1
|
Mylona E, Zaridis DI, Kalantzopoulos CΝ, Tachos NS, Regge D, Papanikolaou N, Tsiknakis M, Marias K, Fotiadis DI. Optimizing radiomics for prostate cancer diagnosis: feature selection strategies, machine learning classifiers, and MRI sequences. Insights Imaging 2024; 15:265. [PMID: 39495422 PMCID: PMC11535140 DOI: 10.1186/s13244-024-01783-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/27/2024] [Indexed: 11/05/2024] Open
Abstract
OBJECTIVES Radiomics-based analyses encompass multiple steps, leading to ambiguity regarding the optimal approaches for enhancing model performance. This study compares the effect of several feature selection methods, machine learning (ML) classifiers, and sources of radiomic features, on models' performance for the diagnosis of clinically significant prostate cancer (csPCa) from bi-parametric MRI. METHODS Two multi-centric datasets, with 465 and 204 patients each, were used to extract 1246 radiomic features per patient and MRI sequence. Ten feature selection methods, such as Boruta, mRMRe, ReliefF, recursive feature elimination (RFE), random forest (RF) variable importance, L1-lasso, etc., four ML classifiers, namely SVM, RF, LASSO, and boosted generalized linear model (GLM), and three sets of radiomics features, derived from T2w images, ADC maps, and their combination, were used to develop predictive models of csPCa. Their performance was evaluated in a nested cross-validation and externally, using seven performance metrics. RESULTS In total, 480 models were developed. In nested cross-validation, the best model combined Boruta with Boosted GLM (AUC = 0.71, F1 = 0.76). In external validation, the best model combined L1-lasso with boosted GLM (AUC = 0.71, F1 = 0.47). Overall, Boruta, RFE, L1-lasso, and RF variable importance were the top-performing feature selection methods, while the choice of ML classifier didn't significantly affect the results. The ADC-derived features showed the highest discriminatory power with T2w-derived features being less informative, while their combination did not lead to improved performance. CONCLUSION The choice of feature selection method and the source of radiomic features have a profound effect on the models' performance for csPCa diagnosis. CRITICAL RELEVANCE STATEMENT This work may guide future radiomic research, paving the way for the development of more effective and reliable radiomic models; not only for advancing prostate cancer diagnostic strategies, but also for informing broader applications of radiomics in different medical contexts. KEY POINTS Radiomics is a growing field that can still be optimized. Feature selection method impacts radiomics models' performance more than ML algorithms. Best feature selection methods: RFE, LASSO, RF, and Boruta. ADC-derived radiomic features yield more robust models compared to T2w-derived radiomic features.
Collapse
Affiliation(s)
- Eugenia Mylona
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Dimitrios I Zaridis
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
- Biomedical Engineering Laboratory, School of Electrical & Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Charalampos Ν Kalantzopoulos
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Nikolaos S Tachos
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Daniele Regge
- Department of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Manolis Tsiknakis
- Computational Biomedicine Laboratory, Institute of Computer Science, FORTH, GR 70013, Heraklion, Greece
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, GR 71004, Heraklion, Greece
| | - Kostas Marias
- Computational Biomedicine Laboratory, Institute of Computer Science, FORTH, GR 70013, Heraklion, Greece
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, GR 71004, Heraklion, Greece
| | - Dimitrios I Fotiadis
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece.
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
2
|
O'Sullivan NJ, Temperley HC, Horan MT, Curtain BMM, O'Neill M, Donohoe C, Ravi N, Corr A, Meaney JFM, Reynolds JV, Kelly ME. Computed tomography (CT) derived radiomics to predict post-operative disease recurrence in gastric cancer; a systematic review and meta-analysis. Curr Probl Diagn Radiol 2024; 53:717-722. [PMID: 39025746 DOI: 10.1067/j.cpradiol.2024.07.011] [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: 03/26/2024] [Revised: 06/10/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Radiomics offers the potential to predict oncological outcomes from pre-operative imaging in order to identify 'high risk' patients at increased risk of recurrence. The application of radiomics in predicting disease recurrence provides tailoring of therapeutic strategies. We aim to comprehensively assess the existing literature regarding the current role of radiomics as a predictor of disease recurrence in gastric cancer. METHODS A systematic search was conducted in Medline, EMBASE, and Web of Science databases. Inclusion criteria encompassed retrospective and prospective studies investigating the use of radiomics to predict post-operative recurrence in ovarian cancer. Study quality was assessed using the QUADAS-2 and Radiomics Quality Score tools. RESULTS Nine studies met the inclusion criteria, involving a total of 6,662 participants. Radiomic-based nomograms demonstrated consistent performance in predicting disease recurrence, as evidenced by satisfactory area under the receiver operating characteristic curve values (AUC range 0.72 - 1). The pooled AUCs calculated using the inverse-variance method for both the training and validation datasets were 0.819 and 0.789 respectively CONCLUSION: Our review provides good evidence supporting the role of radiomics as a predictor of post-operative disease recurrence in gastric cancer. Included studies noted good performance in predicting their primary outcome. Radiomics may enhance personalised medicine by tailoring treatment decision based on predicted prognosis.
Collapse
Affiliation(s)
- Niall J O'Sullivan
- Department of Radiology, St. James's Hospital, Dublin, Ireland; School of Medicine, Trinity College Dublin, Ireland; The National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland.
| | - Hugo C Temperley
- Department of Radiology, St. James's Hospital, Dublin, Ireland; School of Medicine, Trinity College Dublin, Ireland
| | - Michelle T Horan
- Department of Radiology, St. James's Hospital, Dublin, Ireland; The National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland
| | | | - Maeve O'Neill
- Department of Surgery, St. James's Hospital, Dublin, Ireland
| | - Claire Donohoe
- Department of Upper Gastrointestinal Surgery, St. James's Hospital, Dublin, Ireland
| | - Narayanasamy Ravi
- Department of Upper Gastrointestinal Surgery, St. James's Hospital, Dublin, Ireland
| | - Alison Corr
- Department of Radiology, St. James's Hospital, Dublin, Ireland
| | - James F M Meaney
- Department of Radiology, St. James's Hospital, Dublin, Ireland; School of Medicine, Trinity College Dublin, Ireland; The National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland
| | - John V Reynolds
- School of Medicine, Trinity College Dublin, Ireland; Department of Upper Gastrointestinal Surgery, St. James's Hospital, Dublin, Ireland
| | - Michael E Kelly
- School of Medicine, Trinity College Dublin, Ireland; Department of Surgery, St. James's Hospital, Dublin, Ireland; Trinity St James Cancer Institute, Trinity College Dublin, Ireland
| |
Collapse
|
3
|
Morelli L, Paganelli C, Marvaso G, Parrella G, Annunziata S, Vicini MG, Zaffaroni M, Pepa M, Summers PE, De Cobelli O, Petralia G, Jereczek-Fossa BA, Baroni G. Addressing intra- and inter-institution variability of a radiomic framework based on Apparent Diffusion Coefficient in prostate cancer. Med Phys 2024; 51:8096-8107. [PMID: 39172115 DOI: 10.1002/mp.17355] [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: 02/07/2024] [Revised: 06/27/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a highly heterogeneous disease, making tailored treatment approaches challenging. Magnetic resonance imaging (MRI), notably diffusion-weighted imaging (DWI) and the derived Apparent Diffusion Coefficient (ADC) maps, plays a crucial role in PCa characterization. In this context, radiomics is a very promising approach able to disclose insights from MRI data. However, the sensitivity of radiomic features to MRI settings, encompassing DWI protocols and multicenter variations, requires the development of robust and generalizable models. PURPOSE To develop a comprehensive radiomics framework for noninvasive PCa characterization using ADC maps, focusing on identifying reliable imaging biomarkers against intra- and inter-institution variations. MATERIALS AND METHODS Two patient cohorts, including an internal cohort (118 PCa patients) used for both training (75%) and hold-out testing (25%), and an external cohort (50 PCa patients) for independent testing, were employed in the study. DWI images were acquired with three different DWI protocols on two different MRI scanners: two DWI protocols acquired on a 1.5-T scanner for the internal cohort, and one DWI protocol acquired on a 3-T scanner for the external cohort. One hundred and seven radiomics features (i.e., shape, first order, texture) were extracted from ADC maps of the whole prostate gland. To address variations in DWI protocols and multicenter variability, a dedicated pipeline, including two-way ANOVA, sequential-feature-selection (SFS), and ComBat features harmonization was implemented. Mann-Whitney U-tests (α = 0.05) were performed to find statistically significant features dividing patients with different tumor characteristics in terms of Gleason score (GS) and T-stage. Support-Vector-Machine models were then developed to predict GS and T-stage, and the performance was assessed through the area under the curve (AUC) of receiver-operating-characteristic curves. RESULTS Downstream of ANOVA, two subsets of 38 and 41 features stable against DWI protocol were identified for GS and T-stage, respectively. Among these, SFS revealed the most predictive features, yielding an AUC of 0.75 (GS) and 0.70 (T-stage) in the hold-out test. Employing ComBat harmonization improved the external-test performance of the GS model, raising AUC from 0.72 to 0.78. CONCLUSION By incorporating stable features with a harmonization procedure and validating the model on an external dataset, model robustness, and generalizability were assessed, highlighting the potential of ADC and radiomics for PCa characterization.
Collapse
Affiliation(s)
- Letizia Morelli
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Chiara Paganelli
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Giulia Marvaso
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giovanni Parrella
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Simone Annunziata
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Maria Giulia Vicini
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
| | - Mattia Zaffaroni
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
| | - Matteo Pepa
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
| | | | - Ottavio De Cobelli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Urology, European Institute of Oncology (IEO), Milan, Italy
| | - Giuseppe Petralia
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Radiology, European Institute of Oncology (IEO), Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Guido Baroni
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| |
Collapse
|
4
|
Sourlos N, Vliegenthart R, Santinha J, Klontzas ME, Cuocolo R, Huisman M, van Ooijen P. Recommendations for the creation of benchmark datasets for reproducible artificial intelligence in radiology. Insights Imaging 2024; 15:248. [PMID: 39400639 PMCID: PMC11473745 DOI: 10.1186/s13244-024-01833-2] [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: 02/22/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024] Open
Abstract
Various healthcare domains have witnessed successful preliminary implementation of artificial intelligence (AI) solutions, including radiology, though limited generalizability hinders their widespread adoption. Currently, most research groups and industry have limited access to the data needed for external validation studies. The creation and accessibility of benchmark datasets to validate such solutions represents a critical step towards generalizability, for which an array of aspects ranging from preprocessing to regulatory issues and biostatistical principles come into play. In this article, the authors provide recommendations for the creation of benchmark datasets in radiology, explain current limitations in this realm, and explore potential new approaches. CLINICAL RELEVANCE STATEMENT: Benchmark datasets, facilitating validation of AI software performance can contribute to the adoption of AI in clinical practice. KEY POINTS: Benchmark datasets are essential for the validation of AI software performance. Factors like image quality and representativeness of cases should be considered. Benchmark datasets can help adoption by increasing the trustworthiness and robustness of AI.
Collapse
Affiliation(s)
- Nikos Sourlos
- Department of Radiology, University Medical Center of Groningen, Groningen, The Netherlands
- DataScience Center in Health, University Medical Center Groningen, Groningen, The Netherlands
| | - Rozemarijn Vliegenthart
- Department of Radiology, University Medical Center of Groningen, Groningen, The Netherlands
- DataScience Center in Health, University Medical Center Groningen, Groningen, The Netherlands
| | - Joao Santinha
- Digital Surgery LAB, Champalimaud Foundation, Champalimaud Clinical Centre, Lisbon, Portugal
| | - Michail E Klontzas
- Department of Medical Imaging, University Hospital of Heraklion, Heraklion, Greece
- Department of Radiology, School of Medicine, University of Crete, Heraklion, Greece
| | - Renato Cuocolo
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy
| | - Merel Huisman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter van Ooijen
- DataScience Center in Health, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Radiation Oncology, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
5
|
Lu HJ, Shen CY, Chiu YW, Lin WL, Peng CY, Tseng HC, Hsin CH, Chuang CY, Chen CC, Wu MF, Huang WS, Shen WC. Radiomic biomarkers for platinum-refractory head and neck cancer in the era of immunotherapy. Oral Dis 2024; 30:4220-4230. [PMID: 38178608 DOI: 10.1111/odi.14854] [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: 06/16/2023] [Revised: 11/17/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024]
Abstract
OBJECTIVE Immune checkpoint inhibitors (ICI) are recommended as the first-line therapy for platinum-refractory head and neck squamous cell carcinoma (HNSCC), a disease with a poor prognosis. However, biomarkers in this situation are rare. The objective was to identify radiomic features-associated biomarkers to guide the prognosis and treatment opinions in the era of ICI. METHODS A total of 31 platinum-refractory HNSCC patients were retrospectively enrolled. Of these, 65.5% (20/31) received ICI-based therapy and 35.5% (11/31) did not. Radiomic features of the primary site at the onset of recurrent metastatic (R/M) status were extracted. Prognostic and predictive radiomic biomarkers were analysed. RESULTS The median overall survival from R/M status (R/M OS) was 9.6 months. Grey-level co-occurrence matrix-associated texture features were the most important in identifying the patients with or without 9-month R/M death. A radiomic risk-stratification model was established and equally separated the patients into high-, intermittent- and lower-risk groups (1-year R/M death rate, 100.0% vs. 70.8% vs. 27.1%, p = 0.001). Short-run high grey-level emphasis (SRHGE) was more suitable than programmed death ligand 1 (PD-L1) expression in selecting whether patients received ICI-based therapy. CONCLUSIONS Radiomic features were effective prognostic and predictive biomarkers. Future studies are warranted.
Collapse
Affiliation(s)
- Hsueh-Ju Lu
- Division of Hematology and Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
- College of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chao-Yu Shen
- College of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-Wei Chiu
- Department of Stomatology, Chung Shan Medical University Hospital, Taichung, Taiwan
- College of Oral Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wea-Lung Lin
- College of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pathology, Chung Shan Medical University and Hospital, Taichung, Taiwan
| | - Chih-Yu Peng
- Department of Stomatology, Chung Shan Medical University Hospital, Taichung, Taiwan
- College of Oral Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsien-Chun Tseng
- College of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Radiation Oncology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chung-Han Hsin
- College of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Otolaryngology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Yi Chuang
- College of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Otolaryngology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Chia Chen
- College of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Plastic Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Fang Wu
- Division of Hematology and Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
- College of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wei-Shiou Huang
- Division of Hematology and Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
- College of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wei-Chih Shen
- Department of Medical Informatics, Chung Shan Medical University, Taichung, Taiwan
- Artificial Intelligence Center, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
6
|
Ebrahimpour L, Després P, Manem V. Differential Radiomics-Based Signature Predicts Lung Cancer Risk Accounting for Imaging Parameters in NLST Cohort. Cancer Med 2024; 13:e70359. [PMID: 39463128 PMCID: PMC11513548 DOI: 10.1002/cam4.70359] [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: 07/18/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024] Open
Abstract
OBJECTIVE Lung cancer remains the leading cause of cancer-related mortality worldwide, with most cases diagnosed at advanced stages. Hence, there is a need to develop effective predictive models for early detection. This study aims to investigate the impact of imaging parameters and delta radiomic features from temporal scans on lung cancer risk prediction. METHODS Using the National Lung Screening Trial (NLST) within a nested case-control study involving 462 positive screenings, radiomic features were extracted from temporal computed tomography (CT) scans and harmonized with ComBat method to adjust variations in slice thickness category (TC) and reconstruction kernel type (KT). Both harmonized and non-harmonized features from baseline (T0), delta features between T0 and a year later (T1), and combined T0 and delta features were utilized for the analysis. Feature reduction was done using LASSO, followed by five feature selection (FS) methods and nine machine learning (ML) models, evaluated with 5-fold cross-validation repeated 10 times. Synthetic Minority Oversampling Technique (SMOTE) was applied to address class imbalances for lung cancer risk prediction. RESULTS Models using delta features outperformed baseline features, with SMOTE consistently boosting performance when using combination of baseline and delta features. TC-based harmonized features improved performance with SMOTE, but overall, harmonization did not significantly enhance the model performance. The highest test score of 0.76 was achieved in three scenarios: delta features with a Gradient Boosting (GB) model (TC-based harmonization and MultiSurf FS); and T0 + delta features, with both a Support Vector Classifier (SVC) model (KT-based harmonization and F-test FS), and an XGBoost (XGB) model (TC-based harmonization and Mutual Information (MI) FS), all using SMOTE. CONCLUSIONS This study underscores the significance of delta radiomic features and balanced datasets to improve lung cancer prediction. While our findings are based on a subsample of NLST data, they provide a valuable foundation for further exploration. Further research is needed to assess the impact of harmonization on imaging-derived models. Future investigations should explore advanced harmonization techniques and additional imaging parameters to develop robust radiomics-based biomarkers of lung cancer risk.
Collapse
Affiliation(s)
- Leyla Ebrahimpour
- Centre de Recherche du CHU de QuébecUniversité LavalCanada
- Department of Radiology and Imaging SciencesEmory UniversityAtlantaGeorgiaUSA
- Département de Physique, de Génie Physique et D'optiqueUniversité LavalCanada
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de QuébecCanada
| | - Philippe Després
- Département de Physique, de Génie Physique et D'optiqueUniversité LavalCanada
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de QuébecCanada
- Big Data Research CenterUniversité LavalCanada
| | - Venkata S. K. Manem
- Centre de Recherche du CHU de QuébecUniversité LavalCanada
- Big Data Research CenterUniversité LavalCanada
- Cancer Research CenterUniversité LavalCanada
- Département de Biologie Moléculaire, Biochimie Médicale et PathologieUniversité LavalCanada
| |
Collapse
|
7
|
Ibrahim A, Guha S, Lu L, Geng P, Wu Q, Chou Y, Yang H, Wang D, Schwartz LH, Xie CM, Zhao B. The reproducibility and predictivity of radiomic features extracted from dynamic contrast-enhanced computed tomography of hepatocellular carcinoma. PLoS One 2024; 19:e0310486. [PMID: 39269960 PMCID: PMC11398651 DOI: 10.1371/journal.pone.0310486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
PURPOSE To assess the reproducibility of radiomic features (RFs) extracted from dynamic contrast-enhanced computed tomography (DCE-CT) scans of patients diagnosed with hepatocellular carcinoma (HCC) with regards to inter-observer variability and acquisition timing after contrast injection. The predictive ability of reproducible RFs for differentiating between the degrees of HCC differentiation is also investigated. METHODS We analyzed a set of DCE-CT scans of 39 patients diagnosed with HCC. Two radiologists independently segmented the scans, and RFs were extracted from each sequence of the DCE-CT scans. The same lesion was segmented across the DCE-CT sequences of each patient's scan. From each lesion, 127 commonly used RFs were extracted. The reproducibility of RFs was assessed with regard to (i) inter-observer variability, by evaluating the reproducibility of RFs between the two radiologists; and (ii) timing of acquisition following contrast injection (inter- and intra-imaging phase). The reproducibility of RFs was assessed using the concordance correlation coefficient (CCC), with a cut-off value of 0.90. Reproducible RFs were used for building XGBoost classification models for the differentiation of HCC differentiation. RESULTS Inter-observer analyses across the different contrast-enhancement phases showed that the number of reproducible RFs was 29 (22.8%), 52 (40.9%), and 36 (28.3%) for the non-contrast enhanced, late arterial, and portal venous phases, respectively. Intra- and inter-sequence analyses revealed that the number of reproducible RFs ranged between 1 (0.8%) and 47 (37%), inversely related with time interval between the sequences. XGBoost algorithms built using reproducible RFs in each phase were found to be high predictive ability of the degree of HCC tumor differentiation. CONCLUSIONS The reproducibility of many RFs was significantly impacted by inter-observer variability, and a larger number of RFs were impacted by the difference in the time of acquisition after contrast injection. Our findings highlight the need for quality assessment to ensure that scans are analyzed in the same physiologic imaging phase in quantitative imaging studies, or that phase-wide reproducible RFs are selected. Overall, the study emphasizes the importance of reproducibility and quality control when using RFs as biomarkers for clinical applications.
Collapse
Affiliation(s)
- Abdalla Ibrahim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Siddharth Guha
- Department of Radiology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Lin Lu
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Pengfei Geng
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Qian Wu
- First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| | - Yen Chou
- Department of Medical Imaging, Fu Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Hao Yang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Delin Wang
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Lawrence H. Schwartz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Chuan-miao Xie
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Binsheng Zhao
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| |
Collapse
|
8
|
Alkhatib R, Gaede KI. Data Management in Biobanking: Strategies, Challenges, and Future Directions. BIOTECH 2024; 13:34. [PMID: 39311336 PMCID: PMC11417763 DOI: 10.3390/biotech13030034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/23/2024] [Accepted: 08/31/2024] [Indexed: 09/26/2024] Open
Abstract
Biobanking plays a pivotal role in biomedical research by providing standardized processing, precise storing, and management of biological sample collections along with the associated data. Effective data management is a prerequisite to ensure the integrity, quality, and accessibility of these resources. This review provides a current landscape of data management in biobanking, discussing key challenges, existing strategies, and potential future directions. We explore multiple aspects of data management, including data collection, storage, curation, sharing, and ethical considerations. By examining the evolving technologies and methodologies in biobanking, we aim to provide insights into addressing the complexities and maximizing the utility of biobank data for research and clinical applications.
Collapse
Affiliation(s)
- Ramez Alkhatib
- Biomaterial Bank Nord, Research Center Borstel Leibniz Lung Center, Parkallee 35, 23845 Borstel, Germany;
- German Centre for Lung Research (DZL), Airway Research Centre North (ARCN), 22927 Großhansdorf, Germany
| | - Karoline I. Gaede
- Biomaterial Bank Nord, Research Center Borstel Leibniz Lung Center, Parkallee 35, 23845 Borstel, Germany;
- German Centre for Lung Research (DZL), Airway Research Centre North (ARCN), 22927 Großhansdorf, Germany
- PopGen 2.0 Biobanking Network (P2N), University Hospital Schleswig-Holstein, Campus Kiel, Kiel University, 24105 Kiel, Germany
| |
Collapse
|
9
|
Hu Y, Geng Y, Wang H, Chen H, Wang Z, Fu L, Huang B, Jiang W. Improved Prediction of Epidermal Growth Factor Receptor Status by Combined Radiomics of Primary Nonsmall-Cell Lung Cancer and Distant Metastasis. J Comput Assist Tomogr 2024; 48:780-788. [PMID: 38498926 DOI: 10.1097/rct.0000000000001591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
OBJECTIVES This study aimed to investigate radiomics based on primary nonsmall-cell lung cancer (NSCLC) and distant metastases to predict epidermal growth factor receptor (EGFR) mutation status. METHODS A total of 290 patients (mean age, 58.21 ± 9.28) diagnosed with brain (BM, n = 150) or spinal bone metastasis (SM, n = 140) from primary NSCLC were enrolled as a primary cohort. An external validation cohort, consisting of 69 patients (mean age, 59.87 ± 7.23; BM, n = 36; SM, n = 33), was enrolled from another center. Thoracic computed tomography-based features were extracted from the primary tumor and peritumoral area and selected using the least absolute shrinkage and selection operator regression to build a radiomic signature (RS-primary). Contrast-enhanced magnetic resonance imaging-based features were calculated and selected from the BM and SM to build RS-BM and RS-SM, respectively. The RS-BM-Com and RS-SM-Com were developed by integrating the most important features from the primary tumor, BM, and SM. RESULTS Six computed tomography-based features showed high association with EGFR mutation status: 3 from intratumoral and 3 from peritumoral areas. By combination of features from primary tumor and metastases, the developed RS-BM-Com and RS-SM-Com performed well with areas under curve in the training (RS-BM-Com vs RS-BM, 0.936 vs 0.885, P = 0.177; RS-SM-Com vs RS-SM, 0.929 vs 0.843, P = 0.003), internal validation (RS-BM-Com vs RS-BM, 0.920 vs 0.858, P = 0.492; RS-SM-Com vs RS-SM, 0.896 vs 0.859, P = 0.379), and external validation (RS-BM-Com vs RS-BM, 0.882 vs 0.805, P = 0.263; RS-SM-Com vs RS-SM, 0.865 vs 0.816, P = 0.312) cohorts. CONCLUSIONS This study indicates that the accuracy of detecting EGFR mutations significantly enhanced in the presence of metastases in primary NSCLC. The established radiomic signatures from this approach may be useful as new predictors for patients with distant metastases.
Collapse
Affiliation(s)
- Yue Hu
- From the Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yikang Geng
- School of Intelligent Medicine, China Medical University, Liaoning
| | - Huan Wang
- Radiation Oncology Department of Thoracic Cancer, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Liaoning
| | - Huanhuan Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang
| | - Zekun Wang
- Department of Medical Iconography, Liaoning Cancer Hospital & Institute, Liaoning
| | - Langyuan Fu
- School of Intelligent Medicine, China Medical University, Liaoning
| | - Bo Huang
- Department of Pathology, Liaoning Cancer Hospital and Institute, Liaoning
| | - Wenyan Jiang
- Department of Scientific Research and Academic, Liaoning Cancer Hospital and Institute, Liaoning, People's Republic of China
| |
Collapse
|
10
|
Ma Z, Zhang J, Liu X, Teng X, Huang YH, Zhang X, Li J, Pan Y, Sun J, Dong Y, Li T, Chan LWC, Chang ATY, Siu SWK, Cheung ALY, Yang R, Cai J. Comparative Analysis of Repeatability in CT Radiomics and Dosiomics Features under Image Perturbation: A Study in Cervical Cancer Patients. Cancers (Basel) 2024; 16:2872. [PMID: 39199643 PMCID: PMC11352227 DOI: 10.3390/cancers16162872] [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: 06/11/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
This study aims to evaluate the repeatability of radiomics and dosiomics features via image perturbation of patients with cervical cancer. A total of 304 cervical cancer patients with planning CT images and dose maps were retrospectively included. Random translation, rotation, and contour randomization were applied to CT images and dose maps before radiomics feature extraction. The repeatability of radiomics and dosiomics features was assessed using intra-class correlation of coefficient (ICC). Pearson correlation coefficient (r) was adopted to quantify the correlation between the image characteristics and feature repeatability. In general, the repeatability of dosiomics features was lower compared with CT radiomics features, especially after small-sigma Laplacian-of-Gaussian (LoG) and wavelet filtering. More repeatable features (ICC > 0.9) were observed when extracted from the original, Large-sigma LoG filtered, and LLL-/LLH-wavelet filtered images. Positive correlations were found between image entropy and high-repeatable feature number in both CT and dose (r = 0.56, 0.68). Radiomics features showed higher repeatability compared to dosiomics features. These findings highlight the potential of radiomics features for robust quantitative imaging analysis in cervical cancer patients, while suggesting the need for further refinement of dosiomics approaches to enhance their repeatability.
Collapse
Affiliation(s)
- Zongrui Ma
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (Z.M.); (J.Z.); (Y.D.)
| | - Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (Z.M.); (J.Z.); (Y.D.)
| | - Xi Liu
- Department of Radiation Oncology, Cancer Center, Peking University Third Hospital, Beijing 100191, China; (X.L.)
- School of Physics, Beihang University, Beijing 102206, China
| | - Xinzhi Teng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (Z.M.); (J.Z.); (Y.D.)
| | - Yu-Hua Huang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (Z.M.); (J.Z.); (Y.D.)
| | - Xile Zhang
- Department of Radiation Oncology, Cancer Center, Peking University Third Hospital, Beijing 100191, China; (X.L.)
| | - Jun Li
- Department of Radiation Oncology, Cancer Center, Peking University Third Hospital, Beijing 100191, China; (X.L.)
| | - Yuxi Pan
- Department of Radiation Oncology, Cancer Center, Peking University Third Hospital, Beijing 100191, China; (X.L.)
| | - Jiachen Sun
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (Z.M.); (J.Z.); (Y.D.)
| | - Yanjing Dong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (Z.M.); (J.Z.); (Y.D.)
| | - Tian Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (Z.M.); (J.Z.); (Y.D.)
| | - Lawrence Wing Chi Chan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (Z.M.); (J.Z.); (Y.D.)
| | - Amy Tien Yee Chang
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | | | - Andy Lai-Yin Cheung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (Z.M.); (J.Z.); (Y.D.)
- Department of Clinical Oncology, St. Paul’s Hospital, Hong Kong, China
| | - Ruijie Yang
- Department of Radiation Oncology, Cancer Center, Peking University Third Hospital, Beijing 100191, China; (X.L.)
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (Z.M.); (J.Z.); (Y.D.)
| |
Collapse
|
11
|
Del Corso G, Germanese D, Caudai C, Anastasi G, Belli P, Formica A, Nicolucci A, Palma S, Pascali MA, Pieroni S, Trombadori C, Colantonio S, Franchini M, Molinaro S. Adaptive Machine Learning Approach for Importance Evaluation of Multimodal Breast Cancer Radiomic Features. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1642-1651. [PMID: 38478187 PMCID: PMC11300750 DOI: 10.1007/s10278-024-01064-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/26/2024] [Accepted: 02/14/2024] [Indexed: 08/07/2024]
Abstract
Breast cancer holds the highest diagnosis rate among female tumors and is the leading cause of death among women. Quantitative analysis of radiological images shows the potential to address several medical challenges, including the early detection and classification of breast tumors. In the P.I.N.K study, 66 women were enrolled. Their paired Automated Breast Volume Scanner (ABVS) and Digital Breast Tomosynthesis (DBT) images, annotated with cancerous lesions, populated the first ABVS+DBT dataset. This enabled not only a radiomic analysis for the malignant vs. benign breast cancer classification, but also the comparison of the two modalities. For this purpose, the models were trained using a leave-one-out nested cross-validation strategy combined with a proper threshold selection approach. This approach provides statistically significant results even with medium-sized data sets. Additionally it provides distributional variables of importance, thus identifying the most informative radiomic features. The analysis proved the predictive capacity of radiomic models even using a reduced number of features. Indeed, from tomography we achieved AUC-ROC 89.9 % using 19 features and 92.1 % using 7 of them; while from ABVS we attained an AUC-ROC of 72.3 % using 22 features and 85.8 % using only 3 features. Although the predictive power of DBT outperforms ABVS, when comparing the predictions at the patient level, only 8.7% of lesions are misclassified by both methods, suggesting a partial complementarity. Notably, promising results (AUC-ROC ABVS-DBT 71.8 % - 74.1 % ) were achieved using non-geometric features, thus opening the way to the integration of virtual biopsy in medical routine.
Collapse
Affiliation(s)
- Giulio Del Corso
- Institute of Information Science and Technologies "A. Faedo" (ISTI), National Research Council of Italy (CNR), Pisa, Italy.
| | - Danila Germanese
- Institute of Information Science and Technologies "A. Faedo" (ISTI), National Research Council of Italy (CNR), Pisa, Italy
| | - Claudia Caudai
- Institute of Information Science and Technologies "A. Faedo" (ISTI), National Research Council of Italy (CNR), Pisa, Italy
| | - Giada Anastasi
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), Pisa, Italy
- Department of Computer Science, University of Pisa, Pisa, Italy
| | - Paolo Belli
- Policlinico Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessia Formica
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), Pisa, Italy
| | | | | | - Maria Antonietta Pascali
- Institute of Information Science and Technologies "A. Faedo" (ISTI), National Research Council of Italy (CNR), Pisa, Italy
| | - Stefania Pieroni
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), Pisa, Italy
| | | | - Sara Colantonio
- Institute of Information Science and Technologies "A. Faedo" (ISTI), National Research Council of Italy (CNR), Pisa, Italy
| | - Michela Franchini
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), Pisa, Italy.
| | - Sabrina Molinaro
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), Pisa, Italy
| |
Collapse
|
12
|
Maggie Liu SY, Jin ZY, Deng JY, Zhong SM, Ahn MJ, Horinouchi H, Li Y, Wu YL. Drug development and evidence for lung cancer targeted therapy in Eastern Asia. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 49:101090. [PMID: 39381018 PMCID: PMC11459064 DOI: 10.1016/j.lanwpc.2024.101090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/25/2024] [Accepted: 04/29/2024] [Indexed: 10/10/2024]
Abstract
The development of targeted drugs in the Eastern Asia region is going through a flourishing stage. With the continuous advancement of technology and medical research, biotechnology companies and research institutions in the region have made significant progress in cancer field. The Eastern Asian region not only actively participates in clinical trials, but is also committed to developing personalized medical plans to meet the diverse genotypes and phenotypes of patients. The governments and enterprises are increasingly valuing innovation, strengthening international cooperation, and promoting drug development. This paper summarizes the development of genetic testing technology, targeted drugs approval, ongoing promising clinical trials in the field of lung cancer and the important progress made by governments in the Eastern Asian region, and proposed key factors that will contribute to the promising future prospects in the region. The targeted drug market in the Eastern Asian region is expected to drive the medical field forward.
Collapse
Affiliation(s)
- Si-Yang Maggie Liu
- Department of Hematology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Zhen-Yi Jin
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, China
| | - Jia-Yi Deng
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Si-Min Zhong
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yangqiu Li
- Department of Hematology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Zhang S, Wang J, Sun S, Zhang Q, Zhai Y, Wang X, Ge P, Shi Z, Zhang D. CT Angiography Radiomics Combining Traditional Risk Factors to Predict Brain Arteriovenous Malformation Rupture: a Machine Learning, Multicenter Study. Transl Stroke Res 2024; 15:784-794. [PMID: 37311939 DOI: 10.1007/s12975-023-01166-0] [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: 05/04/2023] [Revised: 05/04/2023] [Accepted: 06/06/2023] [Indexed: 06/15/2023]
Abstract
This study aimed to develop a machine learning model for predicting brain arteriovenous malformation (bAVM) rupture using a combination of traditional risk factors and radiomics features. This multicenter retrospective study enrolled 586 patients with unruptured bAVMs from 2010 to 2020. All patients were grouped into the hemorrhage (n = 368) and non-hemorrhage (n = 218) groups. The bAVM nidus were segmented on CT angiography images using Slicer software, and radiomic features were extracted using Pyradiomics. The dataset included a training set and an independent testing set. The machine learning model was developed on the training set and validated on the testing set by merging numerous base estimators and a final estimator based on the stacking method. The area under the receiver operating characteristic (ROC) curve, precision, and the f1 score were evaluated to determine the performance of the model. A total of 1790 radiomics features and 8 traditional risk factors were contained in the original dataset, and 241 features remained for model training after L1 regularization filtering. The base estimator of the ensemble model was Logistic Regression, whereas the final estimator was Random Forest. In the training set, the area under the ROC curve of the model was 0.982 (0.967-0.996) and 0.893 (0.826-0.960) in the testing set. This study indicated that radiomics features are a valuable addition to traditional risk factors for predicting bAVM rupture. In the meantime, ensemble learning can effectively improve the performance of a prediction model.
Collapse
Affiliation(s)
- Shaosen Zhang
- Department of Neurosurgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Junjie Wang
- Department of Neurosurgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Shengjun Sun
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuanren Zhai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaochen Wang
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Peicong Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhiyong Shi
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Dong Zhang
- Department of Neurosurgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
14
|
Trojani V, Bassi MC, Verzellesi L, Bertolini M. Impact of Preprocessing Parameters in Medical Imaging-Based Radiomic Studies: A Systematic Review. Cancers (Basel) 2024; 16:2668. [PMID: 39123396 PMCID: PMC11311340 DOI: 10.3390/cancers16152668] [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: 06/21/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Lately, radiomic studies featuring the development of a signature to use in prediction models in diagnosis or prognosis outcomes have been increasingly published. While the results are shown to be promising, these studies still have many pitfalls and limitations. One of the main issues of these studies is that radiomic features depend on how the images are preprocessed before their computation. Since, in widely known and used software for radiomic features calculation, it is possible to set these preprocessing parameters before the calculation of the radiomic feature, there are ongoing studies assessing the stability and repeatability of radiomic features to find the most suitable preprocessing parameters for every used imaging modality. MATERIALS AND METHODS We performed a comprehensive literature search using four electronic databases: PubMed, Cochrane Library, Embase, and Scopus. Mesh terms and free text were modeled in search strategies for databases. The inclusion criteria were studies where preprocessing parameters' influence on feature values and model predictions was addressed. Records lacking information on image acquisition parameters were excluded, and any eligible studies with full-text versions were included in the review process, while conference proceedings and monographs were disregarded. We used the QUADAS-2 (Quality Assessment of Diagnostic Accuracy Studies 2) tool to investigate the risk of bias. We synthesized our data in a table divided by the imaging modalities subgroups. RESULTS After applying the inclusion and exclusion criteria, we selected 43 works. This review examines the impact of preprocessing parameters on the reproducibility and reliability of radiomic features extracted from multimodality imaging (CT, MRI, CBCT, and PET/CT). Standardized preprocessing is crucial for consistent radiomic feature extraction. Key preprocessing steps include voxel resampling, normalization, and discretization, which influence feature robustness and reproducibility. In total, 44% of the included works studied the effects of an isotropic voxel resampling, and most studies opted to employ a discretization strategy. From 2021, several studies started selecting the best set of preprocessing parameters based on models' best performance. As for comparison metrics, ICC was the most used in MRI studies in 58% of the screened works. CONCLUSIONS From our work, we highlighted the need to harmonize the use of preprocessing parameters and their values, especially in light of future studies of prospective studies, which are still lacking in the current literature.
Collapse
Affiliation(s)
- Valeria Trojani
- Medical Physics, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (L.V.); (M.B.)
| | | | - Laura Verzellesi
- Medical Physics, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (L.V.); (M.B.)
| | - Marco Bertolini
- Medical Physics, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (L.V.); (M.B.)
| |
Collapse
|
15
|
Chen L, Zhu W, Zhang W, Chen E, Zhou W. Magnetic resonance imaging radiomics-based prediction of severe inflammatory response in locally advanced rectal cancer patients after neoadjuvant radiochemotherapy. Langenbecks Arch Surg 2024; 409:218. [PMID: 39017754 PMCID: PMC11255083 DOI: 10.1007/s00423-024-03416-7] [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: 10/19/2023] [Accepted: 07/12/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE To predict severe inflammatory response after neoadjuvant radiochemotherapy in locally advanced rectal cancer (RC) patients using magnetic resonance imaging (MRI) radiomics models. METHODS This retrospective study included patients who underwent radical surgery for RC cancer after neoadjuvant radiochemotherapy between July 2017 and December 2019 at XXX Hospital. MRI radiomics features were extracted from T2WI images before (pre-nRCT-RF) and after (post-nRCT-RF) neoadjuvant radiochemotherapy, and the variation of radiomics features before and after neoadjuvant radiochemotherapy (delta-RF) were calculated. Eight, eight, and five most relevant features were identified for pre-nRCT-RF, post-nRCT-RF, and delta-RF, respectively. RESULTS Eighty-six patients were included and randomized 3:1 to the training and test set (n = 65 and n = 21, respectively). The prediction model based on delta-RF had areas under the curve (AUCs) of 0.80 and 0.85 in the training and test set, respectively. A higher rate of difficult operations was observed in patients with severe inflammation (65.5% vs. 42.9%, P = 0.045). CONCLUSION The prediction model based on MRI delta-RF may be a useful tool for predicting severe inflammatory response after neoadjuvant radiochemotherapy in locally advanced RC patients.
Collapse
Affiliation(s)
- Li Chen
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.
| | - Wenchao Zhu
- Department of Radiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wei Zhang
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Engeng Chen
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wei Zhou
- Department of Colorectal Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Scicolone R, Vacca S, Pisu F, Benson JC, Nardi V, Lanzino G, Suri JS, Saba L. Radiomics and artificial intelligence: General notions and applications in the carotid vulnerable plaque. Eur J Radiol 2024; 176:111497. [PMID: 38749095 DOI: 10.1016/j.ejrad.2024.111497] [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: 03/16/2024] [Revised: 04/14/2024] [Accepted: 05/03/2024] [Indexed: 06/17/2024]
Abstract
Carotid atherosclerosis plays a substantial role in cardiovascular morbidity and mortality. Given the multifaceted impact of this disease, there has been increasing interest in harnessing artificial intelligence (AI) and radiomics as complementary tools for the quantitative analysis of medical imaging data. This integrated approach holds promise not only in refining medical imaging data analysis but also in optimizing the utilization of radiologists' expertise. By automating time consuming tasks, AI allows radiologists to focus on more pertinent responsibilities. Simultaneously, the capacity of AI in radiomics to extract nuanced patterns from raw data enhances the exploration of carotid atherosclerosis, advancing efforts in terms of (1) early detection and diagnosis, (2) risk stratification and predictive modeling, (3) improving workflow efficiency, and (4) contributing to advancements in research. This review provides an overview of general concepts related to radiomics and AI, along with their application in the field of carotid vulnerable plaque. It also offers insights into various research studies conducted on this topic across different imaging techniques.
Collapse
Affiliation(s)
- Roberta Scicolone
- Department of Radiology, Azienda Ospedaliero-Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, Cagliari, Italy
| | - Sebastiano Vacca
- University of Cagliari, School of Medicine and Surgery, Cagliari, Italy
| | - Francesco Pisu
- Department of Radiology, Azienda Ospedaliero-Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, Cagliari, Italy
| | - John C Benson
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Valentina Nardi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, USA
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero-Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, Cagliari, Italy.
| |
Collapse
|
17
|
Ma D, Zhou T, Chen J, Chen J. Radiomics diagnostic performance for predicting lymph node metastasis in esophageal cancer: a systematic review and meta-analysis. BMC Med Imaging 2024; 24:144. [PMID: 38867143 PMCID: PMC11170881 DOI: 10.1186/s12880-024-01278-5] [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: 01/26/2024] [Accepted: 04/22/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Esophageal cancer, a global health concern, impacts predominantly men, particularly in Eastern Asia. Lymph node metastasis (LNM) significantly influences prognosis, and current imaging methods exhibit limitations in accurate detection. The integration of radiomics, an artificial intelligence (AI) driven approach in medical imaging, offers a transformative potential. This meta-analysis evaluates existing evidence on the accuracy of radiomics models for predicting LNM in esophageal cancer. METHODS We conducted a systematic review following PRISMA 2020 guidelines, searching Embase, PubMed, and Web of Science for English-language studies up to November 16, 2023. Inclusion criteria focused on preoperatively diagnosed esophageal cancer patients with radiomics predicting LNM before treatment. Exclusion criteria were applied, including non-English studies and those lacking sufficient data or separate validation cohorts. Data extraction encompassed study characteristics and radiomics technical details. Quality assessment employed modified Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) and Radiomics Quality Score (RQS) tools. Statistical analysis involved random-effects models for pooled sensitivity, specificity, diagnostic odds ratio (DOR), and area under the curve (AUC). Heterogeneity and publication bias were assessed using Deek's test and funnel plots. Analysis was performed using Stata version 17.0 and meta-DiSc. RESULTS Out of 426 initially identified citations, nine studies met inclusion criteria, encompassing 719 patients. These retrospective studies utilized CT, PET, and MRI imaging modalities, predominantly conducted in China. Two studies employed deep learning-based radiomics. Quality assessment revealed acceptable QUADAS-2 scores. RQS scores ranged from 9 to 14, averaging 12.78. The diagnostic meta-analysis yielded a pooled sensitivity, specificity, and AUC of 0.72, 0.76, and 0.74, respectively, representing fair diagnostic performance. Meta-regression identified the use of combined models as a significant contributor to heterogeneity (p-value = 0.05). Other factors, such as sample size (> 75) and least absolute shrinkage and selection operator (LASSO) usage for feature extraction, showed potential influence but lacked statistical significance (0.05 < p-value < 0.10). Publication bias was not statistically significant. CONCLUSION Radiomics shows potential for predicting LNM in esophageal cancer, with a moderate diagnostic performance. Standardized approaches, ongoing research, and prospective validation studies are crucial for realizing its clinical applicability.
Collapse
Affiliation(s)
- Dong Ma
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510900, China
| | - Teli Zhou
- Guangzhou Shiyuan Clinics Co., Ltd, Guangzhou, Guangdong, 510530, China
| | - Jing Chen
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510900, China
| | - Jun Chen
- Dingxi People's Hospital, Dingxi, Gansu, 743000, China.
| |
Collapse
|
18
|
Hoffmann E, Masthoff M, Kunz WG, Seidensticker M, Bobe S, Gerwing M, Berdel WE, Schliemann C, Faber C, Wildgruber M. Multiparametric MRI for characterization of the tumour microenvironment. Nat Rev Clin Oncol 2024; 21:428-448. [PMID: 38641651 DOI: 10.1038/s41571-024-00891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 04/21/2024]
Abstract
Our understanding of tumour biology has evolved over the past decades and cancer is now viewed as a complex ecosystem with interactions between various cellular and non-cellular components within the tumour microenvironment (TME) at multiple scales. However, morphological imaging remains the mainstay of tumour staging and assessment of response to therapy, and the characterization of the TME with non-invasive imaging has not yet entered routine clinical practice. By combining multiple MRI sequences, each providing different but complementary information about the TME, multiparametric MRI (mpMRI) enables non-invasive assessment of molecular and cellular features within the TME, including their spatial and temporal heterogeneity. With an increasing number of advanced MRI techniques bridging the gap between preclinical and clinical applications, mpMRI could ultimately guide the selection of treatment approaches, precisely tailored to each individual patient, tumour and therapeutic modality. In this Review, we describe the evolving role of mpMRI in the non-invasive characterization of the TME, outline its applications for cancer detection, staging and assessment of response to therapy, and discuss considerations and challenges for its use in future medical applications, including personalized integrated diagnostics.
Collapse
Affiliation(s)
- Emily Hoffmann
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Max Masthoff
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Bobe
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Mirjam Gerwing
- Clinic of Radiology, University of Münster, Münster, Germany
| | | | | | - Cornelius Faber
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
19
|
Lo Gullo R, Marcus E, Huayanay J, Eskreis-Winkler S, Thakur S, Teuwen J, Pinker K. Artificial Intelligence-Enhanced Breast MRI: Applications in Breast Cancer Primary Treatment Response Assessment and Prediction. Invest Radiol 2024; 59:230-242. [PMID: 37493391 PMCID: PMC10818006 DOI: 10.1097/rli.0000000000001010] [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] [Indexed: 07/27/2023]
Abstract
ABSTRACT Primary systemic therapy (PST) is the treatment of choice in patients with locally advanced breast cancer and is nowadays also often used in patients with early-stage breast cancer. Although imaging remains pivotal to assess response to PST accurately, the use of imaging to predict response to PST has the potential to not only better prognostication but also allow the de-escalation or omission of potentially toxic treatment with undesirable adverse effects, the accelerated implementation of new targeted therapies, and the mitigation of surgical delays in selected patients. In response to the limited ability of radiologists to predict response to PST via qualitative, subjective assessments of tumors on magnetic resonance imaging (MRI), artificial intelligence-enhanced MRI with classical machine learning, and in more recent times, deep learning, have been used with promising results to predict response, both before the start of PST and in the early stages of treatment. This review provides an overview of the current applications of artificial intelligence to MRI in assessing and predicting response to PST, and discusses the challenges and limitations of their clinical implementation.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
| | - Eric Marcus
- AI for Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Jorge Huayanay
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
- Department of Radiology, National Institute of Neoplastic Diseases, Lima, Peru
| | - Sarah Eskreis-Winkler
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
| | - Sunitha Thakur
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jonas Teuwen
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, the Netherlands
- AI for Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Katja Pinker
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66 Street, New York, NY 10065, USA
| |
Collapse
|
20
|
Robson N, Thekkinkattil DK. Current Role and Future Prospects of Positron Emission Tomography (PET)/Computed Tomography (CT) in the Management of Breast Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:321. [PMID: 38399608 PMCID: PMC10889944 DOI: 10.3390/medicina60020321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024]
Abstract
Breast cancer has become the most diagnosed cancer in women globally, with 2.3 million new diagnoses each year. Accurate early staging is essential for improving survival rates with metastatic spread from loco regional to distant metastasis, decreasing mortality rates by 50%. Current guidelines do not advice the routine use of positron emission tomography (PET)-computed tomography (CT) in the staging of early breast cancer in the absence of symptoms. However, there is a growing body of evidence to suggest that the use of PET-CT in this early stage can benefit the patient by improving staging and as a result treatment and outcomes, as well as psychological burden, without increasing costs to the health service. Ongoing research in PET radiomics and artificial intelligence is showing promising future prospects in its use in diagnosis, staging, prognostication, and assessment of responses to the treatment of breast cancer. Furthermore, ongoing research to address current limitations of PET-CT by improving techniques and tracers is encouraging. In this narrative review, we aim to evaluate the current evidence of the usefulness of PET-CT in the management of breast cancer in different settings along with its future prospects, including the use of artificial intelligence (AI), radiomics, and novel tracers.
Collapse
Affiliation(s)
- Nicole Robson
- Lincoln Medical School, Ross Lucas Medical Sciences Building, University of Lincoln, Lincoln LN6 7FS, UK;
| | | |
Collapse
|
21
|
Brancato V, Esposito G, Coppola L, Cavaliere C, Mirabelli P, Scapicchio C, Borgheresi R, Neri E, Salvatore M, Aiello M. Standardizing digital biobanks: integrating imaging, genomic, and clinical data for precision medicine. J Transl Med 2024; 22:136. [PMID: 38317237 PMCID: PMC10845786 DOI: 10.1186/s12967-024-04891-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/14/2024] [Indexed: 02/07/2024] Open
Abstract
Advancements in data acquisition and computational methods are generating a large amount of heterogeneous biomedical data from diagnostic domains such as clinical imaging, pathology, and next-generation sequencing (NGS), which help characterize individual differences in patients. However, this information needs to be available and suitable to promote and support scientific research and technological development, supporting the effective adoption of the precision medicine approach in clinical practice. Digital biobanks can catalyze this process, facilitating the sharing of curated and standardized imaging data, clinical, pathological and molecular data, crucial to enable the development of a comprehensive and personalized data-driven diagnostic approach in disease management and fostering the development of computational predictive models. This work aims to frame this perspective, first by evaluating the state of standardization of individual diagnostic domains and then by identifying challenges and proposing a possible solution towards an integrative approach that can guarantee the suitability of information that can be shared through a digital biobank. Our analysis of the state of the art shows the presence and use of reference standards in biobanks and, generally, digital repositories for each specific domain. Despite this, standardization to guarantee the integration and reproducibility of the numerical descriptors generated by each domain, e.g. radiomic, pathomic and -omic features, is still an open challenge. Based on specific use cases and scenarios, an integration model, based on the JSON format, is proposed that can help address this problem. Ultimately, this work shows how, with specific standardization and promotion efforts, the digital biobank model can become an enabling technology for the comprehensive study of diseases and the effective development of data-driven technologies at the service of precision medicine.
Collapse
Affiliation(s)
| | - Giuseppina Esposito
- Bio Check Up S.R.L, 80121, Naples, Italy
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131, Naples, Italy
| | | | | | - Peppino Mirabelli
- UOS Laboratori di Ricerca e Biobanca, AORN Santobono-Pausilipon, Via Teresa Ravaschieri, 8, 80122, Naples, Italy
| | - Camilla Scapicchio
- Academic Radiology, Department of Translational Research, University of Pisa, via Roma, 67, 56126, Pisa, Italy
| | - Rita Borgheresi
- Academic Radiology, Department of Translational Research, University of Pisa, via Roma, 67, 56126, Pisa, Italy
| | - Emanuele Neri
- Academic Radiology, Department of Translational Research, University of Pisa, via Roma, 67, 56126, Pisa, Italy
| | | | | |
Collapse
|
22
|
Boubnovski Martell M, Linton-Reid K, Hindocha S, Chen M, Moreno P, Álvarez-Benito M, Salvatierra Á, Lee R, Posma JM, Calzado MA, Aboagye EO. Deep representation learning of tissue metabolome and computed tomography annotates NSCLC classification and prognosis. NPJ Precis Oncol 2024; 8:28. [PMID: 38310164 PMCID: PMC10838282 DOI: 10.1038/s41698-024-00502-3] [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: 06/21/2023] [Accepted: 01/04/2024] [Indexed: 02/05/2024] Open
Abstract
The rich chemical information from tissue metabolomics provides a powerful means to elaborate tissue physiology or tumor characteristics at cellular and tumor microenvironment levels. However, the process of obtaining such information requires invasive biopsies, is costly, and can delay clinical patient management. Conversely, computed tomography (CT) is a clinical standard of care but does not intuitively harbor histological or prognostic information. Furthermore, the ability to embed metabolome information into CT to subsequently use the learned representation for classification or prognosis has yet to be described. This study develops a deep learning-based framework -- tissue-metabolomic-radiomic-CT (TMR-CT) by combining 48 paired CT images and tumor/normal tissue metabolite intensities to generate ten image embeddings to infer metabolite-derived representation from CT alone. In clinical NSCLC settings, we ascertain whether TMR-CT results in an enhanced feature generation model solving histology classification/prognosis tasks in an unseen international CT dataset of 742 patients. TMR-CT non-invasively determines histological classes - adenocarcinoma/squamous cell carcinoma with an F1-score = 0.78 and further asserts patients' prognosis with a c-index = 0.72, surpassing the performance of radiomics models and deep learning on single modality CT feature extraction. Additionally, our work shows the potential to generate informative biology-inspired CT-led features to explore connections between hard-to-obtain tissue metabolic profiles and routine lesion-derived image data.
Collapse
Affiliation(s)
| | | | - Sumeet Hindocha
- Early Diagnosis and Detection Centre, National Institute for Health and Care Research Biomedical Research Centre at the Royal Marsden and Institute of Cancer Research, London, SW3 6JJ, UK
| | - Mitchell Chen
- Imperial College London Hammersmith Campus, London, SW7 2AZ, UK
| | - Paula Moreno
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, 14004, Spain
- Departamento de Cirugía Toráxica y Trasplante de Pulmón, Hospital Universitario Reina Sofía, Córdoba, 14014, Spain
| | - Marina Álvarez-Benito
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, 14004, Spain
- Unidad de Radiodiagnóstico y Cáncer de Mama, Hospital Universitario Reina Sofía, Córdoba, 14004, Spain
| | - Ángel Salvatierra
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, 14004, Spain
- Unidad de Radiodiagnóstico y Cáncer de Mama, Hospital Universitario Reina Sofía, Córdoba, 14004, Spain
| | - Richard Lee
- Early Diagnosis and Detection Centre, National Institute for Health and Care Research Biomedical Research Centre at the Royal Marsden and Institute of Cancer Research, London, SW3 6JJ, UK
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Dovehouse Street, London, SW3 6LY, UK
| | - Joram M Posma
- Imperial College London Hammersmith Campus, London, SW7 2AZ, UK
| | - Marco A Calzado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, 14004, Spain.
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, 14014, Spain.
| | - Eric O Aboagye
- Imperial College London Hammersmith Campus, London, SW7 2AZ, UK.
| |
Collapse
|
23
|
Jiang W, Meng R, Cheng Y, Wang H, Han T, Qu N, Yu T, Hou Y, Xu S. Intra- and Peritumoral Based Radiomics for Assessment of Lymphovascular Invasion in Invasive Breast Cancer. J Magn Reson Imaging 2024; 59:613-625. [PMID: 37199241 DOI: 10.1002/jmri.28776] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND Radiomics has been applied for assessing lymphovascular invasion (LVI) in patients with breast cancer. However, associations between features from peritumoral regions and the LVI status were not investigated. PURPOSE To investigate the value of intra- and peritumoral radiomics for assessing LVI, and to develop a nomogram to assist in making treatment decisions. STUDY TYPE Retrospective. POPULATION Three hundred and sixteen patients were enrolled from two centers and divided into training (N = 165), internal validation (N = 83), and external validation (N = 68) cohorts. FIELD STRENGTH/SEQUENCE 1.5 T and 3.0 T/dynamic contrast-enhanced (DCE) and diffusion-weighted imaging (DWI). ASSESSMENT Radiomics features were extracted and selected based on intra- and peritumoral breast regions in two magnetic resonance imaging (MRI) sequences to create the multiparametric MRI combined radiomics signature (RS-DCE plus DWI). The clinical model was built with MRI-axillary lymph nodes (MRI ALN), MRI-reported peritumoral edema (MPE), and apparent diffusion coefficient (ADC). The nomogram was constructed with RS-DCE plus DWI, MRI ALN, MPE, and ADC. STATISTICAL TESTS Intra- and interclass correlation coefficient analysis, Mann-Whitney U test, and least absolute shrinkage and selection operator regression were used for feature selection. Receiver operating characteristic and decision curve analyses were applied to compare performance of the RS-DCE plus DWI, clinical model, and nomogram. RESULTS A total of 10 features were found to be associated with LVI, 3 from intra- and 7 from peritumoral areas. The nomogram showed good performance in the training (AUCs, nomogram vs. clinical model vs. RS-DCE plus DWI, 0.884 vs. 0.695 vs. 0.870), internal validation (AUCs, nomogram vs. clinical model vs. RS-DCE plus DWI, 0.813 vs. 0.695 vs. 0.794), and external validation (AUCs, nomogram vs. clinical model vs. RS-DCE plus DWI, 0.862 vs. 0.601 vs. 0.849) cohorts. DATA CONCLUSION The constructed preoperative nomogram might effectively assess LVI. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Wenyan Jiang
- Department of Scientific Research and Academic, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Ruiqing Meng
- Department of Biomedical Engineering, China Medical University, Shenyang, China
| | - Yuan Cheng
- Department of Biomedical Engineering, China Medical University, Shenyang, China
| | - Haotian Wang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Tingting Han
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ning Qu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tao Yu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Yang Hou
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shu Xu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
24
|
Liu J, Corti A, Calareso G, Spadarella G, Licitra L, Corino VDA, Mainardi L. Developing a robust two-step machine learning multiclassification pipeline to predict primary site in head and neck carcinoma from lymph nodes. Heliyon 2024; 10:e24377. [PMID: 38312621 PMCID: PMC10835257 DOI: 10.1016/j.heliyon.2024.e24377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
This study aimed to develop a robust multiclassification pipeline to determine the primary tumor location in patients with head and neck carcinoma of unknown primary using radiomics and machine learning techniques. The dataset included 400 head and neck cancer patients with primary tumor in oropharynx, OPC (n = 162), nasopharynx, NPC (n = 137), oral cavity, OC (n = 63), larynx and hypopharynx, HL (n = 38). Two radiomic-based multiclassification pipelines (P1 and P2) were developed. P1 consisted in a direct identification of the primary sites, whereas P2 was based on a two-step approach: in the first step, the number of classes was reduced by merging the two minority classes which were reclassified in the second step. Diverse correlation thresholds (0.75, 0.80, 0.85), feature selection methods (sequential forwards/backwards selection, sequential floating forward selection, neighborhood component analysis and minimum redundancy maximum relevance), and classification models (neural network, decision tree, naïve Bayes, bagged trees and support vector machine) were assessed. P2 outperformed P1, with the best results obtained with the support vector machine classifier including radiomic and clinical features (accuracies of 75.3 % (HL), 75.4 % (OC), 71.3 % (OPC), 92.9 % (NPC)). These results indicate that the two-step multiclassification pipeline integrating radiomics and clinical information is a promising approach to predict the tumor site of unknown primary.
Collapse
Affiliation(s)
- Jiaying Liu
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Anna Corti
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Giuseppina Calareso
- Radiology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Gaia Spadarella
- Postgraduation School in Radiodiagnostics, University of Milan, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Lisa Licitra
- Head and Neck Cancer Medical Oncology Department, Fondazione IRCCS Instituto Nazionale dei Tumori di Milano, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Valentina D A Corino
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
- Cardiotech Lab, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Luca Mainardi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
25
|
Wu Q, Lou J, Liu J, Dong L, Wu Q, Wu Y, Yu X, Wang M. Performance of node reporting and data system (node-RADS): a preliminary study in cervical cancer. BMC Med Imaging 2024; 24:28. [PMID: 38279127 PMCID: PMC10811875 DOI: 10.1186/s12880-024-01205-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Node Reporting and Data System (Node-RADS) was proposed and can be applied to lymph nodes (LNs) across all anatomical sites. This study aimed to investigate the diagnostic performance of Node-RADS in cervical cancer patients. METHODS A total of 81 cervical cancer patients treated with radical hysterectomy and LN dissection were retrospectively enrolled. Node-RADS evaluations were performed by two radiologists on preoperative MRI scans for all patients, both at the LN level and patient level. Chi-square and Fisher's exact tests were employed to evaluate the distribution differences in size and configuration between patients with and without LN metastasis (LNM) in various regions. The receiver operating characteristic (ROC) and the area under the curve (AUC) were used to explore the diagnostic performance of the Node-RADS score for LNM. RESULTS The rates of LNM in the para-aortic, common iliac, internal iliac, external iliac, and inguinal regions were 7.4%, 9.3%, 19.8%, 21.0%, and 2.5%, respectively. At the patient level, as the NODE-RADS score increased, the rate of LNM also increased, with rates of 26.1%, 29.2%, 42.9%, 80.0%, and 90.9% for Node-RADS scores 1, 2, 3, 4, and 5, respectively. At the patient level, the AUCs for Node-RADS scores > 1, >2, > 3, and > 4 were 0.632, 0.752, 0.763, and 0.726, respectively. Both at the patient level and LN level, a Node-RADS score > 3 could be considered the optimal cut-off value with the best AUC and accuracy. CONCLUSIONS Node-RADS is effective in predicting LNM for scores 4 to 5. However, the proportions of LNM were more than 25% at the patient level for scores 1 and 2, which does not align with the expected very low and low probability of LNM for these scores.
Collapse
Affiliation(s)
- Qingxia Wu
- Department of Medical Imaging, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Jianghua Lou
- Department of Medical Imaging, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Jinjin Liu
- Department of Medical Imaging, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Linxiao Dong
- Department of Medical Imaging, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Qingxia Wu
- Beijing United Imaging Research Institute of Intelligent Imaging, United Imaging Intelligence (Beijing) Co., Ltd., Beijing, 100089, China
| | - Yaping Wu
- Department of Medical Imaging, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Xuan Yu
- Department of Medical Imaging, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Meiyun Wang
- Department of Medical Imaging, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China.
- Laboratory of Brain Science and Brain-Like Intelligence Technology, Biomedical Research Institute, Henan Academy of Sciences, No. 266-38, Mingli Road, Zhengzhou, Henan, 450046, China.
| |
Collapse
|
26
|
Zhang X, Zhong Y, Jin C, Hu D, Tian M, Zhang H. Medical image Generative Pre-Trained Transformer (MI-GPT): future direction for precision medicine. Eur J Nucl Med Mol Imaging 2024; 51:332-335. [PMID: 37803245 DOI: 10.1007/s00259-023-06450-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Affiliation(s)
- Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Daoyan Hu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China.
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Shanghai, 201203, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China.
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China.
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| |
Collapse
|
27
|
Jin Y, Yin H, Zhang H, Wang Y, Liu S, Yang L, Song B. Predicting tumor deposits in rectal cancer: a combined deep learning model using T2-MR imaging and clinical features. Insights Imaging 2023; 14:221. [PMID: 38117396 PMCID: PMC10733230 DOI: 10.1186/s13244-023-01564-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: 06/15/2023] [Accepted: 11/05/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Tumor deposits (TDs) are associated with poor prognosis in rectal cancer (RC). This study aims to develop and validate a deep learning (DL) model incorporating T2-MR image and clinical factors for the preoperative prediction of TDs in RC patients. METHODS AND METHODS A total of 327 RC patients with pathologically confirmed TDs status from January 2016 to December 2019 were retrospectively recruited, and the T2-MR images and clinical variables were collected. Patients were randomly split into a development dataset (n = 246) and an independent testing dataset (n = 81). A single-channel DL model, a multi-channel DL model, a hybrid DL model, and a clinical model were constructed. The performance of these predictive models was assessed by using receiver operating characteristics (ROC) analysis and decision curve analysis (DCA). RESULTS The areas under the curves (AUCs) of the clinical, single-DL, multi-DL, and hybrid-DL models were 0.734 (95% CI, 0.674-0.788), 0.710 (95% CI, 0.649-0.766), 0.767 (95% CI, 0.710-0.819), and 0.857 (95% CI, 0.807-0.898) in the development dataset. The AUC of the hybrid-DL model was significantly higher than the single-DL and multi-DL models (both p < 0.001) in the development dataset, and the single-DL model (p = 0.028) in the testing dataset. Decision curve analysis demonstrated the hybrid-DL model had higher net benefit than other models across the majority range of threshold probabilities. CONCLUSIONS The proposed hybrid-DL model achieved good predictive efficacy and could be used to predict tumor deposits in rectal cancer. CRITICAL RELEVANCE STATEMENT The proposed hybrid-DL model achieved good predictive efficacy and could be used to predict tumor deposits in rectal cancer. KEY POINTS • Preoperative non-invasive identification of TDs is of great clinical significance. • The combined hybrid-DL model achieved good predictive efficacy and could be used to predict tumor deposits in rectal cancer. • A preoperative nomogram provides gastroenterologist with an accurate and effective tool.
Collapse
Affiliation(s)
- Yumei Jin
- Department of Medical Imaging Center, Qujing First People's Hospital, Qujing, 655000, Yunnan Province, China.
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China.
| | - Hongkun Yin
- Beijing Infervision Technology Co.Ltd, Beijing, China
| | - Huiling Zhang
- Beijing Infervision Technology Co.Ltd, Beijing, China
| | - Yewu Wang
- Department of Joint and Sports Medicine, Qujing First People's Hospital, Qujing, 655000, Yunnan Province, China
| | - Shengmei Liu
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Ling Yang
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Bin Song
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China.
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China.
- Department of Radiology, Sanya People's Hospital, Sanya, Hainan Province, 572000, China.
| |
Collapse
|
28
|
Manco L, Albano D, Urso L, Arnaboldi M, Castellani M, Florimonte L, Guidi G, Turra A, Castello A, Panareo S. Positron Emission Tomography-Derived Radiomics and Artificial Intelligence in Multiple Myeloma: State-of-the-Art. J Clin Med 2023; 12:7669. [PMID: 38137738 PMCID: PMC10743775 DOI: 10.3390/jcm12247669] [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: 11/01/2023] [Revised: 12/02/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Multiple myeloma (MM) is a heterogeneous neoplasm accounting for the second most prevalent hematologic disorder. The identification of noninvasive, valuable biomarkers is of utmost importance for the best patient treatment selection, especially in heterogeneous diseases like MM. Despite molecular imaging with positron emission tomography (PET) has achieved a primary role in the characterization of MM, it is not free from shortcomings. In recent years, radiomics and artificial intelligence (AI), which includes machine learning (ML) and deep learning (DL) algorithms, have played an important role in mining additional information from medical images beyond human eyes' resolving power. Our review provides a summary of the current status of radiomics and AI in different clinical contexts of MM. A systematic search of PubMed, Web of Science, and Scopus was conducted, including all the articles published in English that explored radiomics and AI analyses of PET/CT images in MM. The initial results have highlighted the potential role of such new features in order to improve the clinical stratification of MM patients, as well as to increase their clinical benefits. However, more studies are warranted before these approaches can be implemented in clinical routines.
Collapse
Affiliation(s)
- Luigi Manco
- Medical Physics Unit, Azienda USL of Ferrara, 45100 Ferrara, Italy; (L.M.); (A.T.)
| | - Domenico Albano
- Nuclear Medicine Department, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy;
| | - Luca Urso
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy;
| | - Mattia Arnaboldi
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.A.); (M.C.); (L.F.)
| | - Massimo Castellani
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.A.); (M.C.); (L.F.)
| | - Luigia Florimonte
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.A.); (M.C.); (L.F.)
| | - Gabriele Guidi
- Medical Physics Unit, University Hospital of Modena, 41125 Modena, Italy;
| | - Alessandro Turra
- Medical Physics Unit, Azienda USL of Ferrara, 45100 Ferrara, Italy; (L.M.); (A.T.)
| | - Angelo Castello
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.A.); (M.C.); (L.F.)
| | - Stefano Panareo
- Nuclear Medicine Unit, Department of Oncology and Hematology, University Hospital of Modena, Via del Pozzo 71, 41124 Modena, Italy;
| |
Collapse
|
29
|
Pasini G, Russo G, Mantarro C, Bini F, Richiusa S, Morgante L, Comelli A, Russo GI, Sabini MG, Cosentino S, Marinozzi F, Ippolito M, Stefano A. A Critical Analysis of the Robustness of Radiomics to Variations in Segmentation Methods in 18F-PSMA-1007 PET Images of Patients Affected by Prostate Cancer. Diagnostics (Basel) 2023; 13:3640. [PMID: 38132224 PMCID: PMC10743045 DOI: 10.3390/diagnostics13243640] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/29/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Radiomics shows promising results in supporting the clinical decision process, and much effort has been put into its standardization, thus leading to the Imaging Biomarker Standardization Initiative (IBSI), that established how radiomics features should be computed. However, radiomics still lacks standardization and many factors, such as segmentation methods, limit study reproducibility and robustness. AIM We investigated the impact that three different segmentation methods (manual, thresholding and region growing) have on radiomics features extracted from 18F-PSMA-1007 Positron Emission Tomography (PET) images of 78 patients (43 Low Risk, 35 High Risk). Segmentation was repeated for each patient, thus leading to three datasets of segmentations. Then, feature extraction was performed for each dataset, and 1781 features (107 original, 930 Laplacian of Gaussian (LoG) features, 744 wavelet features) were extracted. Feature robustness and reproducibility were assessed through the intra class correlation coefficient (ICC) to measure agreement between the three segmentation methods. To assess the impact that the three methods had on machine learning models, feature selection was performed through a hybrid descriptive-inferential method, and selected features were given as input to three classifiers, K-Nearest Neighbors (KNN), Support Vector Machines (SVM), Linear Discriminant Analysis (LDA), Random Forest (RF), AdaBoost and Neural Networks (NN), whose performance in discriminating between low-risk and high-risk patients have been validated through 30 times repeated five-fold cross validation. CONCLUSIONS Our study showed that segmentation methods influence radiomics features and that Shape features were the least reproducible (average ICC: 0.27), while GLCM features the most reproducible. Moreover, feature reproducibility changed depending on segmentation type, resulting in 51.18% of LoG features exhibiting excellent reproducibility (range average ICC: 0.68-0.87) and 47.85% of wavelet features exhibiting poor reproducibility that varied between wavelet sub-bands (range average ICC: 0.34-0.80) and resulted in the LLL band showing the highest average ICC (0.80). Finally, model performance showed that region growing led to the highest accuracy (74.49%), improved sensitivity (84.38%) and AUC (79.20%) in contrast with manual segmentation.
Collapse
Affiliation(s)
- Giovanni Pasini
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Eudossiana 18, 00184 Rome, Italy; (G.P.); (L.M.); (F.M.)
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Contrada, Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.R.); (S.R.); (A.C.); (A.S.)
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Contrada, Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.R.); (S.R.); (A.C.); (A.S.)
- National Laboratory of South, National Institute for Nuclear Physics (LNS-INFN), 95125 Catania, Italy
| | - Cristina Mantarro
- Nuclear Medicine Department, Cannizzaro Hospital, 95125 Catania, Italy; (C.M.); (S.C.); (M.I.)
| | - Fabiano Bini
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Eudossiana 18, 00184 Rome, Italy; (G.P.); (L.M.); (F.M.)
| | - Selene Richiusa
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Contrada, Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.R.); (S.R.); (A.C.); (A.S.)
| | - Lucrezia Morgante
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Eudossiana 18, 00184 Rome, Italy; (G.P.); (L.M.); (F.M.)
| | - Albert Comelli
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Contrada, Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.R.); (S.R.); (A.C.); (A.S.)
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
| | - Giorgio Ivan Russo
- Department of Surgery, Urology Section, University of Catania, 95125 Catania, Italy;
| | | | - Sebastiano Cosentino
- Nuclear Medicine Department, Cannizzaro Hospital, 95125 Catania, Italy; (C.M.); (S.C.); (M.I.)
| | - Franco Marinozzi
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Eudossiana 18, 00184 Rome, Italy; (G.P.); (L.M.); (F.M.)
| | - Massimo Ippolito
- Nuclear Medicine Department, Cannizzaro Hospital, 95125 Catania, Italy; (C.M.); (S.C.); (M.I.)
| | - Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Contrada, Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.R.); (S.R.); (A.C.); (A.S.)
- National Laboratory of South, National Institute for Nuclear Physics (LNS-INFN), 95125 Catania, Italy
| |
Collapse
|
30
|
Allam AK, Anand A, Flores AR, Ropper AE. Computer Vision in Osteoporotic Vertebral Fracture Risk Prediction: A Systematic Review. Neurospine 2023; 20:1112-1123. [PMID: 38171281 PMCID: PMC10762393 DOI: 10.14245/ns.2347022.511] [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: 09/30/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Osteoporotic vertebral fractures (OVFs) are a significant health concern linked to increased morbidity, mortality, and diminished quality of life. Traditional OVF risk assessment tools like bone mineral density (BMD) only capture a fraction of the risk profile. Artificial intelligence, specifically computer vision, has revolutionized other fields of medicine through analysis of videos, histopathology slides and radiological scans. In this review, we provide an overview of computer vision algorithms and current computer vision models used in predicting OVF risk. We highlight the clinical applications, future directions and limitations of computer vision in OVF risk prediction.
Collapse
Affiliation(s)
- Anthony K. Allam
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Adrish Anand
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Alex R. Flores
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | | |
Collapse
|
31
|
Ge X, Gao J, Niu R, Shi Y, Shao X, Wang Y, Shao X. New research progress on 18F-FDG PET/CT radiomics for EGFR mutation prediction in lung adenocarcinoma: a review. Front Oncol 2023; 13:1242392. [PMID: 38094613 PMCID: PMC10716448 DOI: 10.3389/fonc.2023.1242392] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/16/2023] [Indexed: 11/09/2024] Open
Abstract
Lung cancer, the most frequently diagnosed cancer worldwide, is the leading cause of cancer-associated deaths. In recent years, significant progress has been achieved in basic and clinical research concerning the epidermal growth factor receptor (EGFR), and the treatment of lung adenocarcinoma has also entered a new era of individualized, targeted therapies. However, the detection of lung adenocarcinoma is usually invasive. 18F-FDG PET/CT can be used as a noninvasive molecular imaging approach, and radiomics can acquire high-throughput data from standard images. These methods play an increasingly prominent role in diagnosing and treating cancers. Herein, we reviewed the progress in applying 18F-FDG PET/CT and radiomics in lung adenocarcinoma clinical research and how these data are analyzed via traditional statistics, machine learning, and deep learning to predict EGFR mutation status, all of which achieved satisfactory results. Traditional statistics extract features effectively, machine learning achieves higher accuracy with complex algorithms, and deep learning obtains significant results through end-to-end methods. Future research should combine these methods to achieve more accurate predictions, providing reliable evidence for the precision treatment of lung adenocarcinoma. At the same time, facing challenges such as data insufficiency and high algorithm complexity, future researchers must continuously explore and optimize to better apply to clinical practice.
Collapse
Affiliation(s)
- Xinyu Ge
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Jianxiong Gao
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Rong Niu
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Yunmei Shi
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Xiaoliang Shao
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Yuetao Wang
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| | - Xiaonan Shao
- Department of Nuclear Medicine, the Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Clinical Translation of Nuclear Medicine and Molecular Imaging, Soochow University, Changzhou, China
- Department of Nuclear Medicine, Changzhou Clinical Medical Center, Changzhou, China
| |
Collapse
|
32
|
Roisman LC, Kian W, Anoze A, Fuchs V, Spector M, Steiner R, Kassel L, Rechnitzer G, Fried I, Peled N, Bogot NR. Radiological artificial intelligence - predicting personalized immunotherapy outcomes in lung cancer. NPJ Precis Oncol 2023; 7:125. [PMID: 37990050 PMCID: PMC10663598 DOI: 10.1038/s41698-023-00473-x] [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: 07/17/2023] [Accepted: 10/24/2023] [Indexed: 11/23/2023] Open
Abstract
Personalized medicine has revolutionized approaches to treatment in the field of lung cancer by enabling therapies to be specific to each patient. However, physicians encounter an immense number of challenges in providing the optimal treatment regimen for the individual given the sheer complexity of clinical aspects such as tumor molecular profile, tumor microenvironment, expected adverse events, acquired or inherent resistance mechanisms, the development of brain metastases, the limited availability of biomarkers and the choice of combination therapy. The integration of innovative next-generation technologies such as deep learning-a subset of machine learning-and radiomics has the potential to transform the field by supporting clinical decision making in cancer treatment and the delivery of precision therapies while integrating numerous clinical considerations. In this review, we present a brief explanation of the available technologies, the benefits of using these technologies in predicting immunotherapy response in lung cancer, and the expected future challenges in the context of precision medicine.
Collapse
Affiliation(s)
- Laila C Roisman
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel.
- Ben-Gurion University of the Negev, Be'er Sheva, Israel.
| | - Waleed Kian
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
- The Institute of Oncology, Assuta Ashdod, Ashdod, Israel
| | - Alaa Anoze
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Vered Fuchs
- Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Maria Spector
- The Department of Radiology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Roee Steiner
- The Institute for Nuclear Medicine, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Levi Kassel
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Gilad Rechnitzer
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Iris Fried
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Nir Peled
- The Hebrew University, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - Naama R Bogot
- The Department of Radiology, Shaare Zedek Medical Center, Jerusalem, Israel
| |
Collapse
|
33
|
Mohammadi S, Ghaderi S, Ghaderi K, Mohammadi M, Pourasl MH. Automated segmentation of meningioma from contrast-enhanced T1-weighted MRI images in a case series using a marker-controlled watershed segmentation and fuzzy C-means clustering machine learning algorithm. Int J Surg Case Rep 2023; 111:108818. [PMID: 37716060 PMCID: PMC10514425 DOI: 10.1016/j.ijscr.2023.108818] [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: 08/22/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023] Open
Abstract
INTRODUCTION AND IMPORTANCE Accurate segmentation of meningiomas from contrast-enhanced T1-weighted (CE T1-w) magnetic resonance imaging (MRI) is crucial for diagnosis and treatment planning. Manual segmentation is time-consuming and prone to variability. To evaluate an automated segmentation approach for meningiomas using marker-controlled watershed segmentation (MCWS) and fuzzy c-means (FCM) algorithms. CASE PRESENTATION AND METHODS CE T1-w MRI of 3 female patients (aged 59, 44, 67 years) with right frontal meningiomas were analyzed. Images were converted to grayscale and preprocessed with Otsu's thresholding and FCM clustering. MCWS segmentation was performed. Segmentation accuracy was assessed by comparing automated segmentations to manual delineations. CLINICAL DISCUSSION The approach successfully segmented meningiomas in all cases. Mean sensitivity was 0.8822, indicating accurate identification of tumors. Mean Dice similarity coefficient between Otsu's and FCM1 was 0.6599, suggesting good overlap between segmentation methods. CONCLUSION The MCWS and FCM approach enables accurate automated segmentation of meningiomas from CE T1-w MRI. With further validation on larger datasets, this could provide an efficient tool to assist in delineating meningioma boundaries for clinical management.
Collapse
Affiliation(s)
- Sana Mohammadi
- Department of Medical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Kayvan Ghaderi
- Department of Information Technology and Computer Engineering, Faculty of Engineering, University of Kurdistan, Sanandaj 66177-15175, Iran
| | - Mahdi Mohammadi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
34
|
Rinaldi L, Guerini Rocco E, Spitaleri G, Raimondi S, Attili I, Ranghiero A, Cammarata G, Minotti M, Lo Presti G, De Piano F, Bellerba F, Funicelli G, Volpe S, Mora S, Fodor C, Rampinelli C, Barberis M, De Marinis F, Jereczek-Fossa BA, Orecchia R, Rizzo S, Botta F. Association between Contrast-Enhanced Computed Tomography Radiomic Features, Genomic Alterations and Prognosis in Advanced Lung Adenocarcinoma Patients. Cancers (Basel) 2023; 15:4553. [PMID: 37760521 PMCID: PMC10527057 DOI: 10.3390/cancers15184553] [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: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Non-invasive methods to assess mutational status, as well as novel prognostic biomarkers, are warranted to foster therapy personalization of patients with advanced non-small cell lung cancer (NSCLC). This study investigated the association of contrast-enhanced Computed Tomography (CT) radiomic features of lung adenocarcinoma lesions, alone or integrated with clinical parameters, with tumor mutational status (EGFR, KRAS, ALK alterations) and Overall Survival (OS). In total, 261 retrospective and 48 prospective patients were enrolled. A Radiomic Score (RS) was created with LASSO-Logistic regression models to predict mutational status. Radiomic, clinical and clinical-radiomic models were trained on retrospective data and tested (Area Under the Curve, AUC) on prospective data. OS prediction models were trained and tested on retrospective data with internal cross-validation (C-index). RS significantly predicted each alteration at training (radiomic and clinical-radiomic AUC 0.95-0.98); validation performance was good for EGFR (AUC 0.86), moderate for KRAS and ALK (AUC 0.61-0.65). RS was also associated with OS at univariate and multivariable analysis, in the latter with stage and type of treatment. The validation C-index was 0.63, 0.79, and 0.80 for clinical, radiomic, and clinical-radiomic models. The study supports the potential role of CT radiomics for non-invasive identification of gene alterations and prognosis prediction in patients with advanced lung adenocarcinoma, to be confirmed with independent studies.
Collapse
Affiliation(s)
- Lisa Rinaldi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
| | - Elena Guerini Rocco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
| | - Gianluca Spitaleri
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Sara Raimondi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Ilaria Attili
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Alberto Ranghiero
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
| | - Giulio Cammarata
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Marta Minotti
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Giuliana Lo Presti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Francesca De Piano
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Federica Bellerba
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy (F.B.)
| | - Gianluigi Funicelli
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Stefania Volpe
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Serena Mora
- Data Management Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (S.M.); (C.F.)
| | - Cristiana Fodor
- Data Management Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (S.M.); (C.F.)
| | - Cristiano Rampinelli
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
| | - Massimo Barberis
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.G.R.); (A.R.); (M.B.)
| | - Filippo De Marinis
- Division of Thoracic Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.S.); (I.A.); (F.D.M.)
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy; (S.V.)
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Roberto Orecchia
- Division of Radiology, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (M.M.); (C.R.); (R.O.)
- Scientific Direction, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Stefania Rizzo
- Clinica di Radiologia EOC, Istituto Imaging della Svizzera Italiana (IIMSI), Via Tesserete 46, 6900 Lugano, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Via G. Buffi 13, 6900 Lugano, Switzerland
| | - Francesca Botta
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
| |
Collapse
|
35
|
Zhang X, Zhang G, Qiu X, Yin J, Tan W, Yin X, Yang H, Liao L, Wang H, Zhang Y. Radiomics under 2D regions, 3D regions, and peritumoral regions reveal tumor heterogeneity in non-small cell lung cancer: a multicenter study. LA RADIOLOGIA MEDICA 2023; 128:1079-1092. [PMID: 37486526 DOI: 10.1007/s11547-023-01676-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/29/2023] [Indexed: 07/25/2023]
Abstract
PURPOSE Lung cancer has significant genetic and phenotypic heterogeneity, leading to poor prognosis. Radiomic features have emerged as promising predictors of the tumor phenotype. However, the role of underlying information surrounding the cancer remains unclear. MATERIALS AND METHODS We conducted a retrospective study of 508 patients with NSCLC from three institutions. Radiomics models were built using features from six tumor regions and seven classifiers to predict three prognostically significant tumor phenotypes. The models were evaluated and interpreted by the mean area under the receiver operating characteristic curve (AUC) under nested cross-validation and Shapley values. The best-performing predictive models corresponding to six tumor regions and three tumor phenotypes were identified for further comparative analysis. In addition, we designed five experiments with different voxel spacing to assess the sensitivity of the experimental results to the spatial resolution of the voxels. RESULTS Our results demonstrated that models based on 2D, 3D, and peritumoral region features yielded mean AUCs and 95% confidence intervals of 0.759 and [0.747-0.771] for lymphovascular invasion, 0.889 and [0.882-0.896] for pleural invasion, and 0.839 and [0.829-0.849] for T-staging in the testing cohort, which was significantly higher than all other models. Similar results were obtained for the model combining the three regional features at five voxel spacings. CONCLUSION Our study revealed the predictive role of the developed methods with multi-regional features for the preoperative assessment of prognostic factors in NSCLC. The analysis of different voxel spacing and model interpretability strengthens the experimental findings and contributes to understanding the biological significance of the radiological phenotype.
Collapse
Affiliation(s)
- Xingping Zhang
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006, China
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC, 3011, Australia
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, 518000, China
| | - Guijuan Zhang
- Department of Respiratory and Critical Care, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Xingting Qiu
- Department of Radiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Jiao Yin
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC, 3011, Australia
| | - Wenjun Tan
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, 110189, China
| | - Xiaoxia Yin
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006, China
| | - Hong Yang
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006, China
| | - Liefa Liao
- School of Information Engineering, Jiangxi University of Science and Technology, Ganzhou, 341000, China
| | - Hua Wang
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC, 3011, Australia.
| | - Yanchun Zhang
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, 510006, China.
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC, 3011, Australia.
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, 518000, China.
| |
Collapse
|
36
|
Tonneau M, Phan K, Manem VSK, Low-Kam C, Dutil F, Kazandjian S, Vanderweyen D, Panasci J, Malo J, Coulombe F, Gagné A, Elkrief A, Belkaïd W, Di Jorio L, Orain M, Bouchard N, Muanza T, Rybicki FJ, Kafi K, Huntsman D, Joubert P, Chandelier F, Routy B. Generalization optimizing machine learning to improve CT scan radiomics and assess immune checkpoint inhibitors' response in non-small cell lung cancer: a multicenter cohort study. Front Oncol 2023; 13:1196414. [PMID: 37546399 PMCID: PMC10400292 DOI: 10.3389/fonc.2023.1196414] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
Background Recent developments in artificial intelligence suggest that radiomics may represent a promising non-invasive biomarker to predict response to immune checkpoint inhibitors (ICIs). Nevertheless, validation of radiomics algorithms in independent cohorts remains a challenge due to variations in image acquisition and reconstruction. Using radiomics, we investigated the importance of scan normalization as part of a broader machine learning framework to enable model external generalizability to predict ICI response in non-small cell lung cancer (NSCLC) patients across different centers. Methods Radiomics features were extracted and compared from 642 advanced NSCLC patients on pre-ICI scans using established open-source PyRadiomics and a proprietary DeepRadiomics deep learning technology. The population was separated into two groups: a discovery cohort of 512 NSCLC patients from three academic centers and a validation cohort that included 130 NSCLC patients from a fourth center. We harmonized images to account for variations in reconstruction kernel, slice thicknesses, and device manufacturers. Multivariable models, evaluated using cross-validation, were used to estimate the predictive value of clinical variables, PD-L1 expression, and PyRadiomics or DeepRadiomics for progression-free survival at 6 months (PFS-6). Results The best prognostic factor for PFS-6, excluding radiomics features, was obtained with the combination of Clinical + PD-L1 expression (AUC = 0.66 in the discovery and 0.62 in the validation cohort). Without image harmonization, combining Clinical + PyRadiomics or DeepRadiomics delivered an AUC = 0.69 and 0.69, respectively, in the discovery cohort, but dropped to 0.57 and 0.52, in the validation cohort. This lack of generalizability was consistent with observations in principal component analysis clustered by CT scan parameters. Subsequently, image harmonization eliminated these clusters. The combination of Clinical + DeepRadiomics reached an AUC = 0.67 and 0.63 in the discovery and validation cohort, respectively. Conversely, the combination of Clinical + PyRadiomics failed generalizability validations, with AUC = 0.66 and 0.59. Conclusion We demonstrated that a risk prediction model combining Clinical + DeepRadiomics was generalizable following CT scan harmonization and machine learning generalization methods. These results had similar performances to routine oncology practice using Clinical + PD-L1. This study supports the strong potential of radiomics as a future non-invasive strategy to predict ICI response in advanced NSCLC.
Collapse
Affiliation(s)
- Marion Tonneau
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
- Université de Médecine, Lille, France
| | - Kim Phan
- Imagia Canexia Health, Montreal, QC, Canada
| | - Venkata S. K. Manem
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
- Department of Mathematics and Computer Science, University of Quebec at Trois-Rivières, Trois-Rivières, QC, Canada
| | | | | | - Suzanne Kazandjian
- Department of Medical Oncology, Jewish General Hospital, Montreal, QC, Canada
| | - Davy Vanderweyen
- Department of Radiology, Centre Hospitalier de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Justin Panasci
- Department of Medical Oncology, Jewish General Hospital, Montreal, QC, Canada
| | - Julie Malo
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
| | - François Coulombe
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
| | - Andréanne Gagné
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
| | - Arielle Elkrief
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
- Hemato-Oncology Division, Centre Hospitalier de l’université de Montreal, Montreal, QC, Canada
| | - Wiam Belkaïd
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
| | | | - Michele Orain
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
| | - Nicole Bouchard
- Department of Oncology, Centre Hospitalier de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Thierry Muanza
- Department of Medical Oncology, Jewish General Hospital, Montreal, QC, Canada
- Department of Radiation Oncology, Lady Davis Institute of the Jewish General Hospital, Montreal, QC, Canada
| | | | - Kam Kafi
- Imagia Canexia Health, Montreal, QC, Canada
| | | | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
- Department of Pathology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | | | - Bertrand Routy
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
- Hemato-Oncology Division, Centre Hospitalier de l’université de Montreal, Montreal, QC, Canada
| |
Collapse
|
37
|
Cui J, Miao X, Yanghao X, Qin X. Bibliometric research on the developments of artificial intelligence in radiomics toward nervous system diseases. Front Neurol 2023; 14:1171167. [PMID: 37360350 PMCID: PMC10288367 DOI: 10.3389/fneur.2023.1171167] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023] Open
Abstract
Background The growing interest suggests that the widespread application of radiomics has facilitated the development of neurological disease diagnosis, prognosis, and classification. The application of artificial intelligence methods in radiomics has increasingly achieved outstanding prediction results in recent years. However, there are few studies that have systematically analyzed this field through bibliometrics. Our destination is to study the visual relationships of publications to identify the trends and hotspots in radiomics research and encourage more researchers to participate in radiomics studies. Methods Publications in radiomics in the field of neurological disease research can be retrieved from the Web of Science Core Collection. Analysis of relevant countries, institutions, journals, authors, keywords, and references is conducted using Microsoft Excel 2019, VOSviewer, and CiteSpace V. We analyze the research status and hot trends through burst detection. Results On October 23, 2022, 746 records of studies on the application of radiomics in the diagnosis of neurological disorders were retrieved and published from 2011 to 2023. Approximately half of them were written by scholars in the United States, and most were published in Frontiers in Oncology, European Radiology, Cancer, and SCIENTIFIC REPORTS. Although China ranks first in the number of publications, the United States is the driving force in the field and enjoys a good academic reputation. NORBERT GALLDIKS and JIE TIAN published the most relevant articles, while GILLIES RJ was cited the most. RADIOLOGY is a representative and influential journal in the field. "Glioma" is a current attractive research hotspot. Keywords such as "machine learning," "brain metastasis," and "gene mutations" have recently appeared at the research frontier. Conclusion Most of the studies focus on clinical trial outcomes, such as the diagnosis, prediction, and prognosis of neurological disorders. The radiomics biomarkers and multi-omics studies of neurological disorders may soon become a hot topic and should be closely monitored, particularly the relationship between tumor-related non-invasive imaging biomarkers and the intrinsic micro-environment of tumors.
Collapse
|
38
|
Lee K, Liu Z, Chandran U, Kalsekar I, Laxmanan B, Higashi MK, Jun T, Ma M, Li M, Mai Y, Gilman C, Wang T, Ai L, Aggarwal P, Pan Q, Oh W, Stolovitzky G, Schadt E, Wang X. Detecting Ground Glass Opacity Features in Patients With Lung Cancer: Automated Extraction and Longitudinal Analysis via Deep Learning-Based Natural Language Processing. JMIR AI 2023; 2:e44537. [PMID: 38875565 PMCID: PMC11041451 DOI: 10.2196/44537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/30/2023] [Accepted: 03/31/2023] [Indexed: 06/16/2024]
Abstract
BACKGROUND Ground-glass opacities (GGOs) appearing in computed tomography (CT) scans may indicate potential lung malignancy. Proper management of GGOs based on their features can prevent the development of lung cancer. Electronic health records are rich sources of information on GGO nodules and their granular features, but most of the valuable information is embedded in unstructured clinical notes. OBJECTIVE We aimed to develop, test, and validate a deep learning-based natural language processing (NLP) tool that automatically extracts GGO features to inform the longitudinal trajectory of GGO status from large-scale radiology notes. METHODS We developed a bidirectional long short-term memory with a conditional random field-based deep-learning NLP pipeline to extract GGO and granular features of GGO retrospectively from radiology notes of 13,216 lung cancer patients. We evaluated the pipeline with quality assessments and analyzed cohort characterization of the distribution of nodule features longitudinally to assess changes in size and solidity over time. RESULTS Our NLP pipeline built on the GGO ontology we developed achieved between 95% and 100% precision, 89% and 100% recall, and 92% and 100% F1-scores on different GGO features. We deployed this GGO NLP model to extract and structure comprehensive characteristics of GGOs from 29,496 radiology notes of 4521 lung cancer patients. Longitudinal analysis revealed that size increased in 16.8% (240/1424) of patients, decreased in 14.6% (208/1424), and remained unchanged in 68.5% (976/1424) in their last note compared to the first note. Among 1127 patients who had longitudinal radiology notes of GGO status, 815 (72.3%) were reported to have stable status, and 259 (23%) had increased/progressed status in the subsequent notes. CONCLUSIONS Our deep learning-based NLP pipeline can automatically extract granular GGO features at scale from electronic health records when this information is documented in radiology notes and help inform the natural history of GGO. This will open the way for a new paradigm in lung cancer prevention and early detection.
Collapse
Affiliation(s)
| | | | - Urmila Chandran
- Lung Cancer Initiative, Johnson & Johnson, New Brunswick, NJ, United States
| | - Iftekhar Kalsekar
- Lung Cancer Initiative, Johnson & Johnson, New Brunswick, NJ, United States
| | - Balaji Laxmanan
- Lung Cancer Initiative, Johnson & Johnson, New Brunswick, NJ, United States
| | | | - Tomi Jun
- Sema4, Stamford, CT, United States
| | - Meng Ma
- Sema4, Stamford, CT, United States
| | | | - Yun Mai
- Sema4, Stamford, CT, United States
| | | | | | - Lei Ai
- Sema4, Stamford, CT, United States
| | | | - Qi Pan
- Sema4, Stamford, CT, United States
| | - William Oh
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Eric Schadt
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | |
Collapse
|
39
|
Jha AK, Mithun S, Sherkhane UB, Jaiswar V, Shah S, Purandare N, Prabhash K, Maheshwari A, Gupta S, Wee L, Rangarajan V, Dekker A. Development and validation of radiomic signature for predicting overall survival in advanced-stage cervical cancer. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1138552. [PMID: 39355056 PMCID: PMC11440856 DOI: 10.3389/fnume.2023.1138552] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/03/2023] [Indexed: 10/03/2024]
Abstract
Background The role of artificial intelligence and radiomics in prediction model development in cancer has been increasing every passing day. Cervical cancer is the 4th most common cancer in women worldwide, contributing to 6.5% of all cancer types. The treatment outcome of cervical cancer patients varies and individualized prediction of disease outcome is of paramount importance. Purpose The purpose of this study is to develop and validate the digital signature for 5-year overall survival prediction in cervical cancer using robust CT radiomic and clinical features. Materials and Methods Pretreatment clinical features and CT radiomic features of 68 patients, who were treated with chemoradiation therapy in our hospital, were used in this study. Radiomic features were extracted using an in-house developed python script and pyradiomic package. Clinical features were selected by the recursive feature elimination technique. Whereas radiomic feature selection was performed using a multi-step process i.e., step-1: only robust radiomic features were selected based on our previous study, step-2: a hierarchical clustering was performed to eliminate feature redundancy, and step-3: recursive feature elimination was performed to select the best features for prediction model development. Four machine algorithms i.e., Logistic regression (LR), Random Forest (RF), Support vector classifier (SVC), and Gradient boosting classifier (GBC), were used to develop 24 models (six models using each algorithm) using clinical, radiomic and combined features. Models were compared based on the prediction score in the internal validation. Results The average prediction accuracy was found to be 0.65 (95% CI: 0.60-0.70), 0.72 (95% CI: 0.63-0.81), and 0.77 (95% CI: 0.72-0.82) for clinical, radiomic, and combined models developed using four prediction algorithms respectively. The average prediction accuracy was found to be 0.69 (95% CI: 0.62-0.76), 0.79 (95% CI: 0.72-0.86), 0.71 (95% CI: 0.62-0.80), and 0.72 (95% CI: 0.66-0.78) for LR, RF, SVC and GBC models developed on three datasets respectively. Conclusion Our study shows the promising predictive performance of a robust radiomic signature to predict 5-year overall survival in cervical cancer patients.
Collapse
Affiliation(s)
- Ashish Kumar Jha
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, BARC Training School Complex, Mumbai, India
| | - Sneha Mithun
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, BARC Training School Complex, Mumbai, India
| | - Umeshkumar B Sherkhane
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, India
| | - Vinay Jaiswar
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, India
| | - Sneha Shah
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, BARC Training School Complex, Mumbai, India
| | - Nilendu Purandare
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, BARC Training School Complex, Mumbai, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, BARC Training School Complex, Mumbai, India
| | - Amita Maheshwari
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, BARC Training School Complex, Mumbai, India
| | - Sudeep Gupta
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
- Advance Center for Treatment, Research, Education in Cancer, Navi-Mumbai, India
- Homi Bhabha National Institute, BARC Training School Complex, Mumbai, India
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - V Rangarajan
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, BARC Training School Complex, Mumbai, India
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, Netherlands
| |
Collapse
|
40
|
Liu Y, Wei X, Feng X, Liu Y, Feng G, Du Y. Repeatability of radiomics studies in colorectal cancer: a systematic review. BMC Gastroenterol 2023; 23:125. [PMID: 37059990 PMCID: PMC10105401 DOI: 10.1186/s12876-023-02743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 03/22/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Recently, radiomics has been widely used in colorectal cancer, but many variable factors affect the repeatability of radiomics research. This review aims to analyze the repeatability of radiomics studies in colorectal cancer and to evaluate the current status of radiomics in the field of colorectal cancer. METHODS The included studies in this review by searching from the PubMed and Embase databases. Then each study in our review was evaluated using the Radiomics Quality Score (RQS). We analyzed the factors that may affect the repeatability in the radiomics workflow and discussed the repeatability of the included studies. RESULTS A total of 188 studies was included in this review, of which only two (2/188, 1.06%) studies controlled the influence of individual factors. In addition, the median score of RQS was 11 (out of 36), range-1 to 27. CONCLUSIONS The RQS score was moderately low, and most studies did not consider the repeatability of radiomics features, especially in terms of Intra-individual, scanners, and scanning parameters. To improve the generalization of the radiomics model, it is necessary to further control the variable factors of repeatability.
Collapse
Affiliation(s)
- Ying Liu
- School of Medical Imaging, North Sichuan Medical College, Sichuan Province, Nanchong City, 637000, China
| | - Xiaoqin Wei
- School of Medical Imaging, North Sichuan Medical College, Sichuan Province, Nanchong City, 637000, China
| | | | - Yan Liu
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China
| | - Guiling Feng
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China
| | - Yong Du
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China.
| |
Collapse
|
41
|
Chiu FY, Yen Y. Imaging biomarkers for clinical applications in neuro-oncology: current status and future perspectives. Biomark Res 2023; 11:35. [PMID: 36991494 DOI: 10.1186/s40364-023-00476-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
Biomarker discovery and development are popular for detecting the subtle diseases. However, biomarkers are needed to be validated and approved, and even fewer are ever used clinically. Imaging biomarkers have a crucial role in the treatment of cancer patients because they provide objective information on tumor biology, the tumor's habitat, and the tumor's signature in the environment. Tumor changes in response to an intervention complement molecular and genomic translational diagnosis as well as quantitative information. Neuro-oncology has become more prominent in diagnostics and targeted therapies. The classification of tumors has been actively updated, and drug discovery, and delivery in nanoimmunotherapies are advancing in the field of target therapy research. It is important that biomarkers and diagnostic implements be developed and used to assess the prognosis or late effects of long-term survivors. An improved realization of cancer biology has transformed its management with an increasing emphasis on a personalized approach in precision medicine. In the first part, we discuss the biomarker categories in relation to the courses of a disease and specific clinical contexts, including that patients and specimens should both directly reflect the target population and intended use. In the second part, we present the CT perfusion approach that provides quantitative and qualitative data that has been successfully applied to the clinical diagnosis, treatment and application. Furthermore, the novel and promising multiparametric MR imageing approach will provide deeper insights regarding the tumor microenvironment in the immune response. Additionally, we briefly remark new tactics based on MRI and PET for converging on imaging biomarkers combined with applications of bioinformatics in artificial intelligence. In the third part, we briefly address new approaches based on theranostics in precision medicine. These sophisticated techniques merge achievable standardizations into an applicatory apparatus for primarily a diagnostic implementation and tracking radioactive drugs to identify and to deliver therapies in an individualized medicine paradigm. In this article, we describe the critical principles for imaging biomarker characterization and discuss the current status of CT, MRI and PET in finiding imaging biomarkers of early disease.
Collapse
Affiliation(s)
- Fang-Ying Chiu
- Center for Cancer Translational Research, Tzu Chi University, Hualien City, 970374, Taiwan.
- Center for Brain and Neurobiology Research, Tzu Chi University, Hualien City, 970374, Taiwan.
- Teaching and Research Headquarters for Sustainable Development Goals, Tzu Chi University, Hualien City, 970374, Taiwan.
| | - Yun Yen
- Center for Cancer Translational Research, Tzu Chi University, Hualien City, 970374, Taiwan.
- Ph.D. Program for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei City, 110301, Taiwan.
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei City, 110301, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei City, 110301, Taiwan.
- Cancer Center, Taipei Municipal WanFang Hospital, Taipei City, 116081, Taiwan.
| |
Collapse
|
42
|
Elmahdy M, Sebro R. Radiomics analysis in medical imaging research. J Med Radiat Sci 2023; 70:3-7. [PMID: 36762402 PMCID: PMC9977659 DOI: 10.1002/jmrs.662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/21/2023] [Indexed: 02/11/2023] Open
Abstract
This article discusses the current research in the field of radiomics in medical imaging with emphasis on its role in fighting coronavirus disease 2019 (COVID-19). This article covers the building of radiomic models in a simple straightforward manner, while discussing radiomic models potential to help us face this pandemic.
Collapse
Affiliation(s)
- Mahmoud Elmahdy
- Department of RadiologyMayo ClinicJacksonvilleFloridaUSA,Center for Augmented IntelligenceMayo ClinicJacksonvilleFloridaUSA
| | - Ronnie Sebro
- Department of RadiologyMayo ClinicJacksonvilleFloridaUSA,Center for Augmented IntelligenceMayo ClinicJacksonvilleFloridaUSA,Department of Orthopedic SurgeryMayo ClinicJacksonvilleFloridaUSA,Department of BiostatisticsCentre for Quantitative Health SciencesJacksonvilleFloridaUSA
| |
Collapse
|
43
|
Alsyed E, Smith R, Bartley L, Marshall C, Spezi E. A heterogeneous phantom study for investigating the stability of PET images radiomic features with varying reconstruction settings. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1078536. [PMID: 39380957 PMCID: PMC11459985 DOI: 10.3389/fnume.2023.1078536] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/18/2023] [Indexed: 10/10/2024]
Abstract
The purpose of this work was to assess the capability of radiomic features in distinguishing PET image regions with different uptake patterns. Furthermore, we assessed the stability of PET radiomic features with varying image reconstruction settings. An in-house phantom was designed and constructed, consisting of homogenous and heterogenous artificial phantom inserts. Four artificially constructed inserts were placed into a water filled phantom and filled with varying levels of radioactivity to simulate homogeneous and heterogeneous uptake patterns. The phantom was imaged for 80 min. PET images were reconstructed whilst varying reconstruction parameters. The parameters adjusted included, number of ordered subsets, number of iterations, use of time-of-flight and filter cut off. Regions of interest (ROI) were established by segmentation of the phantom inserts from the reconstructed images. In total seventy eight 3D radiomic features for each ROI with unique reconstructed parameters were extracted. The Friedman test was used to determine the statistical power of each radiomic feature in differentiating phantom inserts with different hetero/homogeneous configurations. The Coefficient of Variation (COV) of each feature, with respect to the reconstruction setting was used to determine feature stability. Forty three out of seventy eight radiomic features were found to be stable (COV ≤ 5%) against all reconstruction settings. To provide any utility, stable features are required to differentiate between regions with different hetro/homogeneity. Of the forty three stable features, fifteen (35%) features showed a statistically significant difference between the artificially constructed inserts. Such features included GLCM (Difference average, Difference entropy, Dissimilarity and Inverse difference), GLRL (Long run emphasis, Grey level non uniformity and Run percentage) and NGTDM (Complexity and Strength). The finding of this work suggests that radiomic features are capable of distinguishing between radioactive distribution patterns that demonstrate different levels of heterogeneity. Therefore, radiomic features could serve as an adjuvant diagnostic tool along with traditional imaging. However, the choice of the radiomic features needs to account for variability introduced when different reconstruction settings are used. Standardization of PET image reconstruction settings across sites performing radiomic analysis in multi-centre trials should be considered.
Collapse
Affiliation(s)
- Emad Alsyed
- School of Engineering, Cardiff University, Cardiff, United Kingdom
- Department of Nuclear Engineering, Faculty of Engineering, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rhodri Smith
- Wales Research and Diagnostic Positron Emission Tomography Imaging Centre (PETIC), Cardiff University, Cardiff, United Kingdom
| | - Lee Bartley
- Wales Research and Diagnostic Positron Emission Tomography Imaging Centre (PETIC), Cardiff University, Cardiff, United Kingdom
| | - Christopher Marshall
- Wales Research and Diagnostic Positron Emission Tomography Imaging Centre (PETIC), Cardiff University, Cardiff, United Kingdom
| | - Emiliano Spezi
- School of Engineering, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
44
|
Salome P, Sforazzini F, Grugnara G, Kudak A, Dostal M, Herold-Mende C, Heiland S, Debus J, Abdollahi A, Knoll M. MR Intensity Normalization Methods Impact Sequence Specific Radiomics Prognostic Model Performance in Primary and Recurrent High-Grade Glioma. Cancers (Basel) 2023; 15:cancers15030965. [PMID: 36765922 PMCID: PMC9913466 DOI: 10.3390/cancers15030965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
PURPOSE This study investigates the impact of different intensity normalization (IN) methods on the overall survival (OS) radiomics models' performance of MR sequences in primary (pHGG) and recurrent high-grade glioma (rHGG). METHODS MR scans acquired before radiotherapy were retrieved from two independent cohorts (rHGG C1: 197, pHGG C2: 141) from multiple scanners (15, 14). The sequences are T1 weighted (w), contrast-enhanced T1w (T1wce), T2w, and T2w-FLAIR. Sequence-specific significant features (SF) associated with OS, extracted from the tumour volume, were derived after applying 15 different IN methods. Survival analyses were conducted using Cox proportional hazard (CPH) and Poisson regression (POI) models. A ranking score was assigned based on the 10-fold cross-validated (CV) concordance index (C-I), mean square error (MSE), and the Akaike information criterion (AICs), to evaluate the methods' performance. RESULTS Scatter plots of the 10-CV C-I and MSE against the AIC showed an impact on the survival predictions between the IN methods and MR sequences (C1/C2 C-I range: 0.62-0.71/0.61-0.72, MSE range: 0.20-0.42/0.13-0.22). White stripe showed stable results for T1wce (C1/C2 C-I: 0.71/0.65, MSE: 0.21/0.14). Combat (0.68/0.62, 0.22/0.15) and histogram matching (HM, 0.67/0.64, 0.22/0.15) showed consistent prediction results for T2w models. They were also the top-performing methods for T1w in C2 (Combat: 0.67, 0.13; HM: 0.67, 0.13); however, only HM achieved high predictions in C1 (0.66, 0.22). After eliminating IN impacted SF using Spearman's rank-order correlation coefficient, a mean decrease in the C-I and MSE of 0.05 and 0.03 was observed in all four sequences. CONCLUSION The IN method impacted the predictive power of survival models; thus, performance is sequence-dependent.
Collapse
Affiliation(s)
- Patrick Salome
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- Heidelberg Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Correspondence: (P.S.); (M.K.)
| | - Francesco Sforazzini
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- Heidelberg Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
| | - Gianluca Grugnara
- Department of Neuroradiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Andreas Kudak
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- CCU Radiation Therapy, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
| | - Matthias Dostal
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- CCU Radiation Therapy, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
| | - Christel Herold-Mende
- Brain Tumour Group, European Organization for Research and Treatment of Cancer, 1200 Brussels, Belgium
- Division of Neurosurgical Research, Department of Neurosurgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
| | - Amir Abdollahi
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
| | - Maximilian Knoll
- Clinical Cooperation Unit (CCU) Radiation Oncology, German Cancer Research Centre, INF 280, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core Centre Heidelberg, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Centre (HIT), INF 450, 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
- Correspondence: (P.S.); (M.K.)
| |
Collapse
|
45
|
Berger T, Noble DJ, Yang Z, Shelley LE, McMullan T, Bates A, Thomas S, Carruthers LJ, Beckett G, Duffton A, Paterson C, Jena R, McLaren DB, Burnet NG, Nailon WH. Sub-regional analysis of the parotid glands: model development for predicting late xerostomia with radiomics features in head and neck cancer patients. Acta Oncol 2023; 62:166-173. [PMID: 36802351 DOI: 10.1080/0284186x.2023.2179895] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/08/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND The irradiation of sub-regions of the parotid has been linked to xerostomia development in patients with head and neck cancer (HNC). In this study, we compared the xerostomia classification performance of radiomics features calculated on clinically relevant and de novo sub-regions of the parotid glands of HNC patients. MATERIAL AND METHODS All patients (N = 117) were treated with TomoTherapy in 30-35 fractions of 2-2.167 Gy per fraction with daily mega-voltage-CT (MVCT) acquisition for image-guidance purposes. Radiomics features (N = 123) were extracted from daily MVCTs for the whole parotid gland and nine sub-regions. The changes in feature values after each complete week of treatment were considered as predictors of xerostomia (CTCAEv4.03, grade ≥ 2) at 6 and 12 months. Combinations of predictors were generated following the removal of statistically redundant information and stepwise selection. The classification performance of the logistic regression models was evaluated on train and test sets of patients using the Area Under the Curve (AUC) associated with the different sub-regions at each week of treatment and benchmarked with the performance of models solely using dose and toxicity at baseline. RESULTS In this study, radiomics-based models predicted xerostomia better than standard clinical predictors. Models combining dose to the parotid and xerostomia scores at baseline yielded an AUCtest of 0.63 and 0.61 for xerostomia prediction at 6 and 12 months after radiotherapy while models based on radiomics features extracted from the whole parotid yielded a maximum AUCtest of 0.67 and 0.75, respectively. Overall, across sub-regions, maximum AUCtest was 0.76 and 0.80 for xerostomia prediction at 6 and 12 months. Within the first two weeks of treatment, the cranial part of the parotid systematically yielded the highest AUCtest. CONCLUSION Our results indicate that variations of radiomics features calculated on sub-regions of the parotid glands can lead to earlier and improved prediction of xerostomia in HNC patients.
Collapse
Affiliation(s)
- Thomas Berger
- Department of Oncology Physics, Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
- Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - David J Noble
- Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
- Department of Oncology, The University of Cambridge, Cambridge, UK
- Department of Clinical Oncology, Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
| | - Zhuolin Yang
- Department of Oncology Physics, Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
- School of Engineering, the University of Edinburgh, the King's Buildings, Edinburgh, UK
| | - Leila Ea Shelley
- Department of Oncology Physics, Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
| | - Thomas McMullan
- Department of Oncology Physics, Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
| | - Amy Bates
- Department of Oncology, The University of Cambridge, Cambridge, UK
| | - Simon Thomas
- Department of Medical Physics and Clinical Engineering, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Linda J Carruthers
- Department of Oncology Physics, Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
| | - George Beckett
- Edinburgh Parallel Computing Centre, Bayes Centre, Edinburgh, UK
| | | | | | - Raj Jena
- Department of Oncology, The University of Cambridge, Cambridge, UK
| | - Duncan B McLaren
- Department of Clinical Oncology, Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
| | | | - William H Nailon
- Department of Oncology Physics, Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
- School of Engineering, the University of Edinburgh, the King's Buildings, Edinburgh, UK
| |
Collapse
|
46
|
Current Role of Delta Radiomics in Head and Neck Oncology. Int J Mol Sci 2023; 24:ijms24032214. [PMID: 36768535 PMCID: PMC9916410 DOI: 10.3390/ijms24032214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
The latest developments in the management of head and neck cancer show an increasing trend in the implementation of novel approaches using artificial intelligence for better patient stratification and treatment-related risk evaluation. Radiomics, or the extraction of data from various imaging modalities, is a tool often used to evaluate specific features related to the tumour or normal tissue that are not identifiable by the naked eye and which can add value to existing clinical data. Furthermore, the assessment of feature variations from one time point to another based on subsequent images, known as delta radiomics, was shown to have even higher value for treatment-outcome prediction or patient stratification into risk categories. The information gathered from delta radiomics can, further, be used for decision making regarding treatment adaptation or other interventions found to be beneficial to the patient. The aim of this work is to collate the existing studies on delta radiomics in head and neck cancer and evaluate its role in tumour response and normal-tissue toxicity predictions alike. Moreover, this work also highlights the role of holomics, which brings under the same umbrella clinical and radiomic features, for a more complex patient characterization and treatment optimisation.
Collapse
|
47
|
Capobianco E, Dominietto M. Translating Data Science Results into Precision Oncology Decisions: A Mini Review. J Clin Med 2023; 12:438. [PMID: 36675367 PMCID: PMC9862106 DOI: 10.3390/jcm12020438] [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: 12/08/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
While reviewing and discussing the potential of data science in oncology, we emphasize medical imaging and radiomics as the leading contextual frameworks to measure the impacts of Artificial Intelligence (AI) and Machine Learning (ML) developments. We envision some domains and research directions in which radiomics should become more significant in view of current barriers and limitations.
Collapse
Affiliation(s)
- Enrico Capobianco
- The Jackson Laboratory, 10 Discovery Drive, Farmington, CT 06032, USA
| | - Marco Dominietto
- Paul Scherrer Institut, Forschungsstrasse 111, 5232 Villigen, Switzerland
| |
Collapse
|
48
|
Liao J, Li X, Gan Y, Han S, Rong P, Wang W, Li W, Zhou L. Artificial intelligence assists precision medicine in cancer treatment. Front Oncol 2023; 12:998222. [PMID: 36686757 PMCID: PMC9846804 DOI: 10.3389/fonc.2022.998222] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/22/2022] [Indexed: 01/06/2023] Open
Abstract
Cancer is a major medical problem worldwide. Due to its high heterogeneity, the use of the same drugs or surgical methods in patients with the same tumor may have different curative effects, leading to the need for more accurate treatment methods for tumors and personalized treatments for patients. The precise treatment of tumors is essential, which renders obtaining an in-depth understanding of the changes that tumors undergo urgent, including changes in their genes, proteins and cancer cell phenotypes, in order to develop targeted treatment strategies for patients. Artificial intelligence (AI) based on big data can extract the hidden patterns, important information, and corresponding knowledge behind the enormous amount of data. For example, the ML and deep learning of subsets of AI can be used to mine the deep-level information in genomics, transcriptomics, proteomics, radiomics, digital pathological images, and other data, which can make clinicians synthetically and comprehensively understand tumors. In addition, AI can find new biomarkers from data to assist tumor screening, detection, diagnosis, treatment and prognosis prediction, so as to providing the best treatment for individual patients and improving their clinical outcomes.
Collapse
Affiliation(s)
- Jinzhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Zhou
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, The Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
49
|
Radiomics approach to the condylar head for legal age classification using cone-beam computed tomography: A pilot study. PLoS One 2023; 18:e0280523. [PMID: 36656878 PMCID: PMC9851527 DOI: 10.1371/journal.pone.0280523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
Legal age estimation of living individuals is a critically important issue, and radiomics is an emerging research field that extracts quantitative data from medical images. However, no reports have proposed age-related radiomics features of the condylar head or an age classification model using those features. This study aimed to introduce a radiomics approach for various classifications of legal age (18, 19, 20, and 21 years old) based on cone-beam computed tomography (CBCT) images of the mandibular condylar head, and to evaluate the usefulness of the radiomics features selected by machine learning models as imaging biomarkers. CBCT images from 85 subjects were divided into eight age groups for four legal age classifications: ≤17 and ≥18 years old groups (18-year age classification), ≤18 and ≥19 years old groups (19-year age classification), ≤19 and ≥20 years old groups (20-year age classification) and ≤20 and ≥21 years old groups (21-year age classification). The condylar heads were manually segmented by an expert. In total, 127 radiomics features were extracted from the segmented area of each condylar head. The random forest (RF) method was utilized to select features and develop the age classification model for four legal ages. After sorting features in descending order of importance, the top 10 extracted features were used. The 21-year age classification model showed the best performance, with an accuracy of 91.18%, sensitivity of 80%, and specificity of 95.83%. Radiomics features of the condylar head using CBCT showed the possibility of age estimation, and the selected features were useful as imaging biomarkers.
Collapse
|
50
|
A Framework of Analysis to Facilitate the Harmonization of Multicenter Radiomic Features in Prostate Cancer. J Clin Med 2022; 12:jcm12010140. [PMID: 36614941 PMCID: PMC9821561 DOI: 10.3390/jcm12010140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Pooling radiomic features coming from different centers in a statistical framework is challenging due to the variability in scanner models, acquisition protocols, and reconstruction settings. To remove technical variability, commonly called batch effects, different statistical harmonization strategies have been widely used in genomics but less considered in radiomics. The aim of this work was to develop a framework of analysis to facilitate the harmonization of multicenter radiomic features extracted from prostate T2-weighted magnetic resonance imaging (MRI) and to improve the power of radiomics for prostate cancer (PCa) management in order to develop robust non-invasive biomarkers translating into clinical practice. To remove technical variability and correct for batch effects, we investigated four different statistical methods (ComBat, SVA, Arsynseq, and mixed effect). The proposed approaches were evaluated using a dataset of 210 prostate cancer (PCa) patients from two centers. The impacts of the different statistical approaches were evaluated by principal component analysis and classification methods (LogitBoost, random forest, K-nearest neighbors, and decision tree). The ComBat method outperformed all other methods by achieving 70% accuracy and 78% AUC with the random forest method to automatically classify patients affected by PCa. The proposed statistical framework enabled us to define and develop a standardized pipeline of analysis to harmonize multicenter T2W radiomic features, yielding great promise to support PCa clinical practice.
Collapse
|