1
|
Yu L, Cai Y, Lin S, Zhang H, Yu S. Quantitative MRI radiomics approach for evaluating muscular alteration in Crohn disease: development of a machine learning-nomogram composite diagnostic tool. Abdom Radiol (NY) 2025:10.1007/s00261-025-04896-x. [PMID: 40232416 DOI: 10.1007/s00261-025-04896-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: 02/09/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Emerging evidence underscores smooth muscle hyperplasia and hypertrophy, rather than fibrosis, as the defining characteristics of fibrostenotic lesions in Crohn disease (CD). However, non-invasive methods for quantifying these muscular changes have yet to be fully explored. AIMS To explore the application value of radiomics based on magnetic resonance imaging (MRI) post-contrast T1-weighted images to identify muscular alteration in CD lesions with significant inflammation. METHODS A total of 68 cases were randomly assigned in this study, with 48 cases allocated to the training dataset and the remaining 20 cases assigned to the independent test dataset. Radiomic features were extracted and constructed a diagnosis model by univariate analysis and least absolute shrinkage and selection operator (LASSO) regression. Construct a nomogram based on multivariate logistic regression analysis, integrating radiomics signature, MRI features and clinical characteristics. RESULTS The radiomics model constructed based on the selected features of the post-contrasted T1-weighted images has good diagnostic performance, which yielded a sensitivity of 0.880, a specificity of 0.783, and an accuracy of 0.833 [AUC = 0.856, 95% confidence interval (CI) = 0.765-0.947]. Moreover, the nomogram representing the integrated model achieved good discrimination performances, which yielded a sensitivity of 0.836, a specificity of 0.892, and an accuracy of 0.864 (AUC = 0.926, 95% CI = 0.865-0.988), and it was better than that of the radiomics model alone. CONCLUSIONS The radiomics based on post-contrasted T1-weighted images provides additional biomarkers for Crohn disease. Additionally, integrating DCE-MRI, radiomics, and clinical data into a comprehensive model significantly improves diagnostic accuracy for identifying muscular alteration.
Collapse
Affiliation(s)
- Lin Yu
- Department of Radiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yong Cai
- Department of Radiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Shaowei Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Huijuan Zhang
- Department of Radiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Shun Yu
- Department of Radiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China.
- Fujian Provincial Key Laboratory of Medical Big Data Engineering, Fuzhou, Fujian, China.
| |
Collapse
|
2
|
Isemoto K, Waseda Y, Fujiwara M, Kimura K, Hirahara D, Saho T, Takaya E, Arita Y, Kwee TC, Fukuda S, Tanaka H, Yoshida S, Fujii Y. Predictive Potential of Contrast-Enhanced MRI-Based Delta-Radiomics for Chemoradiation Responsiveness in Muscle-Invasive Bladder Cancer. Diagnostics (Basel) 2025; 15:801. [PMID: 40218151 PMCID: PMC11988543 DOI: 10.3390/diagnostics15070801] [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: 02/24/2025] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Delta-radiomics involves analyzing feature variations at different acquisition time-points. This study aimed to assess the utility of delta-radiomics feature analysis applied to contrast-enhanced (CE) and non-contrast-enhanced (NE) T1-weighted images (WI) in predicting the therapeutic response to chemoradiotherapy (CRT) in patients diagnosed with muscle-invasive bladder cancer (MIBC). Methods: Forty-three patients with non-metastatic MIBC (cT2-4N0M0) who underwent partial or radical cystectomy after induction CRT were, retrospectively, reviewed. Pathological complete response (pCR) to CRT was defined as the absence of residual viable tumor cells in the cystectomy specimen. Identical volumes of interest corresponding to the index bladder cancer lesions on CE- and NE-T1WI on pre-therapeutic 1.5-T MRI were collaboratively delineated by one radiologist and one urologist. Texture analysis was performed using "LIFEx" software. The subtraction of radiological features between CE- and NE-T1WI yielded 112 delta-radiomics features, which were utilized in multiple machine-learning algorithms to construct optimal predictive models for CRT responsiveness. Additionally, the predictive performance of the radiomics model constructed using CE-T1WI alone was assessed. Results: Twenty-one patients (49%) achieved pCR. The best-performing delta-radiomics model, employing the "Extreme Gradient Boosting" algorithm, yielded an area under the receiver operating characteristic curve (AUC) of 0.85 (95% confidence interval [CI]: 0.75-0.95), utilizing four signal intensity-based delta-radiomics features. This outperformed the best model derived from CE-T1WI alone (AUC: 0.63, 95% CI: 0.50-0.75), which incorporated two morphological features and one signal intensity-based radiomics feature. Conclusions: Delta-radiomics analysis applied to pre-therapeutic CE- and NE-MRI demonstrated promising predictive ability for CRT responsiveness prior to treatment initiation.
Collapse
Affiliation(s)
- Kohei Isemoto
- Department of Urology, Institute of Science Tokyo, Tokyo 113-8519, Japan; (K.I.); (M.F.); (S.F.); (H.T.); (S.Y.); (Y.F.)
| | - Yuma Waseda
- Department of Urology, Insured Medical Care Management, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Motohiro Fujiwara
- Department of Urology, Institute of Science Tokyo, Tokyo 113-8519, Japan; (K.I.); (M.F.); (S.F.); (H.T.); (S.Y.); (Y.F.)
| | - Koichiro Kimura
- Department of Radiology, Institute of Science Tokyo, Tokyo 113-8519, Japan;
| | - Daisuke Hirahara
- Department of Management Planning Division, Harada Academy, Kagoshima 891-0113, Japan;
| | - Tatsunori Saho
- Department of Radiological Technology, Kokura Memorial Hospital, Kitakyushu 802-8555, Japan;
| | - Eichi Takaya
- AI Lab, Tohoku University Hospital, Sendai 980-8574, Japan;
| | - Yuki Arita
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Thomas C. Kwee
- Department of Radiology, Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Boston, MA 02114, USA;
| | - Shohei Fukuda
- Department of Urology, Institute of Science Tokyo, Tokyo 113-8519, Japan; (K.I.); (M.F.); (S.F.); (H.T.); (S.Y.); (Y.F.)
| | - Hajime Tanaka
- Department of Urology, Institute of Science Tokyo, Tokyo 113-8519, Japan; (K.I.); (M.F.); (S.F.); (H.T.); (S.Y.); (Y.F.)
| | - Soichiro Yoshida
- Department of Urology, Institute of Science Tokyo, Tokyo 113-8519, Japan; (K.I.); (M.F.); (S.F.); (H.T.); (S.Y.); (Y.F.)
| | - Yasuhisa Fujii
- Department of Urology, Institute of Science Tokyo, Tokyo 113-8519, Japan; (K.I.); (M.F.); (S.F.); (H.T.); (S.Y.); (Y.F.)
| |
Collapse
|
3
|
Crimì F, Turatto F, D'Alessandro C, Sussan G, Iacobone M, Torresan F, Tizianel I, Campi C, Quaia E, Caccese M, Ceccato F. Texture analysis can predict response to etoposide-doxorubicin-cisplatin in patients with adrenocortical carcinoma. J Endocrinol Invest 2025; 48:711-720. [PMID: 39382628 PMCID: PMC11876227 DOI: 10.1007/s40618-024-02476-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND The adrenocortical carcinoma (ACC) is a rare and highly aggressive malignancy originating from the adrenal cortex. These patients usually undergo chemotherapy with etoposide, doxorubicin, cisplatin and mitotane (EDP-M) in case of locally advanced or metastatic ACC. Computed tomography (CT) radiomics showed to be useful in adrenal pathologies. The study aimed to analyze the association between response to EDP-M treatment and CT textural features at diagnosis in patients with locally advanced or metastatic ACCs. METHODS We enrolled 17 patients with advanced or metastatic ACC who underwent CT before and after EDP-M therapy. The response to treatment was evaluated according to RECIST 1.1, Choi, and volumetric criteria. Based on the aforementioned criteria, the patients were classified as responders and not responders. Textural features were extracted from the biggest lesion in contrast-enhanced CT images with LifeX software. ROC curves were drawn for the variables that were significantly different (p < 0.05) between the two groups. RESULTS Long-run high grey level emphasis (LRHGLE_GLRLM) and histogram kurtosis were significantly different between responder and not responder groups (p = 0.04) and the multivariate ROC curve combining the two features showed a very good AUC (0.900; 95%IC: 0.724-1.000) in discriminating responders from not responders. More heterogeneous tissue texture of initial staging CT in locally advanced or metastatic ACC could predict the positive response to EDP-M treatment. CONCLUSIONS Adrenal texture is able to predict the response to EDP-M therapy in patients with advanced ACC.
Collapse
Affiliation(s)
- Filippo Crimì
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Institute of Radiology, University-Hospital of Padova, Padova, Italy
| | - Francesca Turatto
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Institute of Radiology, University-Hospital of Padova, Padova, Italy
| | - Carlo D'Alessandro
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Institute of Radiology, University-Hospital of Padova, Padova, Italy
| | - Giovanni Sussan
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Institute of Radiology, University-Hospital of Padova, Padova, Italy
| | - Maurizio Iacobone
- Endocrine Surgery Unit, Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padova, 35128, Italy
| | - Francesca Torresan
- Endocrine Surgery Unit, Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padova, 35128, Italy
| | - Irene Tizianel
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Endocrinology Unit, Department of Medicine-DIMED, University of Padova, Via Ospedale Civile, Padova, 105 - 35128, Italy
| | - Cristina Campi
- Department of Mathematics, University of Genoa, Genoa, Italy
- Life Science Computational Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genoa, Italy
| | - Emilio Quaia
- Department of Medicine-DIMED, University of Padova, Padova, Italy
- Institute of Radiology, University-Hospital of Padova, Padova, Italy
| | - Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Filippo Ceccato
- Department of Medicine-DIMED, University of Padova, Padova, Italy.
- Endocrinology Unit, Department of Medicine-DIMED, University of Padova, Via Ospedale Civile, Padova, 105 - 35128, Italy.
| |
Collapse
|
4
|
Zhang L, Jin Z, Yang F, Guo Y, Liu Y, Chen M, Xu S, Lin Z, Sun P, Yang M, Zhang P, Tao K, Zhang T, Li X, Zheng C. Added value of histogram analysis of intravoxel incoherent motion and diffusion kurtosis imaging for the evaluation of complete response to neoadjuvant therapy in locally advanced rectal cancer. Eur Radiol 2025; 35:1669-1678. [PMID: 39297948 PMCID: PMC11835893 DOI: 10.1007/s00330-024-11081-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/05/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024]
Abstract
OBJECTIVE To evaluate how intravoxel incoherent motion (IVIM) and diffusion kurtosis imaging (DKI) histogram analysis contribute to assessing complete response (CR) to neoadjuvant therapy (NAT) in locally advanced rectal cancer (LARC). MATERIAL AND METHODS In this prospective study, participants with LARC, who underwent NAT and subsequent surgery, with adequate MR image quality, were enrolled from November 2021 to March 2023. Conventional MRI (T2WI and DWI), IVIM, and DKI were performed before NAT (pre-NAT) and within two weeks before surgery (post-NAT). Image evaluation was independently performed by two experienced radiologists. Pathological complete response (pCR) was used as the reference standard. An IVIM-DKI-added model (a combination of IVIM and DKI histogram parameters with T2WI and DWI) was constructed. Receiver operating characteristic (ROC) curves were generated to evaluate the diagnostic performance of conventional MRI and the IVIM-DKI-added model. RESULTS A total of 59 participants (median age: 58.00 years [IQR: 52.00, 62.00]; 38 [64%] men) were evaluated, including 21 pCR and 38 non-pCR cases. The histogram parameters of DKI, including skewness of kurtosis post-NAT (post-KSkewness) and root mean squared of change ratio of diffusivity (Δ%DDKI-root mean squared), were entered into the IVIM-DKI-added model. The area under the ROC curve (AUC) of the IVIM-DKI-added model for assessing CR to NAT was significantly higher than that of conventional MRI (0.855 [95% CI: 0.749-0.960] vs 0.685 [95% CI: 0.565-0.806], p < 0.001). CONCLUSION IVIM and DKI provide added value in the evaluation of CR to NAT in LARC. KEY POINTS Question The current conventional imaging evaluation system lacks adequacy for assessing CR to NAT in LARC. Findings Significantly improved diagnostic performance was observed with the histogram analysis of IVIM and DKI in conjunction with conventional MRI. Clinical relevance IVIM and DKI provide significant value in evaluating CR to NAT in LARC, which bears significant implications for reducing surgical complications and facilitating organ preservation.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
| | - Ziwei Jin
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
| | - Fan Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
| | - Yiwan Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
| | - Yuan Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
| | - Manman Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
| | - Si Xu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China
| | - Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, Hubei, 430022, China
| | - Peng Sun
- Clinical and Technical Support, Philips Healthcare, Beijing, 100600, China
| | - Ming Yang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, Hubei, 430022, China
| | - Xin Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China.
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, China.
| |
Collapse
|
5
|
Lee Y, Bandara WR, Park S, Lee M, Seo C, Yang S, Lim KJ, Moe SM, Warden SJ, Surowiec RK. Integrating deep learning and machine learning for improved CKD-related cortical bone assessment in HRpQCT images: A pilot study. Bone Rep 2025; 24:101821. [PMID: 39866530 PMCID: PMC11763521 DOI: 10.1016/j.bonr.2024.101821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/18/2024] [Accepted: 12/24/2024] [Indexed: 01/28/2025] Open
Abstract
High resolution peripheral quantitative computed tomography (HRpQCT) offers detailed bone geometry and microarchitecture assessment, including cortical porosity, but assessing chronic kidney disease (CKD) bone images remains challenging. This proof-of-concept study merges deep learning and machine learning to 1) improve automatic segmentation, particularly in cases with severe cortical porosity and trabeculated endosteal surfaces, and 2) maximize image information using machine learning feature extraction to classify CKD-related skeletal abnormalities, surpassing conventional DXA and CT measures. We included 30 individuals (20 non-CKD, 10 stage 3 to 5D CKD) who underwent HRpQCT of the distal and diaphyseal radius and tibia and contributed data to develop and validate four different AI models for each anatomical site. Manually annotated cortical bone was used to train each segmentation deep-learning model. Textural features were extracted via Gray-Level Co-occurrence Matrix (GLCM) and classified as CKD or non-CKD using XGBoost with each segmentation model. For comparison, manufacturer-supplied segmentation was used to extract cortical geometry, microarchitecture, and finite element analysis (FEA) outcomes. Model performance was confirmed using the test dataset and a separate independent validation cohort which included HRpQCT imaging from 42 additional individuals (18 non-CKD, 24 CKD stage 5D). For segmentation, the diaphyseal location showed strong performance on test datasets, with Mean IoUs of 0.96 and 0.95, and accuracies of 0.97 for both radius and tibia sites in CKD. Model 4 developed from the diaphyseal tibia region excelled in classifying test and independent validation datasets, achieving F1 scores of 0.99 and 0.96, AUCs of 0.99 and 0.94, sensitivities of 0.99, and specificities of 0.99 and 0.92. No single parameter, including BMD and cortical porosity, among conventional CT outcomes consistently differentiated CKD from non-CKD across all anatomical sites. Integrating HRpQCT with deep and machine learning, this innovative approach enables precise automatic segmentation of severely deteriorated endocortical surfaces and enhances sensitivity to CKD-related cortical bone changes compared to standard DXA and HRpQCT outcomes.
Collapse
Affiliation(s)
- Youngjun Lee
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| | - Wikum R. Bandara
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| | - Sangjun Park
- Department of Radiation Therapy, Samsung Medical Center, The Sungkyunkwan University of Korea, South Korea
| | - Miran Lee
- Department of Electronic Engineering, Sogang University, Cardiac Sonographer at the Department of Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Choongboem Seo
- Department of Radiology, Seoul National University Bundang Hospital, The Seoul National University of Korea, South Korea
| | - Sunwoo Yang
- Department of Radiological Technology, Shingu University, South Korea
| | - Kenneth J. Lim
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Sharon M. Moe
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Stuart J. Warden
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Indianapolis, Indianapolis, IN, United States of America
| | - Rachel K. Surowiec
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| |
Collapse
|
6
|
Donci DD, Solomon C, Băciuț M, Dinu C, Stoia S, Rusu GM, Csutak C, Lenghel LM, Ciurea A. The Role of MRI Radiomics Using T2-Weighted Images and the Apparent Diffusion Coefficient Map for Discriminating Between Warthin's Tumors and Malignant Parotid Gland Tumors. Cancers (Basel) 2025; 17:620. [PMID: 40002215 PMCID: PMC11852730 DOI: 10.3390/cancers17040620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Differentiating between benign and malignant parotid gland tumors (PGT) is essential for establishing the treatment strategy, which is greatly influenced by the tumor's histology. The objective of this study was to evaluate the role of MRI-based radiomics in the differentiation between Warthin's tumors (WT) and malignant tumors (MT), two entities that proved to present overlapping imaging features on conventional and functional MRI sequences. METHODS In this retrospective study, a total of 106 PGT (66 WT, 40 MT) with confirmed histology were eligible for radiomic analysis, which were randomly split into a training group (79 PGT; 49 WT; 30 MT) and a testing group (27 PGT; 17 WT, 10 MT). The radiomic features were extracted from 3D segmentations of PGT performed on the following sequences: PROPELLER T2-weighted images and the ADC map, using a dedicated software. First- and second-order features were derived for each lesion, using original and filtered images. RESULTS After employing several feature reduction techniques, including LASSO regression, three final radiomic parameters were identified to be the most significant in distinguishing between the two studied groups, with fair AUC values that ranged between 0.703 and 0.767. All three radiomic features were used to construct a Radiomic Score that presented the highest diagnostic performance in distinguishing between WT and MT, achieving an AUC of 0.785 in the training set, and 0.741 in the testing set. CONCLUSIONS MRI-based radiomic features have the potential to serve as promising novel imaging biomarkers for discriminating between Warthin's tumors and malignant tumors in the parotid gland. Nevertheless, it is still to prove how radiomic features can consistently achieve higher diagnostic performance, and if they can outperform alternative imaging methods, ideally in larger, multicentric studies.
Collapse
Affiliation(s)
- Delia Doris Donci
- Radiology and Imaging Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Carolina Solomon
- Radiology and Imaging Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Mihaela Băciuț
- Oro-Maxillo-Facial Surgery Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Cristian Dinu
- Oro-Maxillo-Facial Surgery Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Sebastian Stoia
- Oro-Maxillo-Facial Surgery Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Georgeta Mihaela Rusu
- Radiology and Imaging Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Csaba Csutak
- Radiology and Imaging Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Lavinia Manuela Lenghel
- Radiology and Imaging Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Anca Ciurea
- Radiology and Imaging Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
7
|
Miyamoto Y, Nakaura T, Ohuchi M, Ogawa K, Kato R, Maeda Y, Eto K, Iwatsuki M, Baba Y, Hirai T, Baba H. Radiomics-based Machine Learning Approach to Predict Chemotherapy Responses in Colorectal Liver Metastases. J Anus Rectum Colon 2025; 9:117-126. [PMID: 39882217 PMCID: PMC11772800 DOI: 10.23922/jarc.2024-077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/15/2024] [Indexed: 01/31/2025] Open
Abstract
Objectives This study explored the clinical utility of CT radiomics-driven machine learning as a predictive marker for chemotherapy response in colorectal liver metastasis (CRLM) patients. Methods We included 150 CRLM patients who underwent first-line doublet chemotherapy, dividing them into a training cohort (n=112) and a test cohort (n=38). We manually delineated three-dimensional tumor volumes, selecting the largest liver metastasis for measurement, using pretreatment portal-phase CT images and extracted 107 radiomics features. Treatment response was classified as responder (complete or partial response) or non-responder (stable or progressive disease), based on the best overall response according to RECIST criteria, version 1.1. Employing Random Forest and Boruta algorithms, we identified significant features for responder-non-responder differentiation. Radiomics signatures were developed and validated in the training cohort using five-fold cross-validation, and performance was assessed using the area under the curve (AUC). Results Among the patients, 91 (61%) were responders and 59 (39%) were non-responders. Variable selection with Boruta revealed three key parameters ("DependenceVariance," "ClusterShade," and "RunVariance"). In the training cohort, individual CT texture parameter AUCs ranged from 0.4 to 0.65, while the machine learning analysis incorporating all valid parameters exhibited a significantly higher AUC of 0.94 (p<0.01). The validation cohort also demonstrated strong predictive accuracy, with an AUC of 0.87 for treatment response. Conclusions This study highlights the potential of CT radiomics-driven machine learning in predicting chemotherapy responses among CRLM patients.
Collapse
Affiliation(s)
- Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Nakaura
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Mayuko Ohuchi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsuhiro Ogawa
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Rikako Kato
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuto Maeda
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kojiro Eto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Division of Translational Research and Advanced Treatment Against Gastrointestinal Cancer, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Next-Generation Surgical Therapy Development, Kumamoto University, Kumamoto, Japan
| | - Toshinori Hirai
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
8
|
Pietsch FL, Haag F, Ayx I, Grawe F, Vellala AK, Schoenberg SO, Froelich MF, Tharmaseelan H. Textural heterogeneity of liver lesions in CT imaging - comparison of colorectal and pancreatic metastases. Abdom Radiol (NY) 2024; 49:4295-4306. [PMID: 39115682 PMCID: PMC11522118 DOI: 10.1007/s00261-024-04511-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE Tumoral heterogeneity poses a challenge for personalized cancer treatments. Especially in metastasized cancer, it remains a major limitation for successful targeted therapy, often leading to drug resistance due to tumoral escape mechanisms. This work explores a non-invasive radiomics-based approach to capture textural heterogeneity in liver lesions and compare it between colorectal cancer (CRC) and pancreatic cancer (PDAC). MATERIALS AND METHODS In this retrospective single-center study 73 subjects (42 CRC, 31 PDAC) with 1291 liver metastases (430 CRC, 861 PDAC) were segmented fully automated on contrast-enhanced CT images by a UNet for medical images. Radiomics features were extracted using the Python package Pyradiomics. The mean coefficient of variation (CV) was calculated patient-wise for each feature to quantify the heterogeneity. An unpaired t-test identified features with significant differences in feature variability between CRC and PDAC metastases. RESULTS In both colorectal and pancreatic liver metastases, interlesional heterogeneity in imaging can be observed using quantitative imaging features. 75 second-order features were extracted to compare the varying textural characteristics. In total, 18 radiomics features showed a significant difference (p < 0.05) in their expression between the two malignancies. Out of these, 16 features showed higher levels of variability within the cohort of pancreatic metastases, which, as illustrated in a radar plot, suggests greater textural heterogeneity for this entity. CONCLUSIONS Radiomics has the potential to identify the interlesional heterogeneity of CT texture among individual liver metastases. In this proof-of-concept study for the quantification and comparison of imaging-related heterogeneity in liver metastases a variation in the extent of heterogeneity levels in CRC and PDAC liver metastases was shown.
Collapse
Affiliation(s)
- Friedrich L Pietsch
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Florian Haag
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Isabelle Ayx
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Freba Grawe
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Abhinay K Vellala
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Stefan O Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Matthias F Froelich
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Hishan Tharmaseelan
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
9
|
Zhang J, Zhang Q, Zhao B, Shi G. Deep learning nomogram for predicting neoadjuvant chemotherapy response in locally advanced gastric cancer patients. Abdom Radiol (NY) 2024; 49:3780-3796. [PMID: 38796795 PMCID: PMC11519172 DOI: 10.1007/s00261-024-04331-7] [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: 03/07/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE Developed and validated a deep learning radiomics nomogram using multi-phase contrast-enhanced computed tomography (CECT) images to predict neoadjuvant chemotherapy (NAC) response in locally advanced gastric cancer (LAGC) patients. METHODS This multi-center study retrospectively included 322 patients diagnosed with gastric cancer from January 2013 to June 2023 at two hospitals. Handcrafted radiomics technique and the EfficientNet V2 neural network were applied to arterial, portal venous, and delayed phase CT images to extract two-dimensional handcrafted and deep learning features. A nomogram model was built by integrating the handcrafted signature, the deep learning signature, with clinical features. Discriminative ability was assessed using the receiver operating characteristics (ROC) curve and the precision-recall (P-R) curve. Model fitting was evaluated using calibration curves, and clinical utility was assessed through decision curve analysis (DCA). RESULTS The nomogram exhibited excellent performance. The area under the ROC curve (AUC) was 0.848 [95% confidence interval (CI), 0.793-0.893)], 0.802 (95% CI 0.688-0.889), and 0.751 (95% CI 0.652-0.833) for the training, internal validation, and external validation sets, respectively. The AUCs of the P-R curves were 0.838 (95% CI 0.756-0.895), 0.541 (95% CI 0.329-0.740), and 0.556 (95% CI 0.376-0.722) for the corresponding sets. The nomogram outperformed the clinical model and handcrafted signature across all sets (all P < 0.05). The nomogram model demonstrated good calibration and provided greater net benefit within the relevant threshold range compared to other models. CONCLUSION This study created a deep learning nomogram using CECT images and clinical data to predict NAC response in LAGC patients undergoing surgical resection, offering personalized treatment insights.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Radiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Qiang Zhang
- Department of Radiation Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, People's Republic of China
| | - Bo Zhao
- Department of Medical Imaging, The First Hospital of Qinhuangdao, Qinhuangdao, People's Republic of China
| | - Gaofeng Shi
- Department of Radiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China.
| |
Collapse
|
10
|
Li H, Zhuang Y, Yuan W, Gu Y, Dai X, Li M, Chen H, Zhou H. Radiomics in precision medicine for colorectal cancer: a bibliometric analysis (2013-2023). Front Oncol 2024; 14:1464104. [PMID: 39558950 PMCID: PMC11571149 DOI: 10.3389/fonc.2024.1464104] [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: 07/28/2024] [Accepted: 10/04/2024] [Indexed: 11/20/2024] Open
Abstract
Background The incidence and mortality of colorectal cancer (CRC) have been rising steadily. Early diagnosis and precise treatment are essential for improving patient survival outcomes. Over the past decade, the integration of artificial intelligence (AI) and medical imaging technologies has positioned radiomics as a critical area of research in the diagnosis, treatment, and prognosis of CRC. Methods We conducted a comprehensive review of CRC-related radiomics literature published between 1 January 2013 and 31 December 2023 using the Web of Science Core Collection database. Bibliometric tools such as Bibliometrix, VOSviewer, and CiteSpace were employed to perform an in-depth bibliometric analysis. Results Our search yielded 1,226 publications, revealing a consistent annual growth in CRC radiomics research, with a significant rise after 2019. China led in publication volume (406 papers), followed by the United States (263 papers), whereas the United States dominated in citation numbers. Notable institutions included General Electric, Harvard University, University of London, Maastricht University, and the Chinese Academy of Sciences. Prominent researchers in this field are Tian J from the Chinese Academy of Sciences, with the highest publication count, and Ganeshan B from the University of London, with the most citations. Journals leading in publication and citation counts are Frontiers in Oncology and Radiology. Keyword and citation analysis identified deep learning, texture analysis, rectal cancer, image analysis, and management as prevailing research themes. Additionally, recent trends indicate the growing importance of AI and multi-omics integration, with a focus on improving precision medicine applications in CRC. Emerging keywords such as deep learning and AI have shown rapid growth in citation bursts over the past 3 years, reflecting a shift toward more advanced technological applications. Conclusions Radiomics plays a crucial role in the clinical management of CRC, providing valuable insights for precision medicine. It significantly contributes to predicting molecular biomarkers, assessing tumor aggressiveness, and monitoring treatment efficacy. Future research should prioritize advancing AI algorithms, enhancing multi-omics data integration, and further expanding radiomics applications in CRC precision medicine.
Collapse
Affiliation(s)
- Hao Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yupei Zhuang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Weichen Yuan
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yutian Gu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinyan Dai
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Muhan Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Haibin Chen
- Science and Technology Department, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongguang Zhou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
11
|
Xu Z, Jiang G, Dai J. Tumor therapeutics in the era of "RECIST": past, current insights, and future prospects. Oncol Rev 2024; 18:1435922. [PMID: 39493769 PMCID: PMC11527623 DOI: 10.3389/or.2024.1435922] [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: 05/21/2024] [Accepted: 08/30/2024] [Indexed: 11/05/2024] Open
Abstract
In recent years, advancements in medical treatment and imaging technologies have revolutionized the assessment of tumor response. However, the Response Evaluation Criteria in Solid Tumors (RECIST) has long been established as the gold standard for evaluating tumor treatment. As treatment modalities evolve, the need for continuous refinement and adaptation of RECIST becomes increasingly apparent. This review explores the historical evolution, current applications, limitations, and future directions of RECIST. It discusses the challenges of distinguishing true progression from pseudo-progression in ICIs (immune checkpoint inhibitors), the integration of advanced imaging tools, and the necessity for RECIST criteria tailored to specific therapies like neoadjuvant treatments. The review highlights the ongoing efforts to enhance RECIST's accuracy and reliability in clinical decision-making and the potential for developing new standards to better evaluate treatment efficacy in the rapidly evolving landscape of oncology.
Collapse
Affiliation(s)
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Dai
- *Correspondence: Gening Jiang, ; Jie Dai,
| |
Collapse
|
12
|
Ma T, Zhao M, Li X, Song X, Wang L, Ye Z. A machine learning based radiomics approach for predicting No. 14v station lymph node metastasis in gastric cancer. Front Med (Lausanne) 2024; 11:1464632. [PMID: 39493708 PMCID: PMC11527654 DOI: 10.3389/fmed.2024.1464632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/04/2024] [Indexed: 11/05/2024] Open
Abstract
Purpose To evaluate the potential of radiomics approach for predicting No. 14v station lymph node metastasis (14vM) in gastric cancer (GC). Methods The contrast enhanced CT (CECT) images with corresponding clinical information of 288 GC patients were retrospectively collected. Patients were separated into training set (n = 202) and testing set (n = 86). A total of 1,316 radiomics feature were extracted from portal venous phase images of CECT. Seven machine learning (ML) algorithms including naïve Bayes (NB), k-nearest neighbor (KNN), decision tree (DT), logistic regression (LR), random forest (RF), eXtreme gradient boosting (XGBoost) and support vector machine (SVM) were trained for development of optimal radiomics signature. A combined model was established by combining radiomics with important clinicopathological factors. The diagnostic ability of the signature and model were evaluated. Results LR algorithm was chosen for signature construction. The radiomics signature exhibited good discrimination accuracy of 14vM with AUCs of 0.83 in the training and 0.77 in the testing set. The risk of 14vM showed significant association with higher radiomics score. A combined model exhibited increased predictive ability and good agreement in the training (AUC = 0.87) and testing (AUC = 0.85) sets. Conclusion The ML-based radiomics model provided a promising image biomarker for preoperative detection of 14vM and may help the surgeon to decide whether to add 14v dissection to lymphadenectomy.
Collapse
Affiliation(s)
- Tingting Ma
- Department of Radiology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Mengran Zhao
- Department of Radiology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiangli Li
- Health Management Center, Weifang People’s Hospital, Weifang, China
| | - Xiangchao Song
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lingwei Wang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
13
|
Li J, Zhou M, Tong Y, Chen H, Su R, Tao Y, Zhang G, Sun Z. Tumor Growth Pattern and Intra- and Peritumoral Radiomics Combined for Prediction of Initial TACE Outcome in Patients with Primary Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:1927-1944. [PMID: 39398867 PMCID: PMC11471153 DOI: 10.2147/jhc.s480554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
Purpose Non-invasive methods are urgently needed to assess the efficacy of transarterial chemoembolization (TACE) and to identify patients with hepatocellular carcinoma (HCC) who may benefit from this procedure. This study, therefore, aimed to investigate the predictive ability of tumor growth patterns and radiomics features from contrast-enhanced magnetic resonance imaging (CE-MRI) in predicting tumor response to TACE among patients with HCC. Patients and Methods A retrospective study was conducted on 133 patients with HCC who underwent TACE at three centers between January 2015 and April 2023. Enrolled patients were divided into training, testing, and validation cohorts. Rim arterial phase hyperenhancement (Rim APHE), tumor growth patterns, nonperipheral washout, markedly low apparent diffusion coefficient (ADC) value, intratumoral arteries, and clinical baseline features were documented for all patients. Radiomics features were extracted from the intratumoral and peritumoral regions across the three phases of CE-MRI. Seven prediction models were developed, and their performances were evaluated using receiver operating characteristic (ROC) and decision curve analysis (DCA). Results Tumor growth patterns and albumin-bilirubin (ALBI) score were significantly correlated with tumor response. Tumor growth patterns also showed a positive correlation with tumor burden (r = 0.634, P = 0.000). The Peritumor (AUC = 0.85, 0.71, and 0.77), Clinics_Peritumor (AUC = 0.86, 0.77, and 0.81), and Tumor_Peritumor (AUC = 0.87, 0.77, and 0.80) models significantly outperformed the Clinics and Tumor models (P < 0.05), while the Clinics_Tumor_Peritumor model (AUC = 0.88, 0.81, and 0.81) outperformed the Clinics (AUC = 0.67, 0.77, and 0.75), Tumor (AUC = 0.78, 0.72, and 0.68), and Clinics_Tumor (AUC = 0.82, 0.83, and 0.78) models (P < 0.05 or 0.053, respectively). The DCA curve demonstrated better predictive performance within a specific threshold probability range for Clinics_Tumor_Peritumor. Conclusion Combining tumor growth patterns, intra- and peri-tumoral radiomics features, and ALBI score could be a robust tool for non-invasive and personalized prediction of treatment response to TACE in patients with HCC.
Collapse
Affiliation(s)
- Jiaying Li
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, People's Republic of China
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Minhui Zhou
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Yahan Tong
- Department of Radiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310005, People's Republic of China
| | - Haibo Chen
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, People's Republic of China
| | - Ruisi Su
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Yinghui Tao
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Guodong Zhang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, People's Republic of China
| | - Zhichao Sun
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, People's Republic of China
| |
Collapse
|
14
|
Alqahtani A, Bhattacharjee S, Almopti A, Li C, Nabi G. Radiomics-Based Computed Tomography Urogram Approach for the Prediction of Survival and Recurrence in Upper Urinary Tract Urothelial Carcinoma. Cancers (Basel) 2024; 16:3119. [PMID: 39335090 PMCID: PMC11429600 DOI: 10.3390/cancers16183119] [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: 07/27/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Upper tract urothelial carcinoma (UTUC) is a rare and aggressive malignancy with a poor prognosis. The accurate prediction of survival and recurrence in UTUC is crucial for effective risk stratification and guiding therapeutic decisions. Models combining radiomics and clinicopathological data features derived from computed tomographic urograms (CTUs) can be a way to predict survival and recurrence in UTUC. Thus, preoperative CTUs and clinical data were analyzed from 106 UTUC patients who underwent radical nephroureterectomy. Radiomics features were extracted from segmented tumors, and the Least Absolute Shrinkage and Selection Operator (LASSO) method was used to select the most relevant features. Multivariable Cox models combining radiomics features and clinical factors were developed to predict the survival and recurrence. Harrell's concordance index (C-index) was applied to evaluate the performance and survival distribution analyses were assessed by a Kaplan-Meier analysis. The significant outcome predictors were identified by multivariable Cox models. The combined model achieved a superior predictive accuracy (C-index: 0.73) and higher recurrence prediction (C-index: 0.84). The Kaplan-Meier analysis showed significant differences in the survival (p < 0.0001) and recurrence (p < 0.002) probabilities for the combined datasets. The CTU-based radiomics models effectively predicted survival and recurrence in the UTUC patients, and enhanced the prognostic performance by combining radiomics features with clinical factors.
Collapse
Affiliation(s)
- Abdulsalam Alqahtani
- School of Medicine, Centre for Medical Engineering and Technology, University of Dundee, Dundee DD1 9SY, UK
- Radiology Department, College of Applied Medical Sciences, Najran University, Najran 55461, Saudi Arabia
| | - Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin, D04 W6F6 Dublin, Ireland
| | - Abdulrahman Almopti
- School of Medicine, Centre for Medical Engineering and Technology, University of Dundee, Dundee DD1 9SY, UK
| | - Chunhui Li
- School of Science and Engineering, University of Dundee, Dundee DD1 4HN, UK
| | - Ghulam Nabi
- School of Medicine, Centre for Medical Engineering and Technology, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
15
|
Chen YF, Liu L, Lyu B, Yang Y, Zheng SS, Huang X, Xu Y, Fan YH. Role of artificial intelligence in Crohn's disease intestinal strictures and fibrosis. J Dig Dis 2024; 25:476-483. [PMID: 39191433 DOI: 10.1111/1751-2980.13308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 07/21/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024]
Abstract
Crohn's disease (CD) is a chronic inflammatory disorder of the gastrointestinal tract. Intestinal fibrosis or stricture is one of the most prevalent complications in CD with a high recurrence rate. Manual examination of intestinal fibrosis or stricture by physicians may be biased or inefficient. A rapid development of artificial intelligence (AI) technique in recent years facilitates the detection of existing or possible intestinal fibrosis and stricture in CD through various modalities, including endoscopy, imaging examination, and serological biomarkers. We reviewed the articles on AI application in diagnosing intestinal fibrosis and stricture in CD during the past decade and categorized them into three aspects based on the detection methods, and found that AI helps accurate and expedient identification and prediction of intestinal fibrosis and stenosis in CD.
Collapse
Affiliation(s)
- Yi Fei Chen
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Liu Liu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Bin Lyu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Ye Yang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Si Si Zheng
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Xuan Huang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Yi Xu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Yi Hong Fan
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang Province, China
| |
Collapse
|
16
|
Wang Y, Sun D, Zhang J, Kong Y, Morelli JN, Wen D, Wu G, Li X. Multi-sequence MRI-based radiomics: An objective method to diagnose early-stage osteonecrosis of the femoral head. Eur J Radiol 2024; 177:111563. [PMID: 38897051 DOI: 10.1016/j.ejrad.2024.111563] [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: 03/14/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVES This study investigated the use of radiomics for diagnosing early-stage osteonecrosis of the femoral head (ONFH) by extracting features from multiple MRI sequences and constructing predictive models. MATERIALS AND METHODS We conducted a retrospective review, collected MR images of early-stage ONFH (102 from institution A and 20 from institution B) and healthy femoral heads (102 from institution A and 20 from institution B) from two institutions. We extracted radiomics features, handled batch effects using Combat, and normalized features using z-score. We employed the Least absolute shrinkage and selection operator (LASSO) algorithm, along with Max-Relevance and Min-Redundancy (mRMR), to select optimal features for constructing radiomics models based on single, double, and multi-sequence MRI data. We evaluated performance using receiver operating characteristic (ROC) and precision-recall (PR) curves, and compared area under curve of ROC (AUC-ROC) values with the DeLong test. Additionally, we studied the diagnostic performance of the multi-sequence radiomics model and radiologists, compared the diagnostic outcomes of the model and radiologists using the Fisher exact test. RESULTS We studied 122 early-stage ONFH and 122 normal femoral heads. The multi-sequence model exhibited the best diagnostic performance among all models (AUC-ROC, PR-AUC for training set: 0.96, 0.961; validation set: 0.96, 0.97; test set: 0.94, 0.94), and it outperformed three resident radiologists on the external testing group with an accuracy of 87.5 %, sensitivity of 85.00 %, and specificity of 90.00 % (p < 0.01), highlighting the robustness of our findings. CONCLUSIONS Our study underscored the novelty of the multi-sequence radiomics model in diagnosing early-stage ONFH. By leveraging features extracted from multiple imaging sequences, this approach demonstrated high efficacy, indicating its potential to advance early diagnosis for ONFH. These findings provided important guidance for enhancing early diagnosis of ONFH through radiomics methods, offering new avenues and possibilities for clinical practice and patient care.
Collapse
Affiliation(s)
- Yi Wang
- The Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan 430030, Hubei Province, People's Republic of China
| | - Dong Sun
- The Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan 430030, Hubei Province, People's Republic of China
| | - Jing Zhang
- The Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan 430030, Hubei Province, People's Republic of China
| | - Yuefeng Kong
- Radiology Department, Wuhan Fourth Hospital, No. 473 Hanzheng Street, Wuhan 430030, Hubei Province, People's Republic of China
| | - John N Morelli
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Donglin Wen
- The Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan 430030, Hubei Province, People's Republic of China
| | - Gang Wu
- The Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan 430030, Hubei Province, People's Republic of China.
| | - Xiaoming Li
- The Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan 430030, Hubei Province, People's Republic of China.
| |
Collapse
|
17
|
Gao D, Wu YP, Chen TW. Review and prospects of new progress in intelligent imaging research on lymph node metastasis in esophageal carcinoma. META-RADIOLOGY 2024; 2:100081. [DOI: 10.1016/j.metrad.2024.100081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
18
|
Wang R, Zhou R, Sun S, Yang Z, Chen H. Histograms of computed tomography values in differential diagnosis of benign and malignant osteogenic lesions. Acta Radiol 2024; 65:625-631. [PMID: 38213126 DOI: 10.1177/02841851231225418] [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] [Indexed: 01/13/2024]
Abstract
BACKGROUND The use of histogram analysis of computed tomography (CT) values is a potential method for differentiating between benign osteoblastic lesions (BOLs) and malignant osteoblastic lesions (MOLs). PURPOSE To explore the diagnostic efficacy of histogram analysis in accurately distinguishing between BOLs and MOLs based on CT values. MATERIAL AND METHODS A total of 25 BOLs and 25 MOLs, which were confirmed through pathology or imaging follow-up, were included in this study. FireVoxel software was used to process the lesions and obtain various histogram parameters, including mean value, standard deviation, variance, coefficient of variation, skewness, kurtosis, entropy value, and percentiles ranging from 1st to 99th. Statistical tests, such as two independent-sample t-tests and the Mann-Whitney U test with Bonferroni correction, were employed to compare the differences in histogram parameters between BOLs and MOLs. A receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic efficacy of each parameter. RESULTS Significant differences were observed in several histogram parameters between BOLs and MOLs, including the mean value, coefficient of variation, skewness, and various percentiles. Notably, the 25th percentile demonstrated the highest diagnostic efficacy, as indicated by the largest area under the curve in the ROC curve analysis. CONCLUSION Histogram analysis of CT values provides valuable diagnostic information for accurately differentiating between BOLs and MOLs. Among the different parameters, the 25th percentile parameter proves to be the most effective in this discrimination process.
Collapse
Affiliation(s)
- Ruiqing Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao City, PR China
| | - Ruizhi Zhou
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao City, PR China
| | - Shiqing Sun
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao City, PR China
| | - Zhitao Yang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao City, PR China
| | - Haisong Chen
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao City, PR China
| |
Collapse
|
19
|
Oh J, Wu D, Hong B, Lee D, Kang M, Li Q, Kim K. Texture-preserving low dose CT image denoising using Pearson divergence. Phys Med Biol 2024; 69:115021. [PMID: 38688292 DOI: 10.1088/1361-6560/ad45a4] [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: 12/29/2023] [Accepted: 04/30/2024] [Indexed: 05/02/2024]
Abstract
Objective.The mean squared error (MSE), also known asL2loss, has been widely used as a loss function to optimize image denoising models due to its strong performance as a mean estimator of the Gaussian noise model. Recently, various low-dose computed tomography (LDCT) image denoising methods using deep learning combined with the MSE loss have been developed; however, this approach has been observed to suffer from the regression-to-the-mean problem, leading to over-smoothed edges and degradation of texture in the image.Approach.To overcome this issue, we propose a stochastic function in the loss function to improve the texture of the denoised CT images, rather than relying on complicated networks or feature space losses. The proposed loss function includes the MSE loss to learn the mean distribution and the Pearson divergence loss to learn feature textures. Specifically, the Pearson divergence loss is computed in an image space to measure the distance between two intensity measures of denoised low-dose and normal-dose CT images. The evaluation of the proposed model employs a novel approach of multi-metric quantitative analysis utilizing relative texture feature distance.Results.Our experimental results show that the proposed Pearson divergence loss leads to a significant improvement in texture compared to the conventional MSE loss and generative adversarial network (GAN), both qualitatively and quantitatively.Significance.Achieving consistent texture preservation in LDCT is a challenge in conventional GAN-type methods due to adversarial aspects aimed at minimizing noise while preserving texture. By incorporating the Pearson regularizer in the loss function, we can easily achieve a balance between two conflicting properties. Consistent high-quality CT images can significantly help clinicians in diagnoses and supporting researchers in the development of AI-diagnostic models.
Collapse
Affiliation(s)
- Jieun Oh
- Center for Advanced Medical Computing and Analysis (CAMCA), Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States of America
- Chungnam National University College of Medicine, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea
| | - Dufan Wu
- Center for Advanced Medical Computing and Analysis (CAMCA), Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States of America
| | - Boohwi Hong
- Chungnam National University College of Medicine, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea
| | - Dongheon Lee
- Chungnam National University College of Medicine, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea
| | - Minwoong Kang
- Chungnam National University College of Medicine, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea
| | - Quanzheng Li
- Center for Advanced Medical Computing and Analysis (CAMCA), Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States of America
| | - Kyungsang Kim
- Center for Advanced Medical Computing and Analysis (CAMCA), Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States of America
| |
Collapse
|
20
|
Bülbül HM, Burakgazi G, Kesimal U. Preoperative assessment of grade, T stage, and lymph node involvement: machine learning-based CT texture analysis in colon cancer. Jpn J Radiol 2024; 42:300-307. [PMID: 37874525 DOI: 10.1007/s11604-023-01502-2] [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: 08/17/2023] [Accepted: 10/01/2023] [Indexed: 10/25/2023]
Abstract
PURPOSE To investigate whether texture analysis of primary colonic mass in preoperative abdominal computed tomography (CT) scans of patients diagnosed with colon cancer could predict tumor grade, T stage, and lymph node involvement using machine learning (ML) algorithms. MATERIALS AND METHODS This retrospective study included 73 patients diagnosed with colon cancer. Texture features were extracted from contrast-enhanced CT images using LifeX software. First, feature reduction was performed by two radiologists through reproducibility analysis. Using the analysis of variance method, the parameters that best predicted lymph node involvement, grade, and T stage were determined. The predictive performance of these parameters was assessed using Orange software with the k-nearest neighbor (kNN), random forest, gradient boosting, and neural network models, and their area under the curve values were calculated. RESULTS There was excellent reproducibility between the two radiologists in terms of 49 of the 58 texture parameters that were subsequently subject to further analysis. Considering all four ML algorithms, the mean AUC and accuracy ranges were 0.557-0.800 and 47-76%, respectively, for the prediction of lymph node involvement; 0.666-0.846 and 68-77%, respectively, for the prediction of grade; and 0.768-0.962 and 81-88%, respectively, for the prediction of T stage. The best performance was achieved with the random forest model in the prediction of LN involvement, the kNN model for the prediction of grade, and the gradient boosting model for the prediction of T stage. CONCLUSION The results of this study suggest that the texture analysis of preoperative CT scans obtained for staging purposes in colon cancer can predict the presence of advanced-stage tumors, high tumor grade, and lymph node involvement with moderate specificity and sensitivity rates when evaluated using ML models.
Collapse
Affiliation(s)
- Hande Melike Bülbül
- Department of Radiology, Ministry of Health Recep Tayyip Erdoğan University Training and Research Hospital, Rize, Turkey.
| | - Gülen Burakgazi
- Department of Radiology, Ministry of Health Recep Tayyip Erdoğan University Training and Research Hospital, Rize, Turkey
| | - Uğur Kesimal
- Department of Radiology, Ministry of Health Ankara Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
21
|
Dong Z, Guan C, Yang X. Prediction of Fuhrman pathological grade of renal clear cell carcinoma based on CT texture analysis. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2024; 12:28-35. [PMID: 38500865 PMCID: PMC10944366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/10/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVE To study the predictive performance of the imaging model based on the texture analysis of CT plain scan in distinguishing between low (grade I and II) and high (grade III and IV) of Fuhrman pathological grade of renal clear cell carcinoma. METHODS The clinical data of 94 patients with ccRCC who underwent CT scan and were confirmed by biopsy or surgery in TCGA-KIRC public database were retrospectively analyzed. There were 32 cases of low-grade ccRCC and 62 cases of high-grade ccRCC. The patients were randomly divided into training set and verification set according to the proportion of 7:3 by stratified sampling method. The imaging characteristics of ccRCC were calculated in the plain CT images. Lasso regression was used to reduce the dimensionality of the imaging characteristics of the training set, and binary logistic regression was used to construct the prediction model. Bootstrap method was used to verify the training set model and the validation set model, and the area under the receiver operating characteristic (ROC) curve (AUC) was calculated respectively. RESULTS Binary logistic regression showed that only imaging features were independent risk factors for predicting the Furhman classification of ccRCC. The predictive model was y = 1/[1 + exp (-z)], z = 1.274 × imaging risk score + 0.072. The results of bootstrap internal validation showed that the AUC of the training group was 0.961 (95% CI: 0.900-0.913). The Hosmer-Lemeshow goodness of fit test showed that the prediction model had a good calibration in the training group (P = 0.416). The AUC of prediction model in validation group was 0.731 (95% CI: 0.500-1.000). The Hosmer-Lemeshow goodness of fit test results showed that the prediction model had a good calibration in the validation group (P = 0.592). CONCLUSION The model based on CT texture analysis has a good predictive effect in differentiating low-grade and high-grade ccRCC and can provide reference for the treatment and prognosis of patients.
Collapse
Affiliation(s)
- Zhuang Dong
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical UniversityBengbu 233020, Anhui, China
| | - Chao Guan
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical UniversityBengbu 233020, Anhui, China
| | - Xuezhen Yang
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical UniversityBengbu 233020, Anhui, China
- Department of Urology, Qingdao West Coast New District People’s Hospital, Shandong Second Medical UniversityQingdao 266400, Shandong, China
| |
Collapse
|
22
|
Zheng Y, Shi H, Fu S, Wang H, Wang J, Li X, Li Z, Hai B, Zhang J. A computed tomography urography-based machine learning model for predicting preoperative pathological grade of upper urinary tract urothelial carcinoma. Cancer Med 2024; 13:e6901. [PMID: 38174830 PMCID: PMC10807597 DOI: 10.1002/cam4.6901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
OBJECTIVES Development and validation of a computed tomography urography (CTU)-based machine learning (ML) model for prediction of preoperative pathology grade of upper urinary tract urothelial carcinoma (UTUC). METHODS A total of 140 patients with UTUC who underwent CTU examination from January 2017 to August 2023 were retrospectively enrolled. Tumor lesions on the unenhanced, medullary, and excretory periods of CTU were used to extract Features, respectively. Feature selection was screened by the Pearson and Spearman correlation analysis, least absolute shrinkage and selection operator algorithm, random forest (RF), support vector machine (SVM), and eXtreme Gradient Boosting (XGBoost). The logistic regression (LR) was used to screen for independent influencing factors of clinical baseline characteristics. Machine learning models based on different feature datasets were constructed and validated using algorithms such as LR, RF, SVM, and XGBoost. By computing the selected features, a radiomics score was generated, and a diverse feature dataset was constructed. Based on the training set, 16 ML models were created, and their performance was evaluated using the validation set for metrics including sensitivity, specificity, accuracy, area under the receiver operating characteristic curve (AUC), and others. RESULTS The training set consisted of 98 patients (mean age: 64.5 ± 10.5 years; 30 males), whereas the validation set consisted of 42 patients (mean age: 65.3 ± 9.78 years; 17 males). Hydronephrosis was the best independent influence factor (p < 0.05). The RF model had the best performance in predicting high-grade UTUC, with AUC of 0.914 (95% Confidence Interval [95%CI] 0.852-0.977) and 0.903 (95%CI 0.809-0.997) in the training set and validation set, and accuracy of 0.878 and 0.857, respectively. CONCLUSIONS An ML model based on the RF algorithm exhibits excellent predictive performance, offering a non-invasive approach for predicting preoperative high-grade UTUC.
Collapse
Affiliation(s)
- Yanghuang Zheng
- Department of UrologyThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| | - Hongjin Shi
- Department of UrologyThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| | - Shi Fu
- Department of UrologyThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| | - Haifeng Wang
- Department of UrologyThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| | - Jincheng Wang
- Department of UrologyThe First People's Hospital of Luliang CountyLijiangYunnanPeople's Republic of China
| | - Xin Li
- Department of UrologyThe Cancer Hospital of Yunnan ProvinceKunmingYunnanPeople's Republic of China
| | - Zhi Li
- Department of RadiologyThe First People's Hospital of Yunnan ProvinceKunmingYunnanPeople's Republic of China
| | - Bing Hai
- Department of Respiratory MedicineThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| | - Jinsong Zhang
- Department of UrologyThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| |
Collapse
|
23
|
Cellina M, Irmici G, Pepa GD, Ce M, Chiarpenello V, Alì M, Papa S, Carrafiello G. Radiomics and Artificial Intelligence in Renal Lesion Assessment. Crit Rev Oncog 2024; 29:65-75. [PMID: 38505882 DOI: 10.1615/critrevoncog.2023051084] [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: 03/21/2024]
Abstract
Radiomics, the extraction and analysis of quantitative features from medical images, has emerged as a promising field in radiology with the potential to revolutionize the diagnosis and management of renal lesions. This comprehensive review explores the radiomics workflow, including image acquisition, feature extraction, selection, and classification, and highlights its application in differentiating between benign and malignant renal lesions. The integration of radiomics with artificial intelligence (AI) techniques, such as machine learning and deep learning, can help patients' management and allow the planning of the appropriate treatments. AI models have shown remarkable accuracy in predicting tumor aggressiveness, treatment response, and patient outcomes. This review provides insights into the current state of radiomics and AI in renal lesion assessment and outlines future directions for research in this rapidly evolving field.
Collapse
Affiliation(s)
- Michaela Cellina
- Radiology Department, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, Milano, Piazza Principessa Clotilde 3, 20121, Milan, Italy
| | - Giovanni Irmici
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Gianmarco Della Pepa
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Maurizio Ce
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, 20122 Milan, Italy
| | - Vittoria Chiarpenello
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, 20122 Milan, Italy
| | - Marco Alì
- Radiology Unit, CDI, Centro Diagnostico Italiano, 20147 Milan, Italy
| | - Sergio Papa
- Radiology Unit, CDI, Centro Diagnostico Italiano, Via Simone Saint Bon, 20, 20147 Milan, Italy
| | - Gianpaolo Carrafiello
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy; Radiology Department, Fondazione IRCCS Cà Granda, Policlinico di Milano Ospedale Maggiore, Università di Milano, 20122 Milan, Italy
| |
Collapse
|
24
|
Thapa P, Bhatt S, Mishra D, Mehta DS. Effect of fluorescein dye concentration in oral cancer tissue: Statistical and spectroscopic analysis. Photodiagnosis Photodyn Ther 2023; 44:103889. [PMID: 37949386 DOI: 10.1016/j.pdpdt.2023.103889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Oral cancer screening with exogenous agents is highly demanding due to high sensitivity, as the early diagnosis plays a vital role in achieving favorable outcomes for oral squamous cell carcinomas (OSCC) by facilitating prompt detection and comprehensive surgical removal. Optical techniques utilizing the local application of fluorescein dye or fluorescence-guided surgery offer potential for early OSCC detection. The use of fluorescein dye in oral cancer is significantly less, and there is a need to inspect the local application of fluorescein dye in oral cancer patients. Concentration-based investigations of the dye with OSCC patients are essential to ensure accurate fluorescence-guided surgery and screening with fluorescein labeling and to mitigate possible adverse effects. Additionally, analyzing the dye distribution within OSCC tissues can provide insights into their heterogeneity, a critical indicator of malignancy. The present study includes a concentration-based statistical and spectroscopic analysis of fluorescein dye in ex-vivo and in-vivo OSCC patients. In the ex-vivo examination of OSCC tissues, five concentrations (18.66 ± 0.06, 9.51 ± 0.02, 6.38 ± 0.01, 4.80 ± 0.004, and 3.85 ± 0.002 millimolar) are employed for optical analysis. The ex-vivo OSCC tissues are analyzed for multiple statistical parameters at all concentrations, and the results are thoroughly described. Additionally, spectroscopic analysis is conducted on all concentrations for a comprehensive evaluation. Following optical analysis of all five concentrations in the ex-vivo study, two concentrations, 6.38 ± 0.01 and 4.80 ± 0.004 millimolar, are identified as suitable for conducting in-vivo investigations of oral cancer. A detailed spectroscopic and statistical study of OSCC tissues in-vivo has been done using these two concentrations.
Collapse
Affiliation(s)
- Pramila Thapa
- Bio-photonics and Green-photonics Laboratory, Department of Physics, Indian Institute of Technology Delhi, Hauz-Khas, New Delhi 110016, India
| | - Sunil Bhatt
- Bio-photonics and Green-photonics Laboratory, Department of Physics, Indian Institute of Technology Delhi, Hauz-Khas, New Delhi 110016, India
| | - Deepika Mishra
- Department of Oral Pathology and Microbiology, Center for Dental Education & Research, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi 110029, India
| | - Dalip Singh Mehta
- Bio-photonics and Green-photonics Laboratory, Department of Physics, Indian Institute of Technology Delhi, Hauz-Khas, New Delhi 110016, India.
| |
Collapse
|
25
|
Shi L, Sheng M, Wei Z, Liu L, Zhao J. CT-Based Radiomics Predicts the Malignancy of Pulmonary Nodules: A Systematic Review and Meta-Analysis. Acad Radiol 2023; 30:3064-3075. [PMID: 37385850 DOI: 10.1016/j.acra.2023.05.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 07/01/2023]
Abstract
RATIONALE AND OBJECTIVES More pulmonary nodules (PNs) have been detected with the wide application of computed tomography (CT) in lung cancer screening. Radiomics is a noninvasive approach to predict the malignancy of PNs. We aimed to systematically evaluate the methodological quality of the eligible studies regarding CT-based radiomics models in predicting the malignancy of PNs and evaluate the model performance of the available studies. MATERIALS AND METHODS PubMed, Embase, and Web of Science were searched to retrieve relevant studies. The methodological quality of the included studies was assessed using the Radiomics Quality Score (RQS) and Prediction model Risk of Bias Assessment Tool. A meta-analysis was conducted to evaluate the performance of CT-based radiomics model. Meta-regression and subgroup analyses were employed to investigate the source of heterogeneity. RESULTS In total, 49 studies were eligible for qualitative analysis and 27 studies were included in quantitative synthesis. The median RQS of 49 studies was 13 (range -2 to 20). The overall risk of bias was found to be high, and the overall applicability was of low concern in all included studies. The pooled sensitivity, specificity, and diagnostic odds ratio were 0.86 95% confidence interval (CI): 0.79-0.91, 0.84 95% CI: 0.78-0.88, and 31.55 95% CI: 21.31-46.70, respectively. The overall area under the curve was 0.91 95% CI: 0.89-0.94. Meta-regression showed the type of PNs on heterogeneity. CT-based radiomics models performed better in studies including only solid PNs. CONCLUSION CT-based radiomics models exhibited excellent diagnostic performance in predicting the malignancy of PNs. Prospective, large sample size, and well-devised studies are desired to verify the prediction capabilities of CT-based radiomics model.
Collapse
Affiliation(s)
- Lili Shi
- Medical School, Nantong University, Nantong, China (L.S., Z.W.)
| | - Meihong Sheng
- Department of Radiology, The Second Affiliated Hospital of Nantong University and Nantong First People's Hospital, Nantong, China (M.S.)
| | - Zhichao Wei
- Medical School, Nantong University, Nantong, China (L.S., Z.W.)
| | - Lei Liu
- Institutes of Intelligence Medicine, Fudan University, Shanghai, China (L.L.)
| | - Jinli Zhao
- Department of Radiology, Affiliated Hospital of Nantong University, Nantong, China (J.Z.).
| |
Collapse
|
26
|
Khodabakhshi Z, Amini M, Hajianfar G, Oveisi M, Shiri I, Zaidi H. Dual-Centre Harmonised Multimodal Positron Emission Tomography/Computed Tomography Image Radiomic Features and Machine Learning Algorithms for Non-small Cell Lung Cancer Histopathological Subtype Phenotype Decoding. Clin Oncol (R Coll Radiol) 2023; 35:713-725. [PMID: 37599160 DOI: 10.1016/j.clon.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 06/10/2023] [Accepted: 08/05/2023] [Indexed: 08/22/2023]
Abstract
AIMS We aimed to build radiomic models for classifying non-small cell lung cancer (NSCLC) histopathological subtypes through a dual-centre dataset and comprehensively evaluate the effect of ComBat harmonisation on the performance of single- and multimodality radiomic models. MATERIALS AND METHODS A public dataset of NSCLC patients from two independent centres was used. Two image fusion methods, namely guided filtering-based fusion and image fusion based on visual saliency map and weighted least square optimisation, were used. Radiomic features were extracted from each scan, including first-order, texture and moment-invariant features. Subsequently, ComBat harmonisation was applied to the extracted features from computed tomography (CT), positron emission tomography (PET) and fused images to correct the centre effect. For feature selection, least absolute shrinkage and selection operator (Lasso) and recursive feature elimination (RFE) were investigated. For machine learning, logistic regression (LR), support vector machine (SVM) and AdaBoost were evaluated for classifying NSCLC subtypes. Training and evaluation of the models were carried out in a robust framework to offset plausible errors and performance was reported using area under the curve, balanced accuracy, sensitivity and specificity before and after harmonisation. N-way ANOVA was used to assess the effect of different factors on the performance of the models. RESULTS Support vector machine fed with selected features by recursive feature elimination from a harmonised PET feature set achieved the highest performance (area under the curve = 0.82) in classifying NSCLC histopathological subtypes. Although the performance of the models did not significantly improve for CT images after harmonisation, the performance of PET and guided filtering-based fusion feature signatures significantly improved for almost all models. Although the selection of the image modality and feature selection methods was effective on the performance of the model (ANOVA P-values <0.001), machine learning and harmonisation did not change the performance significantly (ANOVA P-values = 0.839 and 0.292, respectively). CONCLUSION This study confirmed the potential of radiomic analysis on PET, CT and hybrid images for histopathological classification of NSCLC subtypes.
Collapse
Affiliation(s)
- Z Khodabakhshi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - M Amini
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - G Hajianfar
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - M Oveisi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran; Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, Kings College London, London, UK; Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - I Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - H Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland; Geneva University Neurocenter, Geneva University, Geneva, Switzerland; Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
27
|
Xia F, Guo F, Liu Z, Zeng J, Ma X, Yu C, Li C. Enhanced CT combined with texture analysis for differential diagnosis of pleomorphic adenoma and adenolymphoma. BMC Med Imaging 2023; 23:169. [PMID: 37891554 PMCID: PMC10612226 DOI: 10.1186/s12880-023-01129-9] [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: 03/13/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
OBJECTIVE This study sought to evaluate the worth of the general characteristics of enhanced CT images and the histogram parameters of each stage in distinguishing pleomorphic adenoma (PA) and adenolymphoma (AL). METHODS The imaging features and histogram parameters of preoperative enhanced CT images in 20 patients with PA and 29 patients with AL were analyzed. Tumor morphology and histogram parameters of PA and AL were compared. Area under the curve (AUC), sensitivity, and subject operational feature specificity (ROC) analysis were used to determine the differential diagnostic effect of single-stage or multi-stage parameter combinations. RESULTS The difference in CT value and net enhancement value of arterial phase (AP) were significant (p < 0.05); Flat sweep phase (FSP), AP mean, percentiles, 10th, 50th, 90th, 99th and arterial period variance and venous phase (VP) kurtosis in the nine histogram parameters of each period (p < 0.05). An analysis of the ROC curve revealed a maximum area beneath the curve (AUC) in the 90th percentile of FSP for a single-parameter differential diagnosis to be 0.870. The diagnostic efficacy of the mean value of FSP + The 90th percentile of AP + Kurtosis of VP was the best in multi-parameter combination diagnosis, with an AUC of 0.925, and the sensitivity and specificity of 0.900 and 0.850, respectively. CONCLUSION The histogram analysis of enhanced CT images is valuable for the differentiation of PA and AL. Moreover, the combination of single-stage parameters or multi-stage parameters can improve the differential diagnosis efficiency.
Collapse
Affiliation(s)
- Feifei Xia
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Shihezi University, Shihezi, 832000, China
| | - Foqing Guo
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Shihezi University, Shihezi, 832000, China
| | - Zhe Liu
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Shihezi University, Shihezi, 832000, China
| | - Jie Zeng
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Shihezi University, Shihezi, 832000, China
| | - Xuehua Ma
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Shihezi University, Shihezi, 832000, China
| | - Chongqing Yu
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Shihezi University, Shihezi, 832000, China
| | - Changxue Li
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Shihezi University, Shihezi, 832000, China.
| |
Collapse
|
28
|
Zhang Y, Ran C, Li W. Central and peripheral pulmonary sclerosing pneumocytomas: multi-phase CT study and comparison with Ki-67. Radiol Oncol 2023; 57:310-316. [PMID: 37665739 PMCID: PMC10476905 DOI: 10.2478/raon-2023-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/21/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND This study aimed to evaluate the multi-phase CT findings of central and peripheral pulmonary sclerosing pneumocytomas (PSPs) and compared them with Ki-67 to reveal their neoplastic nature. PATIENTS AND METHODS Multi-phase CT and clinical data of 33 PSPs (15 central PSPs and 18 peripheral PSPs) were retrospectively analyzed and compared their multi-phase CT features and Ki-67 levels. RESULTS For quantitative indicators, central PSPs were larger than peripheral PSPs (10.39 ± 3.25 cm3 vs. 4.65 ± 2.61 cm3, P = 0.013), and tumor size was negatively correlated with acceleration index (r = -0.845, P < 0.001). The peak enhancement of central PSPs appeared in the delayed phase, with a longer time to peak enhancement (TTP, 100.81 ± 19.01 s), lower acceleration index (0.63 ± 0.17), progressive enhancement, and higher Ki-67 level. The peak enhancement of peripheral PSPs appeared in the venous phase, with the shorter TTP (62.67 ± 20.96 s, P < 0.001), higher acceleration index (0.99 ± 0.25, P < 0.001), enhancement washout, and lower Ki-67 level. For qualitative indicators, the overlying vessel sign (86.67% vs. 44.44%, P = 0.027), prominent pulmonary artery sign (73.33% vs. 27.78%, P = 0.015), and obstructive inflammation/atelectasis (26.67% vs. 0%, P = 0.033) were more common in central PSPs, while peripheral PSPs were more common with halo sign (38.89% vs. 6.67%, P = 0.046). CONCLUSIONS The location of PSP is a possible contributing factor to its diverse imaging-pathological findings. The tumor size, multi-phase enhancement, qualitative signs, and Ki-67 were different between central and peripheral PSPs. Combined tumor size, multi-phase findings, and Ki-67 level are helpful to reveal the nature of the borderline tumor.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Clinical Pharmacy, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Chao Ran
- Department of Radiology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Wei Li
- Department of Medical Imaging, Affiliated Hospital of Yangzhou University, Yangzhou, China
| |
Collapse
|
29
|
Özgül HA, Akin IB, Mutlu U, Balci A. Diagnostic value of machine learning-based computed tomography texture analysis for differentiating multiple myeloma from osteolytic metastatic bone lesions in the peripheral skeleton. Skeletal Radiol 2023; 52:1703-1711. [PMID: 37014470 DOI: 10.1007/s00256-023-04333-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/25/2023] [Accepted: 03/26/2023] [Indexed: 04/05/2023]
Abstract
OBJECTIVES To report the diagnostic performance of machine learning-based CT texture analysis for differentiating multiple myeloma from osteolytic metastatic bone lesions in the peripheral skeleton. METHODS We retrospectively evaluated 172 patients with multiple myeloma (n = 70) and osteolytic metastatic bone lesions (n = 102) in the peripheral skeleton. Two radiologists individually used two-dimensional manual segmentation to extract texture features from non-contrast CT. In total, 762 radiomic features were extracted. Dimension reduction was performed in three stages: inter-observer agreement analysis, collinearity analysis, and feature selection. Data were randomly divided into training (n = 120) and test (n = 52) groups. Eight machine learning algorithms were used for model development. The primary performance metrics were the area under the receiver operating characteristic curve and accuracy. RESULTS In total, 476 of the 762 texture features demonstrated excellent interobserver agreement. The number of features was reduced to 22 after excluding those with strong collinearity. Of these features, six were included in the machine learning algorithms using the wrapper-based classifier-specific technique. When all eight machine learning algorithms were considered for differentiating multiple myeloma from osteolytic metastatic bone lesions in the peripheral skeleton, the area under the receiver operating characteristic curve and accuracy were 0.776-0.932 and 78.8-92.3%, respectively. The k-nearest neighbors model performed the best, with the area under the receiver operating characteristic curve and accuracy values of 0.902 and 92.3%, respectively. CONCLUSION Machine learning-based CT texture analysis is a promising method for discriminating multiple myeloma from osteolytic metastatic bone lesions.
Collapse
Affiliation(s)
- Hakan Abdullah Özgül
- Department of Radiology, Kemalpaşa State Hospital, Kırovası Küme Street, Kemalpaşa, 35730, Izmir, Turkey.
| | - Işıl Başara Akin
- Department of Radiology, Dokuz Eylul University, Faculty of Medicine, Izmir, Turkey
| | - Uygar Mutlu
- Department of Radiology, Yozgat State Hospital, Yozgat, Turkey
| | - Ali Balci
- Department of Radiology, Dokuz Eylul University, Faculty of Medicine, Izmir, Turkey
| |
Collapse
|
30
|
Lee K, Goh J, Jang J, Hwang J, Kwak J, Kim J, Eom K. Feasibility study of computed tomography texture analysis for evaluation of canine primary adrenal gland tumors. Front Vet Sci 2023; 10:1126165. [PMID: 37711438 PMCID: PMC10499047 DOI: 10.3389/fvets.2023.1126165] [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: 12/17/2022] [Accepted: 08/01/2023] [Indexed: 09/16/2023] Open
Abstract
Objective This study aimed to investigate the feasibility of computed tomography (CT) texture analysis for distinguishing canine adrenal gland tumors and its usefulness in clinical decision-making. Materials and methods The medical records of 25 dogs with primary adrenal masses who underwent contrast CT and a histopathological examination were retrospectively reviewed, of which 12 had adenomas (AAs), 7 had adenocarcinomas (ACCs), and 6 had pheochromocytomas (PHEOs). Conventional CT evaluation of each adrenal gland tumor included the mean, maximum, and minimum attenuation values in Hounsfield units (HU), heterogeneity of the tumor parenchyma, and contrast enhancement (type, pattern, and degree), respectively, in each phase. In CT texture analysis, precontrast and delayed-phase images of 18 adrenal gland tumors, which could be applied for ComBat harmonization were used, and 93 radiomic features (18 first-order and 75 second-order statistics) were extracted. Then, ComBat harmonization was applied to compensate for the batch effect created by the different CT protocols. The area under the receiver operating characteristic curve (AUC) for each significant feature was used to evaluate the diagnostic performance of CT texture analysis. Results Among the conventional features, PHEO showed significantly higher mean and maximum precontrast HU values than ACC (p < 0.05). Eight second-order features on the precontrast images showed significant differences between the adrenal gland tumors (p < 0.05). However, none of them were significantly different between AA and PHEO, or between precontrast images and delayed-phase images. This result indicates that ACC exhibited more heterogeneous and complex textures and more variable intensities with lower gray-level values than AA and PHEO. The correlation, maximal correlation coefficient, and gray level non-uniformity normalized were significantly different between AA and ACC, and between ACC and PHEO. These features showed high AUCs in discriminating ACC and PHEO, which were comparable or higher than the precontrast mean and maximum HU (AUC = 0.865 and 0.860, respectively). Conclusion Canine primary adrenal gland tumor differentiation can be achieved with CT texture analysis on precontrast images and may have a potential role in clinical decision-making. Further prospective studies with larger populations and cross-validation are warranted.
Collapse
Affiliation(s)
- Kyungsoo Lee
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jinhyong Goh
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jaeyoung Jang
- Jang Jae Young Veterinary Surgery Center, Seong-nam, Gyunggi-do, Republic of Korea
| | | | - Jungmin Kwak
- Saram and Animal Medical Center, Yongin-si, Gyunggi-do, Republic of Korea
| | - Jaehwan Kim
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Kidong Eom
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
31
|
Varghese BA, Fields BKK, Hwang DH, Duddalwar VA, Matcuk GR, Cen SY. Spatial assessments in texture analysis: what the radiologist needs to know. FRONTIERS IN RADIOLOGY 2023; 3:1240544. [PMID: 37693924 PMCID: PMC10484588 DOI: 10.3389/fradi.2023.1240544] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023]
Abstract
To date, studies investigating radiomics-based predictive models have tended to err on the side of data-driven or exploratory analysis of many thousands of extracted features. In particular, spatial assessments of texture have proven to be especially adept at assessing for features of intratumoral heterogeneity in oncologic imaging, which likewise may correspond with tumor biology and behavior. These spatial assessments can be generally classified as spatial filters, which detect areas of rapid change within the grayscale in order to enhance edges and/or textures within an image, or neighborhood-based methods, which quantify gray-level differences of neighboring pixels/voxels within a set distance. Given the high dimensionality of radiomics datasets, data dimensionality reduction methods have been proposed in an attempt to optimize model performance in machine learning studies; however, it should be noted that these approaches should only be applied to training data in order to avoid information leakage and model overfitting. While area under the curve of the receiver operating characteristic is perhaps the most commonly reported assessment of model performance, it is prone to overestimation when output classifications are unbalanced. In such cases, confusion matrices may be additionally reported, whereby diagnostic cut points for model predicted probability may hold more clinical significance to clinical colleagues with respect to related forms of diagnostic testing.
Collapse
Affiliation(s)
- Bino A. Varghese
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Brandon K. K. Fields
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Darryl H. Hwang
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Vinay A. Duddalwar
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - George R. Matcuk
- Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Steven Y. Cen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
32
|
Durot C, Durot E, Mulé S, Morland D, Godard F, Quinquenel A, Delmer A, Soyer P, Hoeffel C. Pretreatment CT Texture Parameters as Predictive Biomarkers of Progression-Free Survival in Follicular Lymphoma Treated with Immunochemotherapy and Rituximab Maintenance. Diagnostics (Basel) 2023; 13:2237. [PMID: 37443630 DOI: 10.3390/diagnostics13132237] [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: 05/03/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The purpose of this study was to determine whether texture analysis features present on pretreatment unenhanced computed tomography (CT) images, derived from 18F-fluorodeoxyglucose positron emission/computed tomography (18-FDG PET/CT), can predict progression-free survival (PFS), progression-free survival at 24 months (PFS 24), time to next treatment (TTNT), and overall survival in patients with high-tumor-burden follicular lymphoma treated with immunochemotherapy and rituximab maintenance. Seventy-two patients with follicular lymphoma were retrospectively included. Texture analysis was performed on unenhanced CT images extracted from 18-FDG PET/CT examinations that were obtained within one month before treatment. Skewness at a fine texture scale (SSF = 2) was an independent predictor of PFS (hazard ratio = 3.72 (95% CI: 1.15, 12.11), p = 0.028), PFS 24 (hazard ratio = 13.38; 95% CI: 1.29, 138.13; p = 0.029), and TTNT (hazard ratio = 5.11; 95% CI: 1.18, 22.13; p = 0.029). Skewness values above -0.015 at SSF = 2 were significantly associated with lower PFS, PFS 24, and TTNT. Kurtosis without filtration was an independent predictor of PFS (SSF = 0; HR = 1.22 (95% CI: 1.04, 1.44), p = 0.013), and TTNT (SSF = 0; hazard ratio = 1.23; 95% CI: 1.04, 1.46; p = 0.013). This study shows that pretreatment unenhanced CT texture analysis-derived tumor skewness and kurtosis may be used as predictive biomarkers of PFS and TTNT in patients with high-tumor-burden follicular lymphoma treated with immunochemotherapy and rituximab maintenance.
Collapse
Affiliation(s)
- Carole Durot
- Department of Radiology, Reims University Hospital, 45 Rue Cognacq-Jay, 51092 Reims, France
| | - Eric Durot
- Department of Hematology, Reims University Hospital, 45 Rue Cognacq-Jay, 51092 Reims, France
| | - Sébastien Mulé
- Department of Radiology, Henri Mondor University Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
- Faculté de Médecine, Université Paris-Est Créteil, 61 Avenue du Général de Gaulle, 94000 Créteil, France
| | - David Morland
- Department of Nuclear Medicine, Godinot Institute, 1 Rue du Général Koenig, 51100 Reims, France
- CReSTIC, EA 3804, University of Reims Champagne-Ardenne, UFR Moulin de la Housse, 51867 Reims, France
| | - François Godard
- Department of Radiology, Henri Mondor University Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Anne Quinquenel
- Department of Hematology, Reims University Hospital, 45 Rue Cognacq-Jay, 51092 Reims, France
| | - Alain Delmer
- Department of Hematology, Reims University Hospital, 45 Rue Cognacq-Jay, 51092 Reims, France
| | - Philippe Soyer
- Department of Radiology, Hôpital Cochin, AP-HP, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France
- Faculté de Médecine, Université Paris Cité, 75006 Paris, France
| | - Christine Hoeffel
- Department of Radiology, Reims University Hospital, 45 Rue Cognacq-Jay, 51092 Reims, France
- CReSTIC, EA 3804, University of Reims Champagne-Ardenne, UFR Moulin de la Housse, 51867 Reims, France
| |
Collapse
|
33
|
Muntean DD, Dudea SM, Băciuț M, Dinu C, Stoia S, Solomon C, Csaba C, Rusu GM, Lenghel LM. The Role of an MRI-Based Radiomic Signature in Predicting Malignancy of Parotid Gland Tumors. Cancers (Basel) 2023; 15:3319. [PMID: 37444429 PMCID: PMC10340186 DOI: 10.3390/cancers15133319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/11/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of this study was to assess the ability of MRI radiomic features to differentiate between benign parotid gland tumors (BPGT) and malignant parotid gland tumors (MPGT). This retrospective study included 93 patients who underwent MRI examinations of the head and neck region (78 patients presenting unique PGT, while 15 patients presented double PGT). A total of 108 PGT with histological confirmation were eligible for the radiomic analysis and were assigned to a training group (n = 83; 58 BPGT; 25 MPGT) and a testing group (n = 25; 16 BPGT; 9 MPGT). The radiomic features were extracted from 3D segmentations of the PGT on the T2-weighted and fat-saturated, contrast-enhanced T1-weighted images. Following feature reduction techniques, including LASSO regression analysis, a radiomic signature (RS) was built with five radiomic features. The RS presented a good diagnostic performance in differentiating between PGT, achieving an area under the curve (AUC) of 0.852 (p < 0.001) in the training set and 0.786 (p = 0.017) in the testing set. In both datasets, the RS proved to have lower values in the BPGT group as compared to MPGT group (p < 0.001 and p = 0.023, respectively). The multivariate analysis revealed that RS was independently associated with PGT malignancy, together with the ill-defined margin pattern (p = 0.031, p = 0.001, respectively). The complex model, using clinical data, MRI features and the RS, presented a higher diagnostic performance (AUC of 0.976) in comparison to the RS alone. MRI-based radiomic features could be considered potential additional imaging biomarkers able to discriminate between benign and malignant parotid gland tumors.
Collapse
Affiliation(s)
- Delia Doris Muntean
- Department of Radiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.D.M.); (S.M.D.); (C.C.); (G.M.R.); (L.M.L.)
| | - Sorin Marian Dudea
- Department of Radiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.D.M.); (S.M.D.); (C.C.); (G.M.R.); (L.M.L.)
| | - Mihaela Băciuț
- Department of Maxillofacial Surgery and Implantology, Faculty of Dentistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (M.B.); (C.D.); (S.S.)
| | - Cristian Dinu
- Department of Maxillofacial Surgery and Implantology, Faculty of Dentistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (M.B.); (C.D.); (S.S.)
| | - Sebastian Stoia
- Department of Maxillofacial Surgery and Implantology, Faculty of Dentistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (M.B.); (C.D.); (S.S.)
| | - Carolina Solomon
- Department of Radiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.D.M.); (S.M.D.); (C.C.); (G.M.R.); (L.M.L.)
| | - Csutak Csaba
- Department of Radiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.D.M.); (S.M.D.); (C.C.); (G.M.R.); (L.M.L.)
| | - Georgeta Mihaela Rusu
- Department of Radiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.D.M.); (S.M.D.); (C.C.); (G.M.R.); (L.M.L.)
| | - Lavinia Manuela Lenghel
- Department of Radiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.D.M.); (S.M.D.); (C.C.); (G.M.R.); (L.M.L.)
| |
Collapse
|
34
|
Gao X, Cui J, Wang L, Wang Q, Ma T, Yang J, Ye Z. The value of machine learning based radiomics model in preoperative detection of perineural invasion in gastric cancer: a two-center study. Front Oncol 2023; 13:1205163. [PMID: 37388227 PMCID: PMC10303108 DOI: 10.3389/fonc.2023.1205163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/30/2023] [Indexed: 07/01/2023] Open
Abstract
Purpose To establish and validate a machine learning based radiomics model for detection of perineural invasion (PNI) in gastric cancer (GC). Methods This retrospective study included a total of 955 patients with GC selected from two centers; they were separated into training (n=603), internal testing (n=259), and external testing (n=93) sets. Radiomic features were derived from three phases of contrast-enhanced computed tomography (CECT) scan images. Seven machine learning (ML) algorithms including least absolute shrinkage and selection operator (LASSO), naïve Bayes (NB), k-nearest neighbor (KNN), decision tree (DT), logistic regression (LR), random forest (RF), eXtreme gradient boosting (XGBoost) and support vector machine (SVM) were trained for development of optimal radiomics signature. A combined model was constructed by aggregating the radiomic signatures and important clinicopathological characteristics. The predictive ability of the radiomic model was then assessed with receiver operating characteristic (ROC) and calibration curve analyses in all three sets. Results The PNI rates for the training, internal testing, and external testing sets were 22.1, 22.8, and 36.6%, respectively. LASSO algorithm was selected for signature establishment. The radiomics signature, consisting of 8 robust features, revealed good discrimination accuracy for the PNI in all three sets (training set: AUC = 0.86; internal testing set: AUC = 0.82; external testing set: AUC = 0.78). The risk of PNI was significantly associated with higher radiomics scores. A combined model that integrated radiomics and T stage demonstrated enhanced accuracy and excellent calibration in all three sets (training set: AUC = 0.89; internal testing set: AUC = 0.84; external testing set: AUC = 0.82). Conclusion The suggested radiomics model exhibited satisfactory prediction performance for the PNI in GC.
Collapse
Affiliation(s)
- Xujie Gao
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Radiology, National Clinical Research Center for Cancer, Tianjin, China
- Department of Radiology, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jingli Cui
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Radiology, National Clinical Research Center for Cancer, Tianjin, China
- Department of Radiology, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of General Surgery, Weifang People’s Hospital, Weifang, Shandong, China
| | - Lingwei Wang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Radiology, National Clinical Research Center for Cancer, Tianjin, China
- Department of Radiology, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Qiuyan Wang
- Department of Radiology, Weifang People’s Hospital, Weifang, Shandong, China
| | - Tingting Ma
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Radiology, National Clinical Research Center for Cancer, Tianjin, China
- Department of Radiology, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Radiology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Jilong Yang
- Department of Radiology, National Clinical Research Center for Cancer, Tianjin, China
- Department of Radiology, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Radiology, National Clinical Research Center for Cancer, Tianjin, China
- Department of Radiology, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
35
|
Kiryu S, Akai H, Yasaka K, Tajima T, Kunimatsu A, Yoshioka N, Akahane M, Abe O, Ohtomo K. Clinical Impact of Deep Learning Reconstruction in MRI. Radiographics 2023; 43:e220133. [PMID: 37200221 DOI: 10.1148/rg.220133] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Deep learning has been recognized as a paradigm-shifting tool in radiology. Deep learning reconstruction (DLR) has recently emerged as a technology used in the image reconstruction process of MRI, which is an essential procedure in generating MR images. Denoising, which is the first DLR application to be realized in commercial MRI scanners, improves signal-to-noise ratio. When applied to lower magnetic field-strength scanners, the signal-to-noise ratio can be increased without extending the imaging time, and image quality is comparable to that of higher-field-strength scanners. Shorter imaging times decrease patient discomfort and reduce MRI scanner running costs. The incorporation of DLR into accelerated acquisition imaging techniques, such as parallel imaging or compressed sensing, shortens the reconstruction time. DLR is based on supervised learning using convolutional layers and is divided into the following three categories: image domain, k-space learning, and direct mapping types. Various studies have reported other derivatives of DLR, and several have shown the feasibility of DLR in clinical practice. Although DLR efficiently reduces Gaussian noise from MR images, denoising makes image artifacts more prominent, and a solution to this problem is desired. Depending on the training of the convolutional neural network, DLR may change the imaging features of lesions and obscure small lesions. Therefore, radiologists may need to adopt the habit of questioning whether any information has been lost on images that appear clean. ©RSNA, 2023 Quiz questions for this article are available in the supplemental material.
Collapse
Affiliation(s)
- Shigeru Kiryu
- From the Department of Radiology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita 286-0124, Japan (S.K., H.A., K.Y., T.T., A.K., N.Y., M.A.); Department of Radiology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (H.A.); Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (K.Y., O.A.); Department of Radiology, International University of Health and Welfare Mita Hospital, Tokyo, Japan (T.T., A.K.); and International University of Health and Welfare, Otawara, Japan (K.O.)
| | - Hiroyuki Akai
- From the Department of Radiology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita 286-0124, Japan (S.K., H.A., K.Y., T.T., A.K., N.Y., M.A.); Department of Radiology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (H.A.); Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (K.Y., O.A.); Department of Radiology, International University of Health and Welfare Mita Hospital, Tokyo, Japan (T.T., A.K.); and International University of Health and Welfare, Otawara, Japan (K.O.)
| | - Koichiro Yasaka
- From the Department of Radiology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita 286-0124, Japan (S.K., H.A., K.Y., T.T., A.K., N.Y., M.A.); Department of Radiology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (H.A.); Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (K.Y., O.A.); Department of Radiology, International University of Health and Welfare Mita Hospital, Tokyo, Japan (T.T., A.K.); and International University of Health and Welfare, Otawara, Japan (K.O.)
| | - Taku Tajima
- From the Department of Radiology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita 286-0124, Japan (S.K., H.A., K.Y., T.T., A.K., N.Y., M.A.); Department of Radiology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (H.A.); Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (K.Y., O.A.); Department of Radiology, International University of Health and Welfare Mita Hospital, Tokyo, Japan (T.T., A.K.); and International University of Health and Welfare, Otawara, Japan (K.O.)
| | - Akira Kunimatsu
- From the Department of Radiology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita 286-0124, Japan (S.K., H.A., K.Y., T.T., A.K., N.Y., M.A.); Department of Radiology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (H.A.); Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (K.Y., O.A.); Department of Radiology, International University of Health and Welfare Mita Hospital, Tokyo, Japan (T.T., A.K.); and International University of Health and Welfare, Otawara, Japan (K.O.)
| | - Naoki Yoshioka
- From the Department of Radiology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita 286-0124, Japan (S.K., H.A., K.Y., T.T., A.K., N.Y., M.A.); Department of Radiology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (H.A.); Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (K.Y., O.A.); Department of Radiology, International University of Health and Welfare Mita Hospital, Tokyo, Japan (T.T., A.K.); and International University of Health and Welfare, Otawara, Japan (K.O.)
| | - Masaaki Akahane
- From the Department of Radiology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita 286-0124, Japan (S.K., H.A., K.Y., T.T., A.K., N.Y., M.A.); Department of Radiology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (H.A.); Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (K.Y., O.A.); Department of Radiology, International University of Health and Welfare Mita Hospital, Tokyo, Japan (T.T., A.K.); and International University of Health and Welfare, Otawara, Japan (K.O.)
| | - Osamu Abe
- From the Department of Radiology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita 286-0124, Japan (S.K., H.A., K.Y., T.T., A.K., N.Y., M.A.); Department of Radiology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (H.A.); Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (K.Y., O.A.); Department of Radiology, International University of Health and Welfare Mita Hospital, Tokyo, Japan (T.T., A.K.); and International University of Health and Welfare, Otawara, Japan (K.O.)
| | - Kuni Ohtomo
- From the Department of Radiology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita 286-0124, Japan (S.K., H.A., K.Y., T.T., A.K., N.Y., M.A.); Department of Radiology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan (H.A.); Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (K.Y., O.A.); Department of Radiology, International University of Health and Welfare Mita Hospital, Tokyo, Japan (T.T., A.K.); and International University of Health and Welfare, Otawara, Japan (K.O.)
| |
Collapse
|
36
|
Zheng T, Pan J, Du D, Liang X, Yi H, Du J, Wu S, Liu L, Shi G. Preoperative assessment of high-grade endometrial cancer using a radiomic signature and clinical indicators. Future Oncol 2023; 19:587-601. [PMID: 37097730 DOI: 10.2217/fon-2022-0631] [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] [Indexed: 04/26/2023] Open
Abstract
Aim: To develop and validate a radiomics-based combined model (ModelRC) to predict the pathological grade of endometrial cancer. Methods: A total of 403 endometrial cancer patients from two independent centers were enrolled as training, internal validation and external validation sets. Radiomic features were extracted from T2-weighted images, apparent diffusion coefficient map and contrast-enhanced 3D volumetric interpolated breath-hold examination images. Results: Compared with the clinical model and radiomics model, ModelRC showed superior performance; the areas under the receiver operating characteristic curves were 0.920 (95% CI: 0.864-0.962), 0.882 (95% CI: 0.779-0.955) and 0.881 (95% CI: 0.815-0.939) for the training, internal validation and external validation sets, respectively. Conclusion: ModelRC, which incorporated clinical and radiomic features, exhibited excellent performance in the prediction of high-grade endometrial cancer.
Collapse
Affiliation(s)
- Tao Zheng
- Department of Radiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, PR China
| | - Jiangyang Pan
- Department of Radiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, PR China
| | - Dan Du
- Department of Medical Imaging Center, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, PR China
| | - Xin Liang
- Department of Medical Imaging Center, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, PR China
| | - Huiling Yi
- Department of Medical Imaging Center, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, PR China
| | - Juan Du
- Department of Medical Imaging Center, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, PR China
| | - Shuo Wu
- Department of Medical Imaging Center, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, PR China
| | - Lanxiang Liu
- Department of Medical Imaging Center, The First Hospital of Qinhuangdao, Qinhuangdao, 066000, PR China
| | - Gaofeng Shi
- Department of Radiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, PR China
| |
Collapse
|
37
|
Rengo M, Onori A, Caruso D, Bellini D, Carbonetti F, De Santis D, Vicini S, Zerunian M, Iannicelli E, Carbone I, Laghi A. Development and Validation of Artificial-Intelligence-Based Radiomics Model Using Computed Tomography Features for Preoperative Risk Stratification of Gastrointestinal Stromal Tumors. J Pers Med 2023; 13:jpm13050717. [PMID: 37240887 DOI: 10.3390/jpm13050717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND preoperative risk assessment of gastrointestinal stromal tumors (GISTS) is required for optimal and personalized treatment planning. Radiomics features are promising tools to predict risk assessment. The purpose of this study is to develop and validate an artificial intelligence classification algorithm, based on CT features, to define GIST's prognosis as determined by the Miettinen classification. METHODS patients with histological diagnosis of GIST and CT studies were retrospectively enrolled. Eight morphologic and 30 texture CT features were extracted from each tumor and combined to obtain three models (morphologic, texture and combined). Data were analyzed using a machine learning classification (WEKA). For each classification process, sensitivity, specificity, accuracy and area under the curve were evaluated. Inter- and intra-reader agreement were also calculated. RESULTS 52 patients were evaluated. In the validation population, highest performances were obtained by the combined model (SE 85.7%, SP 90.9%, ACC 88.8%, and AUC 0.954) followed by the morphologic (SE 66.6%, SP 81.8%, ACC 76.4%, and AUC 0.742) and texture (SE 50%, SP 72.7%, ACC 64.7%, and AUC 0.613) models. Reproducibility was high of all manual evaluations. CONCLUSIONS the AI-based radiomics model using a CT feature demonstrates good predictive performance for preoperative risk stratification of GISTs.
Collapse
Affiliation(s)
- Marco Rengo
- Department of Medical-Surgical Sciences and Biotechnologies, Academic Diagnostic Imaging Division, I.C.O.T. Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | - Alessandro Onori
- Department of Radiological, Oncological and Pathological Sciences, Academic Diagnostic Imaging Division, I.C.O.T. Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | - Damiano Caruso
- Department of Surgical and Medical Sciences and Translational Medicine, Radiology Unit, Sant'Andrea University Hospital, University of Rome Sapienza, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Davide Bellini
- Department of Medical-Surgical Sciences and Biotechnologies, Academic Diagnostic Imaging Division, I.C.O.T. Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | - Francesco Carbonetti
- Radiology Unit, Sant'Eugenio Hospital, Piazzale dell'Umanesimo 10, 00144 Rome, Italy
| | - Domenico De Santis
- Department of Surgical and Medical Sciences and Translational Medicine, Radiology Unit, Sant'Andrea University Hospital, University of Rome Sapienza, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Simone Vicini
- Department of Medical-Surgical Sciences and Biotechnologies, Academic Diagnostic Imaging Division, I.C.O.T. Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | - Marta Zerunian
- Department of Surgical and Medical Sciences and Translational Medicine, Radiology Unit, Sant'Andrea University Hospital, University of Rome Sapienza, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Elsa Iannicelli
- Department of Surgical and Medical Sciences and Translational Medicine, Radiology Unit, Sant'Andrea University Hospital, University of Rome Sapienza, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Iacopo Carbone
- Department of Radiological, Oncological and Pathological Sciences, Academic Diagnostic Imaging Division, I.C.O.T. Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | - Andrea Laghi
- Department of Surgical and Medical Sciences and Translational Medicine, Radiology Unit, Sant'Andrea University Hospital, University of Rome Sapienza, Via di Grottarossa 1035, 00189 Rome, Italy
| |
Collapse
|
38
|
Xue G, Liu H, Cai X, Zhang Z, Zhang S, Liu L, Hu B, Wang G. Impact of deep learning image reconstruction algorithms on CT radiomic features in patients with liver tumors. Front Oncol 2023; 13:1167745. [PMID: 37091167 PMCID: PMC10113560 DOI: 10.3389/fonc.2023.1167745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
ObjectiveTo evaluate the impact of deep learning image reconstruction (DLIR) and adaptive statistical iterative reconstruction-Veo (ASIR-V) on abdominal CT radiomic features acquired in portal venous phase in liver tumor patients.MethodsSixty patients with liver tumors who underwent contrast-enhanced abdominal CT were retrospectively enrolled. Six groups including filtered back projection (FBP), ASIR-V (30%, 70%) and DLIR at low (DLIR-L), medium (DLIR-M and high (DLIR-H), were reconstructed using portal venous phase data. CT-based radiomic features (first-order, texture and wavelet features) were extracted from 2D and 3D liver tumors, peritumor and liver parenchyma. All features were analyzed for comparison. P < 0.05 indicated statistically different. The consistency of 3D lesion feature extraction was assessed by calculating intraclass correlation coefficient (ICC).ResultsDifferent reconstruction algorithms influenced most radiomic features. The percentages of first-order, texture and wavelet features without statistical difference among 2D and 3D lesions, peritumor and liver parenchyma for all six groups were 27.78% (5/18), 5.33% (4/75) and 5.56% (1/18), respectively (all p > 0.05), and they decreased while the level of reconstruction strengthened for both ASIR-V and DLIR. Compared with FBP, the features of ASIR-V30% and 70% without statistical difference decreased from 71.31% to 23.95%, and DLIR-L, DLIR-M, and DLIR-H decreased from 31.65% to 27.11% and 23.73%. Among texture features, unaffected features of peritumor were larger than those of lesions and liver parenchyma, and unaffected 3D lesions features were larger than those of 2D lesions. The consistency of 3D lesion first-order features was excellent, with intra- and inter-observer ICCs ranging from 0.891 to 0.999 and 0.880 to 0.998.ConclusionsBoth ASIR-V and DLIR algorithms with different strengths influenced the radiomic features of abdominal CT images in portal venous phase, and the influences aggravated as reconstruction strength increased.
Collapse
Affiliation(s)
- Gongbo Xue
- Department of Radiology, Qingdao Municipal Hospital, Qingdao, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Hongyan Liu
- Department of Radiology, Qingdao Municipal Hospital, Qingdao, China
| | - Xiaoyi Cai
- Department of Radiology, Qingdao Municipal Hospital, Qingdao, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Zhen Zhang
- Department of Radiology, Qingdao Municipal Hospital, Qingdao, China
| | - Shuai Zhang
- CT Imaging Research Center, GE Healthcare China, Shanghai, China
| | - Ling Liu
- CT Imaging Research Center, GE Healthcare China, Shanghai, China
| | - Bin Hu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Guohua Wang, ; Bin Hu,
| | - Guohua Wang
- Department of Radiology, Qingdao Municipal Hospital, Qingdao, China
- *Correspondence: Guohua Wang, ; Bin Hu,
| |
Collapse
|
39
|
Truong LUF, Kleiber JC, Durot C, Brenet E, Barbe C, Hoeffel C, Bazin A, Labrousse M, Dubernard X. The study of predictive factors for the evolution of vestibular schwannomas. Eur Arch Otorhinolaryngol 2023; 280:1661-1670. [PMID: 36114332 DOI: 10.1007/s00405-022-07651-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE The primary objective was to determine whether the analysis of textural heterogeneity of vestibular schwannomas on MRI at diagnosis was predictive of their radiological evolutivity. The secondary objective was to determine whether some clinical or radiological factors could also be predictive of growth. METHODS We conducted a pilot, observational and retrospective study of patients with a vestibular schwannoma, initially monitored, between April 2001 and November 2019 within the Oto-Neurosurgical Institute of Champagne Ardenne, Texture analysis was performed on gadolinium injected T1 and CISS T2 MRI sequences and six parameters were extracted: mean greyscale intensity, standard deviation of the greyscale histogram distribution, entropy, mean positive pixels, skewness and kurtosis, which were analysed by the Lasso method, using statistically penalised Cox models. Extrameatal location, tumour necrosis, perceived hearing loss < 2 years with objectified tone audiometry asymmetry, tinnitus at diagnosis, were investigated by the Log-Rank test to obtain univariate survival analyses. RESULTS 78 patients were included and divided into 2 groups: group A comprising 39 "stable patients", and B comprising the remaining 39 "progressive patients". Independent analysis of the texture factors did not predict the growth potential of vestibular schwannomas. Among the clinical or radiological signs of interest, hearing loss < 2 years was identified as a prognostic factor for tumour progression with a significant trend (p = 0.05). CONCLUSIONS This study did not identify an association between texture analysis and vestibular schwannomas growth. Decreased hearing in the 2 years prior to diagnosis appears to predict potential radiological progression.
Collapse
Affiliation(s)
- Le-Uyen France Truong
- Department of Oto-Rhino-Laryngology and Head and neck surgery of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
| | - Jean Charles Kleiber
- Department of Neurosurgery of the CHU of Reims, Hôpital Maison Blanche, 45 rue Cognacq-Jay, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Carole Durot
- Department of Radiology of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Esteban Brenet
- Department of Oto-Rhino-Laryngology and Head and neck surgery of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Coralie Barbe
- Research and Public Health Unit of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Christine Hoeffel
- Department of Radiology of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Arnaud Bazin
- Department of Neurosurgery of the CHU of Reims, Hôpital Maison Blanche, 45 rue Cognacq-Jay, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Marc Labrousse
- Department of Oto-Rhino-Laryngology and Head and neck surgery of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Xavier Dubernard
- Department of Oto-Rhino-Laryngology and Head and neck surgery of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France.
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France.
- Service d'ORL et Chirurgie cervico-faciale, CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France.
| |
Collapse
|
40
|
Ricardo ALF, da Silva GA, Ogawa CM, Nussi AD, De Rosa CS, Martins JS, de Castro Lopes SLP, Appenzeller S, Braz-Silva PH, Costa ALF. Magnetic resonance imaging texture analysis for quantitative evaluation of the mandibular condyle in juvenile idiopathic arthritis. Oral Radiol 2023; 39:329-340. [PMID: 35948783 DOI: 10.1007/s11282-022-00641-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/13/2022] [Indexed: 10/15/2022]
Abstract
OBJECTIVES Juvenile idiopathic arthritis (JIA) is a chronic inflammatory disease that affects the joints and other organs, including the development of the former in a growing child. This study aimed to evaluate the feasibility of texture analysis (TA) based on magnetic resonance imaging (MRI) to provide biomarkers that serve to identify patients likely to progress to temporomandibular joint damage by associating JIA with age, gender and disease onset age. METHODS The radiological database was retrospectively reviewed. A total of 45 patients were first divided into control group (23) and JIA group (22). TA was performed using grey-level co-occurrence matrix (GLCM) parameters, in which 11 textural parameters were calculated using MaZda software. These 11 parameters were ranked based on the p value obtained with ANOVA and then correlated with age, gender and disease onset age. RESULTS Significant differences in texture parameters of condyle were demonstrated between JIA group and control group (p < 0.05). There was a progressive loss of uniformity in the grayscale pixels of MRI with an increasing age in JIA group. CONCLUSIONS MRI TA of the condyle can make it possible to detect the alterations in bone marrow of patients with JIA and promising tool which may help the image analysis.
Collapse
Affiliation(s)
- Ana Lúcia Franco Ricardo
- Postgraduate Program in Dentistry, Cruzeiro Do Sul University (UNICSUL), São Paulo, 01506-000, Brazil
| | - Gabriel Araújo da Silva
- Division of Oral Radiology, Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas (UNICAMP), Campinas, Brazil
| | - Celso Massahiro Ogawa
- Postgraduate Program in Dentistry, Cruzeiro Do Sul University (UNICSUL), São Paulo, 01506-000, Brazil
| | - Amanda D Nussi
- Postgraduate Program in Dentistry, Cruzeiro Do Sul University (UNICSUL), São Paulo, 01506-000, Brazil
| | | | - Jaqueline Serra Martins
- Rheumatology Department, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Sérgio Lúcio Pereira de Castro Lopes
- Department of Diagnosis and Surgery, São José Dos Campos School of Dentistry, São Paulo State University (UNESP), São José dos Campos, SP, Brazil
| | - Simone Appenzeller
- Rheumatology Department, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Andre Luiz Ferreira Costa
- Postgraduate Program in Dentistry, Cruzeiro Do Sul University (UNICSUL), São Paulo, 01506-000, Brazil.
| |
Collapse
|
41
|
Guo W, Liu J, Wang X, Yuan H. Predicting the Risk of Thymic Tumors Using Texture Analysis of Contrast-Enhanced Chest Computed Tomography. J Comput Assist Tomogr 2023:00004728-990000000-00164. [PMID: 36944121 DOI: 10.1097/rct.0000000000001467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
OBJECTIVE This study aimed to explore the value of contrast-enhanced computed tomography texture features for predicting the risk of malignant thymic epithelial tumor. METHODS Data of 97 patients with pathologically confirmed thymic epithelial tumors treated at in our hospital from March 2015 to October 2021 were retrospectively analyzed. Based on the World Health Organization classification of thymic epithelial tumors, patients were divided into a high-risk group (types B2, B3, and C; n = 45) and a low-risk group (types A, AB, and B1; n = 52). Texture analysis was performed using a first-order, gray-level histogram method. Six features were evaluated: mean, variance, skewness, kurtosis, energy, and entropy. The association between contrast-enhanced computed tomography texture features and the risk of malignancy in thymic epithelial tumors was analyzed. The predictive thresholds of predictive texture features were determined by receiver operating characteristics analysis. RESULTS The mean, skewness, and entropy were significantly greater in the high-risk group than in the low-risk group (P < 0.05); however, variance, kurtosis, and energy were comparable in the two groups (P > 0.05). The area under curve of mean, skewness, and entropy was 0.670, 0.760, and 0.880, respectively. The optimal cutoff value of entropy for predicting risk of malignancy was 7.74, with sensitivity, specificity, and accuracy of 80.0%, 80.0%, and 75%, respectively. CONCLUSIONS Contrast-enhanced computed tomography texture features, especially entropy, may be a useful tool to predict the risk of malignancy in thymic epithelial tumors.
Collapse
Affiliation(s)
- Wei Guo
- From the Department of Radiology, Peking University Third Hospital, Beijing
| | - Jianfang Liu
- Department of Radiology, Union Hospital of Fujian Medical University, Fuzhou, PR China
| | - Xiaohua Wang
- From the Department of Radiology, Peking University Third Hospital, Beijing
| | - Huishu Yuan
- From the Department of Radiology, Peking University Third Hospital, Beijing
| |
Collapse
|
42
|
Haneberg AG, Pierre K, Winter-Reinhold E, Hochhegger B, Peters KR, Grajo J, Arreola M, Asadizanjani N, Bian J, Mancuso A, Forghani R. Introduction to Radiomics and Artificial Intelligence: A Primer for Radiologists. Semin Roentgenol 2023; 58:152-157. [PMID: 37087135 DOI: 10.1053/j.ro.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 04/03/2023]
Abstract
Health informatics and artificial intelligence (AI) are expected to transform the healthcare enterprise and the future practice of radiology. There is an increasing body of literature on radiomics and deep learning/AI applications in medical imaging. There are also a steadily increasing number of FDA cleared AI applications in radiology. It is therefore essential for radiologists to have a basic understanding of these approaches, whether in academia or private practice. In this article, we will provide an overview of the field and familiarize the readers with the fundamental concepts behind these approaches.
Collapse
|
43
|
Patkulkar P, Subbalakshmi AR, Jolly MK, Sinharay S. Mapping Spatiotemporal Heterogeneity in Tumor Profiles by Integrating High-Throughput Imaging and Omics Analysis. ACS OMEGA 2023; 8:6126-6138. [PMID: 36844580 PMCID: PMC9948167 DOI: 10.1021/acsomega.2c06659] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/05/2023] [Indexed: 05/14/2023]
Abstract
Intratumoral heterogeneity associates with more aggressive disease progression and worse patient outcomes. Understanding the reasons enabling the emergence of such heterogeneity remains incomplete, which restricts our ability to manage it from a therapeutic perspective. Technological advancements such as high-throughput molecular imaging, single-cell omics, and spatial transcriptomics allow recording of patterns of spatiotemporal heterogeneity in a longitudinal manner, thus offering insights into the multiscale dynamics of its evolution. Here, we review the latest technological trends and biological insights from molecular diagnostics as well as spatial transcriptomics, both of which have witnessed burgeoning growth in the recent past in terms of mapping heterogeneity within tumor cell types as well as the stromal constitution. We also discuss ongoing challenges, indicating possible ways to integrate insights across these methods to have a systems-level spatiotemporal map of heterogeneity in each tumor and a more systematic investigation of the implications of heterogeneity for patient outcomes.
Collapse
|
44
|
Jiang J, Wei J, Zhu Y, Wei L, Wei X, Tian H, Zhang L, Wang T, Cheng Y, Zhao Q, Sun Z, Du H, Huang Y, Liu H, Li Y. Clot-based radiomics model for cardioembolic stroke prediction with CT imaging before recanalization: a multicenter study. Eur Radiol 2023; 33:970-980. [PMID: 36066731 DOI: 10.1007/s00330-022-09116-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/11/2022] [Accepted: 08/12/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVES To develop a clot-based radiomics model using CT imaging radiomic features and machine learning to identify cardioembolic (CE) stroke before mechanical thrombectomy (MTB) in patients with acute ischemic stroke (AIS). MATERIALS AND METHODS This retrospective four-center study consecutively included 403 patients with AIS who sequentially underwent CT and MTB between April 2016 and July 2021. These were grouped into training, testing, and external validation cohorts. Thrombus-extracted radiomic features and basic information were gathered to construct a machine learning model to predict CE stroke. The radiological characteristics and basic information were used to build a routine radiological model. A combined radiomics and radiological features model was also developed. The performances of all models were evaluated and compared in the validation cohort. A histological analysis helped further assess the proposed model in all patients. RESULTS The radiomics model yielded an area under the curve (AUC) of 0.838 (95% confidence interval [CI], 0.771-0.891) for predicting CE stroke in the validation cohort, significantly higher than the radiological model (AUC, 0.713; 95% CI, 0.636-0.781; p = 0.007) but similar to the combined model (AUC, 0.855; 95% CI, 0.791-0.906; p = 0.14). The thrombus radiomic features achieved stronger correlations with red blood cells (|rmax|, 0.74 vs. 0.32) and fibrin and platelet (|rmax|, 0.68 vs. 0.18) than radiological characteristics. CONCLUSION The proposed CT-based radiomics model could reliably predict CE stroke in AIS, performing better than the routine radiological method. KEY POINTS • Admission CT imaging could offer valuable information to identify the acute ischemic stroke source by radiomics analysis. • The proposed CT imaging-based radiomics model yielded a higher area under the curve (0.838) than the routine radiological method (0.713; p = 0.007). • Several radiomic features showed significantly stronger correlations with two main thrombus constituents (red blood cells, |rmax|, 0.74; fibrin and platelet, |rmax|, 0.68) than routine radiological characteristics.
Collapse
Affiliation(s)
- Jingxuan Jiang
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.,Department of Radiology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Jianyong Wei
- Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yueqi Zhu
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Liming Wei
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Xiaoer Wei
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Hao Tian
- Department of Radiology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Lei Zhang
- Department of Radiology, Wuxi Second People's Hospital, Wuxi, 214000, China
| | - Tianle Wang
- Department of Radiology, Affiliated No. 1 People's Hospital of Nantong University, Nantong, 226001, China
| | - Yue Cheng
- Department of Radiology, Wuxi Second People's Hospital, Wuxi, 214000, China
| | - Qianqian Zhao
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Zheng Sun
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Haiyan Du
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Yu Huang
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Hui Liu
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Yuehua Li
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
45
|
McCague C, Ramlee S, Reinius M, Selby I, Hulse D, Piyatissa P, Bura V, Crispin-Ortuzar M, Sala E, Woitek R. Introduction to radiomics for a clinical audience. Clin Radiol 2023; 78:83-98. [PMID: 36639175 DOI: 10.1016/j.crad.2022.08.149] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/31/2022] [Indexed: 01/12/2023]
Abstract
Radiomics is a rapidly developing field of research focused on the extraction of quantitative features from medical images, thus converting these digital images into minable, high-dimensional data, which offer unique biological information that can enhance our understanding of disease processes and provide clinical decision support. To date, most radiomics research has been focused on oncological applications; however, it is increasingly being used in a raft of other diseases. This review gives an overview of radiomics for a clinical audience, including the radiomics pipeline and the common pitfalls associated with each stage. Key studies in oncology are presented with a focus on both those that use radiomics analysis alone and those that integrate its use with other multimodal data streams. Importantly, clinical applications outside oncology are also presented. Finally, we conclude by offering a vision for radiomics research in the future, including how it might impact our practice as radiologists.
Collapse
Affiliation(s)
- C McCague
- Department of Radiology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - S Ramlee
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - M Reinius
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - I Selby
- Department of Radiology, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - D Hulse
- Department of Radiology, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - P Piyatissa
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - V Bura
- Department of Radiology, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Radiology and Medical Imaging, County Clinical Emergency Hospital, Cluj-Napoca, Romania
| | - M Crispin-Ortuzar
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Department of Oncology, University of Cambridge, Cambridge, UK
| | - E Sala
- Department of Radiology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - R Woitek
- Department of Radiology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Research Centre for Medical Image Analysis and Artificial Intelligence (MIAAI), Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| |
Collapse
|
46
|
Liu J, Wang X, Sahin IH, Imanirad I, Felder SI, Kim RD, Xie H. Tumor Response-speed Heterogeneity as a Novel Prognostic Factor in Patients With Metastatic Colorectal Cancer. Am J Clin Oncol 2023; 46:50-57. [PMID: 36606664 DOI: 10.1097/coc.0000000000000972] [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: 01/07/2023]
Abstract
PURPOSE Differential tumor response to therapy is partially attributed to tumor heterogeneity. Additional efforts are needed to identify tumor heterogeneity parameters in response to therapy that is easily applicable in clinical practice. We aimed to describe tumor response-speed heterogeneity and evaluate its prognostic value in patients with metastatic colorectal cancer. PATIENTS AND METHODS Individual patient data from Amgen (NCT00364013) and Sanofi (NCT00305188; NCT00272051) trials were retrieved from Project Data Sphere. Patients in the Amgen 5-fluorouracil, leucovorin, oxaliplatin (FOLFOX) arm were used to establish response-speed heterogeneity. Its prognostic value was subsequently validated in the Sanofi FOLFOX arms and the Amgen panitumumab+FOLFOX arm. Kaplan-Meier method and Cox proportional hazards models were used for survival analyses. RESULTS Patients with high response-speed heterogeneity in the Amgen FOLFOX cohort had significantly shorter ( P <0.001) median progression-free survival (PFS) of 7.27 months (95% CI, 6.12-7.96 mo) and overall survival (OS) of 16.0 months (95% CI, 13.8-18.2 mo) than patients with low response-speed heterogeneity with median PFS of 9.41 months (95% CI, 8.75-10.89 mo) and OS of 22.4 months (95% CI, 20.1-26.7 mo), respectively. Tumor response-speed heterogeneity was a poor prognostic factor of shorter PFS (hazard ratio, 4.17; 95% CI, 2.49-6.99; P <0.001) and shorter OS (hazard ratio, 2.57; 95% CI, 1.64-4.01; P <0.001), after adjustment for other common prognostic factors. Comparable findings were found in the external validation cohorts. CONCLUSION Tumor response-speed heterogeneity to first-line chemotherapy was a novel prognostic factor associated with early disease progression and shorter survival in patients with metastatic colorectal cancer.
Collapse
Affiliation(s)
- Junjia Liu
- Albert Einstein College of Medicine, Bronx, New York
| | | | - Ibrahim H Sahin
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Iman Imanirad
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Seth I Felder
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Richard D Kim
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Hao Xie
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
47
|
Stoehr F, Kloeckner R, Pinto dos Santos D, Schnier M, Müller L, Mähringer-Kunz A, Dratsch T, Schotten S, Weinmann A, Galle PR, Mittler J, Düber C, Hahn F. Radiomics-Based Prediction of Future Portal Vein Tumor Infiltration in Patients with HCC-A Proof-of-Concept Study. Cancers (Basel) 2022; 14:cancers14246036. [PMID: 36551521 PMCID: PMC9775514 DOI: 10.3390/cancers14246036] [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: 11/11/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Portal vein infiltration (PVI) is a typical complication of HCC. Once diagnosed, it leads to classification as BCLC C with an enormous impact on patient management, as systemic therapies are henceforth recommended. Our aim was to investigate whether radiomics analysis using imaging at initial diagnosis can predict the occurrence of PVI in the course of disease. Between 2008 and 2018, we retrospectively identified 44 patients with HCC and an in-house, multiphase CT scan at initial diagnosis who presented without CT-detectable PVI but developed it in the course of disease. Accounting for size and number of lesions, growth type, arterial enhancement pattern, Child-Pugh stage, AFP levels, and subsequent therapy, we matched 44 patients with HCC who did not develop PVI to those developing PVI in the course of disease (follow-up ended December 2021). After segmentation of the tumor at initial diagnosis and texture analysis, we used LASSO regression to find radiomics features suitable for PVI detection in this matched set. Using an 80:20 split between training and holdout validation dataset, 17 radiomics features remained in the fitted model. Applying the model to the holdout validation dataset, sensitivity to detect occurrence of PVI was 0.78 and specificity was 0.78. Radiomics feature extraction had the ability to detect aggressive HCC morphology likely to result in future PVI. An additional radiomics evaluation at initial diagnosis might be a useful tool to identify patients with HCC at risk for PVI during follow-up benefiting from a closer surveillance.
Collapse
Affiliation(s)
- Fabian Stoehr
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Roman Kloeckner
- Institute of Interventional Radiology, University Hospital Schleswig-Holstein—Campus Luebeck, 23562 Luebeck, Germany
| | - Daniel Pinto dos Santos
- Institute of Diagnostic and Interventional Radiology, University Hospital Frankfurt, 60590 Frankfurt, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital of Cologne, 50937 Cologne, Germany
| | - Mira Schnier
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Lukas Müller
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Aline Mähringer-Kunz
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Thomas Dratsch
- Department of Diagnostic and Interventional Radiology, University Hospital of Cologne, 50937 Cologne, Germany
| | - Sebastian Schotten
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, Helios Dr. Horst Schmidt Kliniken Wiesbaden, 65199 Wiesbaden, Germany
| | - Arndt Weinmann
- Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Peter Robert Galle
- Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Jens Mittler
- Department of General, Visceral and Transplant Surgery, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Christoph Düber
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Felix Hahn
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Correspondence: ; Tel.: +49-6131172019
| |
Collapse
|
48
|
Ma T, Cui J, Wang L, Li H, Ye Z, Gao X. A CT-based radiomics signature for prediction of HER2 overexpression and treatment efficacy of trastuzumab in advanced gastric cancer. Transl Cancer Res 2022; 11:4326-4337. [PMID: 36644192 PMCID: PMC9834583 DOI: 10.21037/tcr-22-1690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/26/2022] [Indexed: 11/20/2022]
Abstract
Background Accurate evaluation of human epidermal growth factor receptor 2 (HER2) status is very important for appropriate management of advanced gastric cancer (AGC) patients. In this study, we aimed to develop and validate a computed tomography (CT)-based radiomics signature for preoperative prediction of HER2 overexpression and treatment efficacy of trastuzumab in AGC. Methods We retrospectively enrolled 536 consecutive AGC patients (median age, 59 years; interquartile range, 52-65 years; 377 male, 159 female) and separated them into a training set (n=357) and a testing set (n=179). Radiomic features were extracted from 3 different phase images of contrast-enhanced CT scans, and a radiomics signature was built based on highly reproducible features using the least absolute shrinkage and selection operator (LASSO) method. The predictive performance of the radiomics signature was assessed in the training and testing sets. Univariable and multivariable logistical regression analyses were used to identify independent risk factors of HER2 overexpression. Univariable and multivariable Cox regression analyses were used to identify the risk factors of overall survival (OS) and progression-free survival (PFS). The predictive value of the radiomics signature for treatment efficacy of trastuzumab was also evaluated. Results The radiomics signature comprised eight robust features that demonstrated good discrimination ability for HER2 overexpression in the training set [area under the curve (AUC) =0.85] and the testing set (AUC =0.81). Multivariable Cox regression analysis revealed that the radiomics signature was an independent risk factor for OS [hazard ratio (HR) =2.01, P=0.001] and PFS (HR =1.32, P=0.01). The radiomics score of patients who achieved disease control was significantly lower than that of patients with progressive disease (P=0.023). Conclusions The proposed radiomics signature showed favorable accuracy for prediction of HER2 overexpression and prognosis in AGC. It has promising potential as a noninvasive approach for selecting patients for target therapy.
Collapse
Affiliation(s)
- Tingting Ma
- Department of Radiology, Tianjin Cancer Hospital Airport Hospital, Tianjin, China;,Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,National Clinical Research Center for Cancer, Tianjin, China;,Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jingli Cui
- National Clinical Research Center for Cancer, Tianjin, China;,Tianjin’s Clinical Research Center for Cancer, Tianjin, China;,Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,Department of General Surgery, Weifang People’s Hospital, Weifang, China
| | - Lingwei Wang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,National Clinical Research Center for Cancer, Tianjin, China;,Tianjin’s Clinical Research Center for Cancer, Tianjin, China;,The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hui Li
- National Clinical Research Center for Cancer, Tianjin, China;,Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,National Clinical Research Center for Cancer, Tianjin, China;,Tianjin’s Clinical Research Center for Cancer, Tianjin, China;,The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xujie Gao
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China;,National Clinical Research Center for Cancer, Tianjin, China;,Tianjin’s Clinical Research Center for Cancer, Tianjin, China;,The Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
49
|
Celli V, Guerreri M, Pernazza A, Cuccu I, Palaia I, Tomao F, Di Donato V, Pricolo P, Ercolani G, Ciulla S, Colombo N, Leopizzi M, Di Maio V, Faiella E, Santucci D, Soda P, Cordelli E, Perniola G, Gui B, Rizzo S, Della Rocca C, Petralia G, Catalano C, Manganaro L. MRI- and Histologic-Molecular-Based Radio-Genomics Nomogram for Preoperative Assessment of Risk Classes in Endometrial Cancer. Cancers (Basel) 2022; 14:5881. [PMID: 36497362 PMCID: PMC9739755 DOI: 10.3390/cancers14235881] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
High- and low-risk endometrial carcinoma (EC) differ in whether or not a lymphadenectomy is performed. We aimed to develop MRI-based radio-genomic models able to preoperatively assess lymph-vascular space invasion (LVSI) and discriminate between low- and high-risk EC according to the ESMO-ESGO-ESTRO 2020 guidelines, which include molecular risk classification proposed by "ProMisE". This is a retrospective, multicentric study that included 64 women with EC who underwent 3T-MRI before a hysterectomy. Radiomics features were extracted from T2WI images and apparent diffusion coefficient maps (ADC) after manual segmentation of the gross tumor volume. We constructed a multiple logistic regression approach from the most relevant radiomic features to distinguish between low- and high-risk classes under the ESMO-ESGO-ESTRO 2020 guidelines. A similar approach was taken to assess LVSI. Model diagnostic performance was assessed via ROC curves, accuracy, sensitivity and specificity on training and test sets. The LVSI predictive model used a single feature from ADC as a predictor; the risk class model used two features as predictors from both ADC and T2WI. The low-risk predictive model showed an AUC of 0.74 with an accuracy, sensitivity, and specificity of 0.74, 0.76, 0.94; the LVSI model showed an AUC of 0.59 with an accuracy, sensitivity, and specificity of 0.60, 0.50, 0.61. MRI-based radio-genomic models are useful for preoperative EC risk stratification and may facilitate therapeutic management.
Collapse
Affiliation(s)
- Veronica Celli
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy
| | - Michele Guerreri
- Department of Computer Science & Centre for Medical Image Computing, University College London, London WC1E 6BT, UK
- AINOSTICS, Manchester M2 3NG, UK
- Radiomics Core Research Facility, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, 00168 Roma, Italy
| | - Angelina Pernazza
- Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino-Sapienza University, 04100 Latina, Italy
| | - Ilaria Cuccu
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico 166, 00161 Rome, Italy
| | - Innocenza Palaia
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico 166, 00161 Rome, Italy
| | - Federica Tomao
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico 166, 00161 Rome, Italy
| | - Violante Di Donato
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico 166, 00161 Rome, Italy
| | - Paola Pricolo
- Department of Radiology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giada Ercolani
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy
| | - Sandra Ciulla
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy
| | - Nicoletta Colombo
- Division of Gynecologic Oncology, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20099 Milano, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Monza, Italy
| | - Martina Leopizzi
- Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino-Sapienza University, 04100 Latina, Italy
| | - Valeria Di Maio
- Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino-Sapienza University, 04100 Latina, Italy
| | - Eliodoro Faiella
- Medical Oncology Department, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Domiziana Santucci
- Unit of Diagnostic Imaging, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Paolo Soda
- Unit of Computer Systems and Bioinformatics, Department of Engineering, University Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Ermanno Cordelli
- Unit of Computer Systems and Bioinformatics, Department of Engineering, University Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Giorgia Perniola
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico 166, 00161 Rome, Italy
| | - Benedetta Gui
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Stefania Rizzo
- Clinica di Radiologia EOC, Istituto Imaging della Svizzera Italiana (IIMSI), 6900 Lugano, Switzerland
| | - Carlo Della Rocca
- Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino-Sapienza University, 04100 Latina, Italy
| | - Giuseppe Petralia
- Department of Radiology, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Carlo Catalano
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy
| | - Lucia Manganaro
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
50
|
Mehta P, Sinha S, Kashid S, Chakraborty D, Mhatre R, Murthy V. Exploring Texture Analysis to Optimize Bladder Preservation in Muscle Invasive Bladder Cancer. Clin Genitourin Cancer 2022; 21:e138-e144. [PMID: 36628695 DOI: 10.1016/j.clgc.2022.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
Abstract
PURPOSE To explore if texture analysis of Muscle Invasive Bladder Cancer (MIBC) can aid in better patient selection for bladder preservation. METHODS Pretreatment noncontrast CT images of 41 patients of MIBC treated with bladder preservation were included. The visible tumor was contoured on all slices by a single observer. The primary endpoint was to identify texture parameters associated with disease recurrence posttreatment. The secondary endpoints included intra and interobserver variability, single and multislice analysis, and differentiating the texture features of normal bladder and tumor. For interobserver variability of bladder tumor texture features, 3 observers contoured the visible tumor on all slices independently. Observer 1 contoured again at an interval of 1 month for intraobserver variability. RESULTS The median follow-up was 30 months with 12 patients having a recurrence. In the primary endpoint analysis, the mean of the pixels at Spatial Scaling Filter (SSF) 2 for the no recurrence group and recurrence group was 6.44 v 13.73 respectively (P = .031) and the same at SSF-3 was 11.95 and 22.32 respectively (P = .034). The texture features that could significantly differentiate tumor and normal bladder were mean, standard deviation and kurtosis of the pixels at SSF-2 and entropy and kurtosis of the pixels at SSF-3. Overall, there was an excellent intra and interobserver concordance in texture features. Only multislice analysis and not single-slice could differentiate recurrence and no recurrence posttreatment. CONCLUSIONS Texture analysis can be explored as a modality for patient selection for bladder preservation along with the established clinical parameters to improve outcomes.
Collapse
Affiliation(s)
- Prachi Mehta
- Department of Radiation Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Shwetabh Sinha
- Department of Radiation Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sheetal Kashid
- Department of Radiation Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Debanjan Chakraborty
- Department of Radiation Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Ritesh Mhatre
- Department of Medical Physics, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Vedang Murthy
- Department of Radiation Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|