1
|
Eghbali R, Nedelec P, Weiss D, Bhalerao R, Xie L, Rudie JD, Liu C, Sugrue LP, Rauschecker AM. Automated Lesion and Feature Extraction Pipeline for Brain MRIs with Interpretability. Neuroinformatics 2025; 23:2. [PMID: 39786657 PMCID: PMC11717894 DOI: 10.1007/s12021-024-09708-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] [Accepted: 09/11/2024] [Indexed: 01/12/2025]
Abstract
This paper introduces the Automated Lesion and Feature Extraction (ALFE) pipeline, an open-source, Python-based pipeline that consumes MR images of the brain and produces anatomical segmentations, lesion segmentations, and human-interpretable imaging features describing the lesions in the brain. ALFE pipeline is modeled after the neuroradiology workflow and generates features that can be used by physicians for quantitative analysis of clinical brain MRIs and for machine learning applications. The pipeline uses a decoupled design which allows the user to customize the image processing, image registrations, and AI segmentation tools without the need to change the business logic of the pipeline. In this manuscript, we give an overview of ALFE, present the main aspects of ALFE pipeline design philosophy, and present case studies.
Collapse
Affiliation(s)
- Reza Eghbali
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA.
- Berekely Institute for Data Science, University of California, Berkeley, Berkeley, CA, USA.
| | - Pierre Nedelec
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - David Weiss
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Radhika Bhalerao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Long Xie
- Siemens Healthineers, Erlangen, Germany
| | - Jeffrey D Rudie
- Department of Radiology, University of California, San Diego, San Diego, CA, USA
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Leo P Sugrue
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Andreas M Rauschecker
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
2
|
Liu J, Tu J, Yao L, Peng L, Fang R, Lu Y, He F, Xiong J, Li Y. MRI-based radiomics virtual biopsy for BCL6 in primary central nervous system lymphoma. Clin Radiol 2025; 80:106746. [PMID: 39615185 DOI: 10.1016/j.crad.2024.106746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/06/2024] [Accepted: 11/04/2024] [Indexed: 01/18/2025]
Abstract
AIM To establish a machine learning model based on a radiomic signature for predicting B-cell lymphoma 6 (BCL-6) rearrangement in primary central nervous system lymphoma (PCNSL). MATERIALS AND METHODS Retrospective study on 102 PCNSL patients (31 with BCL-6 rearrangement positive, 71 with BCL-6 rearrangement negative) were randomly divided into the training and validation sets at a ratio of 7:3. Radiomics models based on contrast-enhanced T1-weighted imaging (CE-T1WI) and fluid-attenuated inversion recovery (FLAIR) in different regions, including VOItumour core and VOIperitumoural oedema. Radiomics features were extracted and selected using LASSO regression, and radiomics score (rad-score) were calculated using the weighted coefficients. Four machine learning models (logistic regression, random forest, support vector machine, K-nearest neighbours) were developed and evaluated based on rad-score. The optimal radiomics model was integrated into the clinical or radiological factors to construct a predictive model through logistic regression analysis. A nomogram was constructed based on independent significant features for individualised prediction. RESULTS All rad-scores based on CE-T1WI and FLAIR sequences were significantly associated with BCL6 rearrangement (p < 0.05) in univariate regression analysis. The logistic regression machine learning model performed best with AUCs of 0.935 (training) and 0.923 (validation). Rad-scores from CE-T1WI tumour core and peritumoural oedema were independent significant predictors. CONCLUSION Radiomics signatures based on CE-T1WI and FLAIR sequences have significant value in distinguishing BCL6 rearrangement. The CE-T1WI radiomics model based on VOItumour core and VOIperitumoural oedema are robust markers for identifying BCL6 rearrangement.
Collapse
Affiliation(s)
- J Liu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - J Tu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - L Yao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China
| | - L Peng
- Department of Radiology, Guangdong Provincial People Hospital Nanhai Hospital, Foshan, Guangdong Province, China
| | - R Fang
- Department of Radiology, Chizhou People Hospital, Chizhou, Anhui Province, China
| | - Y Lu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - F He
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - J Xiong
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Y Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Mylona E, Zaridis DI, Kalantzopoulos CΝ, Tachos NS, Regge D, Papanikolaou N, Tsiknakis M, Marias K, Fotiadis DI. Optimizing radiomics for prostate cancer diagnosis: feature selection strategies, machine learning classifiers, and MRI sequences. Insights Imaging 2024; 15:265. [PMID: 39495422 PMCID: PMC11535140 DOI: 10.1186/s13244-024-01783-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/27/2024] [Indexed: 11/05/2024] Open
Abstract
OBJECTIVES Radiomics-based analyses encompass multiple steps, leading to ambiguity regarding the optimal approaches for enhancing model performance. This study compares the effect of several feature selection methods, machine learning (ML) classifiers, and sources of radiomic features, on models' performance for the diagnosis of clinically significant prostate cancer (csPCa) from bi-parametric MRI. METHODS Two multi-centric datasets, with 465 and 204 patients each, were used to extract 1246 radiomic features per patient and MRI sequence. Ten feature selection methods, such as Boruta, mRMRe, ReliefF, recursive feature elimination (RFE), random forest (RF) variable importance, L1-lasso, etc., four ML classifiers, namely SVM, RF, LASSO, and boosted generalized linear model (GLM), and three sets of radiomics features, derived from T2w images, ADC maps, and their combination, were used to develop predictive models of csPCa. Their performance was evaluated in a nested cross-validation and externally, using seven performance metrics. RESULTS In total, 480 models were developed. In nested cross-validation, the best model combined Boruta with Boosted GLM (AUC = 0.71, F1 = 0.76). In external validation, the best model combined L1-lasso with boosted GLM (AUC = 0.71, F1 = 0.47). Overall, Boruta, RFE, L1-lasso, and RF variable importance were the top-performing feature selection methods, while the choice of ML classifier didn't significantly affect the results. The ADC-derived features showed the highest discriminatory power with T2w-derived features being less informative, while their combination did not lead to improved performance. CONCLUSION The choice of feature selection method and the source of radiomic features have a profound effect on the models' performance for csPCa diagnosis. CRITICAL RELEVANCE STATEMENT This work may guide future radiomic research, paving the way for the development of more effective and reliable radiomic models; not only for advancing prostate cancer diagnostic strategies, but also for informing broader applications of radiomics in different medical contexts. KEY POINTS Radiomics is a growing field that can still be optimized. Feature selection method impacts radiomics models' performance more than ML algorithms. Best feature selection methods: RFE, LASSO, RF, and Boruta. ADC-derived radiomic features yield more robust models compared to T2w-derived radiomic features.
Collapse
Affiliation(s)
- Eugenia Mylona
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Dimitrios I Zaridis
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
- Biomedical Engineering Laboratory, School of Electrical & Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Charalampos Ν Kalantzopoulos
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Nikolaos S Tachos
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece
| | - Daniele Regge
- Department of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Manolis Tsiknakis
- Computational Biomedicine Laboratory, Institute of Computer Science, FORTH, GR 70013, Heraklion, Greece
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, GR 71004, Heraklion, Greece
| | - Kostas Marias
- Computational Biomedicine Laboratory, Institute of Computer Science, FORTH, GR 70013, Heraklion, Greece
- Department of Electrical and Computer Engineering, Hellenic Mediterranean University, GR 71004, Heraklion, Greece
| | - Dimitrios I Fotiadis
- Biomedical Research Institute, FORTH, GR 45110, Ioannina, Greece.
- Unit of Medical Technology Intelligent Information Systems, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
4
|
Ko CC, Liu YL, Hung KC, Yang CC, Lim SW, Yeh LR, Chen JH, Su MY. MRI-Based Machine Learning for Prediction of Clinical Outcomes in Primary Central Nervous System Lymphoma. Life (Basel) 2024; 14:1290. [PMID: 39459590 PMCID: PMC11509076 DOI: 10.3390/life14101290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
A portion of individuals diagnosed with primary central nervous system lymphomas (PCNSL) may experience early relapse or refractory (R/R) disease following treatment. This research explored the potential of MRI-based radiomics in forecasting R/R cases in PCNSL. Forty-six patients with pathologically confirmed PCNSL diagnosed between January 2008 and December 2020 were included in this study. Only patients who underwent pretreatment brain MRIs and complete postoperative follow-up MRIs were included. Pretreatment contrast-enhanced T1WI, T2WI, and T2 FLAIR imaging were analyzed. A total of 107 radiomic features, including 14 shape-based, 18 first-order statistical, and 75 texture features, were extracted from each sequence. Predictive models were then built using five different machine learning algorithms to predict R/R in PCNSL. Of the included 46 PCNSL patients, 20 (20/46, 43.5%) patients were found to have R/R. In the R/R group, the median scores in predictive models such as support vector machine, k-nearest neighbors, linear discriminant analysis, naïve Bayes, and decision trees were significantly higher, while the apparent diffusion coefficient values were notably lower compared to those without R/R (p < 0.05). The support vector machine model exhibited the highest performance, achieving an overall prediction accuracy of 83%, a precision rate of 80%, and an AUC of 0.78. Additionally, when analyzing tumor progression, patients with elevated support vector machine and naïve Bayes scores demonstrated a significantly reduced progression-free survival (p < 0.05). These findings suggest that preoperative MRI-based radiomics may provide critical insights for treatment strategies in PCNSL.
Collapse
Affiliation(s)
- Ching-Chung Ko
- Department of Medical Imaging, Chi Mei Medical Center, Tainan 71004, Taiwan;
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Yan-Lin Liu
- Department of Radiological Sciences, University of California, Irvine, CA 92697, USA; (Y.-L.L.); (J.-H.C.); (M.-Y.S.)
| | - Kuo-Chuan Hung
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 710, Taiwan;
- Department of Hospital and Health Care Administration, College of Recreation and Health Management, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| | - Cheng-Chun Yang
- Department of Medical Imaging, Chi Mei Medical Center, Tainan 71004, Taiwan;
| | - Sher-Wei Lim
- Department of Neurosurgery, Chi Mei Medical Center, Chiali, Tainan 722, Taiwan;
- Department of Nursing, Min-Hwei College of Health Care Management, Tainan 736, Taiwan
| | - Lee-Ren Yeh
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung 824, Taiwan;
- Department of Medical Imaging and Radiological Sciences, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan
| | - Jeon-Hor Chen
- Department of Radiological Sciences, University of California, Irvine, CA 92697, USA; (Y.-L.L.); (J.-H.C.); (M.-Y.S.)
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung 824, Taiwan;
| | - Min-Ying Su
- Department of Radiological Sciences, University of California, Irvine, CA 92697, USA; (Y.-L.L.); (J.-H.C.); (M.-Y.S.)
| |
Collapse
|
5
|
Li H, Xiong M, Li M, Sun C, Zheng D, Yuan L, Chen Q, Lin S, Liu Z, Ren X. Radiomic prediction for durable response to high-dose methotrexate-based chemotherapy in primary central nervous system lymphoma. Cancer Med 2024; 13:e70182. [PMID: 39253996 PMCID: PMC11386301 DOI: 10.1002/cam4.70182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND The rarity of primary central nervous system lymphoma (PCNSL) and treatment heterogeneity contributes to a lack of prognostic models for evaluating posttreatment remission. This study aimed to develop and validate radiomic-based models to predict the durable response (DR) to high-dose methotrexate (HD-MTX)-based chemotherapy in PCNSL patients. METHODS A total of 159 patients pathologically diagnosed with PCNSL between 2011 and 2021 across two institutions were enrolled. According to the NCCN guidelines, the DR was defined as the remission lasting ≥1 year after receiving HD-MTX-based chemotherapy. For each patient, a total of 1218 radiomic features were extracted from prebiopsy T1 contrast-enhanced MR images. Multiple machine-learning algorithms were utilized for feature selection and classification to build a radiomic signature. The radiomic-clinical integrated models were developed using the random forest method. Model performance was externally validated to verify its clinical utility. RESULTS A total of 105 PCNSL patients were enrolled after excluding 54 cases with ineligibility. The training and validation cohorts comprised 76 and 29 individuals, respectively. Among them, 65 patients achieved DR. The radiomic signature, consisting of 8 selected features, demonstrated strong predictive performance, with area under the curves of 0.994 in training cohort and 0.913 in validation cohort. This signature was independently associated with the DR in both cohorts. Both the radiomic signature and integrated models significantly outperformed the clinical models in two cohorts. Decision curve analysis underscored the clinical utility of the established models. CONCLUSIONS This radiomic signature and integrated models have the potential to accurately predict the DR to HD-MTX-based chemotherapy in PCNSL patients, providing valuable therapeutic insights.
Collapse
Affiliation(s)
- Haoyi Li
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Mingming Xiong
- National Genomics Data CenterBeijing Institute of Genomics, Chinese Academy of Sciences and China National Center for BioinformationBeijingChina
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of SciencesBeijingChina
| | - Ming Li
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Caixia Sun
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of SciencesBeijingChina
| | - Dao Zheng
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Leilei Yuan
- Department of Nuclear MedicineBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Qian Chen
- Department of Nuclear MedicineBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Song Lin
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Zhenyu Liu
- School of Artificial Intelligence, University of Chinese Academy of SciencesBeijingChina
| | - Xiaohui Ren
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
6
|
Liu J, Tu J, Xu L, Liu F, Lu Y, He F, Li A, Li Y, Liu S, Xiong J. MRI-based radiomics signatures for preoperative prediction of Ki-67 index in primary central nervous system lymphoma. Eur J Radiol 2024; 178:111603. [PMID: 38976966 DOI: 10.1016/j.ejrad.2024.111603] [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: 04/30/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
PURPOSE The aim of this study was to develop and validate radiomics signatures based on MRI for preoperative prediction of Ki-67 proliferative index (PI) expression in primary central nervous system lymphoma (PCNSL). METHODS A total of 341 patients with PCNSL were retrospectively analyzed, including 286 patients in one center as the training set and 55 patients in another two centers as the external validation set. Radiomics features were extracted and selected from preoperative contrast-enhanced T1-weighted images, fluid attenuation inversion recovery to build radiomics signatures according to the Ki-67 PI. The predictive performances of the radiomics model were evaluated using four classifiers including random forest, K-Nearest Neighbors, Neural Network and Decision Tree. A combined model was built by incorporating radiomics signature, clinical variables and MRI radiological characteristics using multivariate logistic regression analysis, and a nomogram was established to predict the expression of Ki-67 individually. The predictive performances of the models were evaluated using area under receiver operating characteristic curve (AUC) and decision curve analysis (DCA). RESULTS Radiomics signatures were independent predictors of the expression level of Ki-67 (OR: 2.523, P < 0.001). RF radiomics models had the highest accuracy (0.934 in the training set and 0.811 in the external validation set) and F1 Score (0.920 in the training set and 0.836 in the external validation set). The clinic-radiologic-radiomics nomogram showed better predictive performance with AUCs of 0.877(95 % CI: 0.837-0.918) in the training set and 0.866(95 % CI: 0.774-0.957) in the external validation set. The calibration curve and DCA demonstrated goodness-of-fit and improved benefits in clinical practice of the nomogram. CONCLUSIONS Nomograms integrating MRI-based radiomics and clinical-radiological characteristics could effectively predict Ki-67 PI in primary PCNSL.
Collapse
Affiliation(s)
- Jianpeng Liu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Tu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Linghui Xu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fangfei Liu
- Department of Nuclear Medicine, The Second Affiliated Hospital, Shandong First Medical University, Tai'an, Shandong, China
| | - Yucheng Lu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fanru He
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Anning Li
- Department of Radiology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Yuxin Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuyong Liu
- Department of Nuclear Medicine, The Second Affiliated Hospital, Shandong First Medical University, Tai'an, Shandong, China.
| | - Ji Xiong
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Isavand P, Aghamiri SS, Amin R. Applications of Multimodal Artificial Intelligence in Non-Hodgkin Lymphoma B Cells. Biomedicines 2024; 12:1753. [PMID: 39200217 PMCID: PMC11351272 DOI: 10.3390/biomedicines12081753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Given advancements in large-scale data and AI, integrating multimodal artificial intelligence into cancer research can enhance our understanding of tumor behavior by simultaneously processing diverse biomedical data types. In this review, we explore the potential of multimodal AI in comprehending B-cell non-Hodgkin lymphomas (B-NHLs). B-cell non-Hodgkin lymphomas (B-NHLs) represent a particular challenge in oncology due to tumor heterogeneity and the intricate ecosystem in which tumors develop. These complexities complicate diagnosis, prognosis, and therapy response, emphasizing the need to use sophisticated approaches to enhance personalized treatment strategies for better patient outcomes. Therefore, multimodal AI can be leveraged to synthesize critical information from available biomedical data such as clinical record, imaging, pathology and omics data, to picture the whole tumor. In this review, we first define various types of modalities, multimodal AI frameworks, and several applications in precision medicine. Then, we provide several examples of its usage in B-NHLs, for analyzing the complexity of the ecosystem, identifying immune biomarkers, optimizing therapy strategy, and its clinical applications. Lastly, we address the limitations and future directions of multimodal AI, highlighting the need to overcome these challenges for better clinical practice and application in healthcare.
Collapse
Affiliation(s)
- Pouria Isavand
- Department of Radiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan 4513956184, Iran
| | | | - Rada Amin
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68503, USA
| |
Collapse
|
8
|
Liu J, Tu J, Hu B, Li C, Piao S, Lu Y, Li A, Ding T, Xiong J, Zhu F, Li Y. Prognostic Assessment in Patients With Primary Diffuse Large B-Cell Lymphoma of the Central Nervous System Using MRI-Based Radiomics. J Magn Reson Imaging 2024. [PMID: 38970331 DOI: 10.1002/jmri.29533] [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: 02/22/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Primary central nervous system lymphoma (PCNSL) carries a poor prognosis. Radiomics may hold potential value in prognostic assessment. PURPOSE To develop and validate an MRI-based radiomics model and combine it with clinical factors to assess progression-free survival (PFS) and overall survival (OS) of patients with PCNSL. STUDY TYPE Retrospective and prospective. POPULATION Three hundred seventy-nine patients (179 female, 53 ± 7 years) from 2014 to 2022. FIELD STRENGTH/SEQUENCE T2/fluid-attenuated inversion recovery, contrast-enhanced T1WI and diffusion-weighted echo-planar imaging sequences on 3.0 T. ASSESSMENT Radiomics features were extracted from enhanced tumor regions on preoperative multi-sequence MRI. Using a least absolute shrinkage and selection operator (LASSO) Cox regression model to select radiomic signatures in training cohort (N = 169). Cox proportional hazards models were constructed for clinical, radiomics, and combined models, with internal (N = 72) and external (N = 32) cohorts validating model performance. STATISTICAL TESTS Chi-squared, Mann-Whitney, Kaplan-Meier, log-rank, LASSO, Cox, decision curve analysis, time-dependent Receiver Operating Characteristic, area under the curve (AUC), and likelihood ratio test. P-value <0.05 was considered significant. RESULTS Follow-up duration was 28.79 ± 22.59 months (median: 25). High-risk patients, determined by the median radiomics score, showed significantly lower survival rates than low-risk patients. Compared with NCCN-IPI, conventional imaging and clinical models, the combined model achieved the highest C-index for both PFS (0.660 internal, 0.802 external) and OS (0.733 internal, 0.781 external) in validation. Net benefit was greater with radiomics than with clinical alone. The combined model exhibited performance with AUCs of 0.680, 0.752, and 0.830 for predicting 1-year, 3-year, and 5-year PFS, and 0.770, 0.789, and 0.863 for OS in internal validation, with PFS AUCs of 0.860 and 0.826 and OS AUCs of 0.859 and 0.748 for 1-year and 3-year survival in external validation. DATA CONCLUSION Incorporating a multi-sequence MR-based radiomics model into clinical models enhances the assess accuracy for the prognosis of PCNSL. EVIDENCE LEVEL 4 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jianpeng Liu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Tu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bin Hu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chao Li
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Sirong Piao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yucheng Lu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Anning Li
- Department of Radiology, Qilu Hospital, Shandong University, Jinan, China
| | - Tianling Ding
- Department of Haematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ji Xiong
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fengping Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuxin Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Boubnovski Martell M, Linton-Reid K, Hindocha S, Chen M, Moreno P, Álvarez-Benito M, Salvatierra Á, Lee R, Posma JM, Calzado MA, Aboagye EO. Deep representation learning of tissue metabolome and computed tomography annotates NSCLC classification and prognosis. NPJ Precis Oncol 2024; 8:28. [PMID: 38310164 PMCID: PMC10838282 DOI: 10.1038/s41698-024-00502-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 01/04/2024] [Indexed: 02/05/2024] Open
Abstract
The rich chemical information from tissue metabolomics provides a powerful means to elaborate tissue physiology or tumor characteristics at cellular and tumor microenvironment levels. However, the process of obtaining such information requires invasive biopsies, is costly, and can delay clinical patient management. Conversely, computed tomography (CT) is a clinical standard of care but does not intuitively harbor histological or prognostic information. Furthermore, the ability to embed metabolome information into CT to subsequently use the learned representation for classification or prognosis has yet to be described. This study develops a deep learning-based framework -- tissue-metabolomic-radiomic-CT (TMR-CT) by combining 48 paired CT images and tumor/normal tissue metabolite intensities to generate ten image embeddings to infer metabolite-derived representation from CT alone. In clinical NSCLC settings, we ascertain whether TMR-CT results in an enhanced feature generation model solving histology classification/prognosis tasks in an unseen international CT dataset of 742 patients. TMR-CT non-invasively determines histological classes - adenocarcinoma/squamous cell carcinoma with an F1-score = 0.78 and further asserts patients' prognosis with a c-index = 0.72, surpassing the performance of radiomics models and deep learning on single modality CT feature extraction. Additionally, our work shows the potential to generate informative biology-inspired CT-led features to explore connections between hard-to-obtain tissue metabolic profiles and routine lesion-derived image data.
Collapse
Affiliation(s)
| | | | - Sumeet Hindocha
- Early Diagnosis and Detection Centre, National Institute for Health and Care Research Biomedical Research Centre at the Royal Marsden and Institute of Cancer Research, London, SW3 6JJ, UK
| | - Mitchell Chen
- Imperial College London Hammersmith Campus, London, SW7 2AZ, UK
| | - Paula Moreno
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, 14004, Spain
- Departamento de Cirugía Toráxica y Trasplante de Pulmón, Hospital Universitario Reina Sofía, Córdoba, 14014, Spain
| | - Marina Álvarez-Benito
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, 14004, Spain
- Unidad de Radiodiagnóstico y Cáncer de Mama, Hospital Universitario Reina Sofía, Córdoba, 14004, Spain
| | - Ángel Salvatierra
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, 14004, Spain
- Unidad de Radiodiagnóstico y Cáncer de Mama, Hospital Universitario Reina Sofía, Córdoba, 14004, Spain
| | - Richard Lee
- Early Diagnosis and Detection Centre, National Institute for Health and Care Research Biomedical Research Centre at the Royal Marsden and Institute of Cancer Research, London, SW3 6JJ, UK
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Dovehouse Street, London, SW3 6LY, UK
| | - Joram M Posma
- Imperial College London Hammersmith Campus, London, SW7 2AZ, UK
| | - Marco A Calzado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, 14004, Spain.
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, 14014, Spain.
| | - Eric O Aboagye
- Imperial College London Hammersmith Campus, London, SW7 2AZ, UK.
| |
Collapse
|
10
|
Calimeri T, Steidl C, Fiore P, Ferreri AJM. New hopes in relapsed refractory primary central nervous system lymphoma. Curr Opin Oncol 2023; 35:364-372. [PMID: 37551946 DOI: 10.1097/cco.0000000000000980] [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: 08/09/2023]
Abstract
PURPOSE OF REVIEW Patients with relapsed/refractory primary central nervous system lymphoma (rrPCNSL) have poor prognosis, with a median survival after relapse of 6.8 months. In this review, we discuss the evolving landscape and the possible future directions related to this important unmet clinical need. RECENT FINDINGS The modern two-phase approach for newly diagnosed PCNSL based on an induction using high-dose methotrexate (HD-MTX) combinations and a subsequent consolidation, has significantly improved the outcome in this setting. However, this strategy is able to cure more or less 50% of patients. rrPCNSL patients have a very poor prognosis with a reported 5-year overall survival of 18%. Late relapses (after third year) and use of high-dose chemotherapy and autologous stem cell transplantation (HDT-ASCT) represent important factors associated with a better outcome in this setting. On the basis of the growing acquisition of knowledge on the molecular characteristics of PCNSL, the use of non-chemotherapeutic drugs such as bruton tyrosine kinase inhibitors (BTK-is), immunomodulatory drugs (IMiDs) and immune checkpoint blockers (ICBs) is increasing in the last years along with the introduction of novel approaches (CAR-T cells and blood--brain barrier disruption). However, despite high responses in some cases, durations are often short, translating in outcome results still unsatisfactory. SUMMARY Treatment of rrPCNSL patients is challenging. As no standard of care exist in this setting, it is of paramount importance to acquire new knowledge related to this condition and start multidisciplinary collaboration in order to improve pts outcome.
Collapse
Affiliation(s)
| | | | - Paolo Fiore
- Lymphoma Unit, IRCCS San Raffaele Scientific Institute
- University 'Vita-Salute San Raffaele', Milan, Italy
| | | |
Collapse
|
11
|
Cornell I, Al Busaidi A, Wastling S, Anjari M, Cwynarski K, Fox CP, Martinez-Calle N, Poynton E, Maynard J, Thust SC. Early MRI Predictors of Relapse in Primary Central Nervous System Lymphoma Treated with MATRix Immunochemotherapy. J Pers Med 2023; 13:1182. [PMID: 37511795 PMCID: PMC10381964 DOI: 10.3390/jpm13071182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Primary Central Nervous System Lymphoma (PCNSL) is a highly malignant brain tumour. We investigated dynamic changes in tumour volume and apparent diffusion coefficient (ADC) measurements for predicting outcome following treatment with MATRix chemotherapy in PCNSL. Patients treated with MATRix (n = 38) underwent T1 contrast-enhanced (T1CE) and diffusion-weighted imaging (DWI) before treatment, after two cycles and after four cycles of chemotherapy. Response was assessed using the International PCNSL Collaborative Group (IPCG) imaging criteria. ADC histogram parameters and T1CE tumour volumes were compared among response groups, using one-way ANOVA testing. Logistic regression was performed to examine those imaging parameters predictive of response. Response after two cycles of chemotherapy differed from response after four cycles; of the six patients with progressive disease (PD) after four cycles of treatment, two (33%) had demonstrated a partial response (PR) or complete response (CR) after two cycles. ADCmean at baseline, T1CE at baseline and T1CE percentage volume change differed between response groups (0.005 < p < 0.038) and were predictive of MATRix treatment response (area under the curve: 0.672-0.854). Baseline ADC and T1CE metrics are potential biomarkers for risk stratification of PCNSL patients early during remission induction therapy with MATRix. Standard interim response assessment (after two cycles) according to IPCG imaging criteria does not reliably predict early disease progression in the context of a conventional treatment approach.
Collapse
Affiliation(s)
- Isabel Cornell
- UCL Institute of Neurology, Department of Brain Rehabilitation and Repair, Queen Square, London WC1N 3BG, UK
| | - Ayisha Al Busaidi
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK
- Neuroradiology Department, Kings College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Stephen Wastling
- UCL Institute of Neurology, Department of Brain Rehabilitation and Repair, Queen Square, London WC1N 3BG, UK
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK
| | - Mustafa Anjari
- UCL Institute of Neurology, Department of Brain Rehabilitation and Repair, Queen Square, London WC1N 3BG, UK
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK
- Radiology Department, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - Kate Cwynarski
- Haematology Department, University College London Hospitals NHS Foundation Trust, London NW1 2BU, UK
| | - Christopher P Fox
- School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | | | - Edward Poynton
- Haematology Department, University College London Hospitals NHS Foundation Trust, London NW1 2BU, UK
| | - John Maynard
- UCL Institute of Neurology, Department of Brain Rehabilitation and Repair, Queen Square, London WC1N 3BG, UK
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK
| | - Steffi C Thust
- UCL Institute of Neurology, Department of Brain Rehabilitation and Repair, Queen Square, London WC1N 3BG, UK
- Precision Imaging Beacon, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- Neuroradiology Department, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, UK
| |
Collapse
|
12
|
Nenning KH, Gesperger J, Furtner J, Nemc A, Roetzer-Pejrimovsky T, Choi SW, Mitter C, Leber SL, Hofmanninger J, Klughammer J, Ergüner B, Bauer M, Brada M, Chong K, Brandner-Kokalj T, Freyschlag CF, Grams A, Haybaeck J, Hoenigschnabl S, Hoffermann M, Iglseder S, Kiesel B, Kitzwoegerer M, Kleindienst W, Marhold F, Moser P, Oberndorfer S, Pinggera D, Scheichel F, Sherif C, Stockhammer G, Stultschnig M, Thomé C, Trenkler J, Urbanic-Purkart T, Weis S, Widhalm G, Wuertz F, Preusser M, Baumann B, Simonitsch-Klupp I, Nam DH, Bock C, Langs G, Woehrer A. Radiomic features define risk and are linked to DNA methylation attributes in primary CNS lymphoma. Neurooncol Adv 2023; 5:vdad136. [PMID: 38024240 PMCID: PMC10676053 DOI: 10.1093/noajnl/vdad136] [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] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Abstract
Background The prognostic roles of clinical and laboratory markers have been exploited to model risk in patients with primary CNS lymphoma, but these approaches do not fully explain the observed variation in outcome. To date, neuroimaging or molecular information is not used. The aim of this study was to determine the utility of radiomic features to capture clinically relevant phenotypes, and to link those to molecular profiles for enhanced risk stratification. Methods In this retrospective study, we investigated 133 patients across 9 sites in Austria (2005-2018) and an external validation site in South Korea (44 patients, 2013-2016). We used T1-weighted contrast-enhanced MRI and an L1-norm regularized Cox proportional hazard model to derive a radiomic risk score. We integrated radiomic features with DNA methylation profiles using machine learning-based prediction, and validated the most relevant biological associations in tissues and cell lines. Results The radiomic risk score, consisting of 20 mostly textural features, was a strong and independent predictor of survival (multivariate hazard ratio = 6.56 [3.64-11.81]) that remained valid in the external validation cohort. Radiomic features captured gene regulatory differences such as in BCL6 binding activity, which was put forth as testable treatment target for a subset of patients. Conclusions The radiomic risk score was a robust and complementary predictor of survival and reflected characteristics in underlying DNA methylation patterns. Leveraging imaging phenotypes to assess risk and inform epigenetic treatment targets provides a concept on which to advance prognostic modeling and precision therapy for this aggressive cancer.
Collapse
Affiliation(s)
- Karl-Heinz Nenning
- Department of Biomedical Imaging and Image-guided Therapy, Computational Imaging Research Laboratory, Medical University of Vienna, Vienna, Austria
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, New York, USA
| | - Johanna Gesperger
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Julia Furtner
- Division of Neuroradiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Research Center for Medical Image Analysis and Artificial Intelligence (MIAAI), Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| | - Amelie Nemc
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Roetzer-Pejrimovsky
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Seung-Won Choi
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Christian Mitter
- Division of Neuroradiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Stefan L Leber
- Division of Neuroradiology, Vascular, and Interventional Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Johannes Hofmanninger
- Department of Biomedical Imaging and Image-guided Therapy, Computational Imaging Research Laboratory, Medical University of Vienna, Vienna, Austria
| | - Johanna Klughammer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Biochemistry, Gene Center, Ludwig-Maximilians-University, München, Germany
| | - Bekir Ergüner
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Marlies Bauer
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Martina Brada
- Department of Pathology, Klinik Landstraße, Vienna, Austria
| | - Kyuha Chong
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | - Astrid Grams
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
- Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Diagnostic and Research, Graz, Austria
| | | | - Markus Hoffermann
- Department of Neurosurgery, State Hospital Feldkirch, Feldkirch, Austria
| | - Sarah Iglseder
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Kiesel
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Melitta Kitzwoegerer
- Department of Pathology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Waltraud Kleindienst
- Department of Neurology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Franz Marhold
- Department of Neurosurgery, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Patrizia Moser
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria
- Department of Pathology, Innpath, Tirolkliniken, Innsbruck, Austria
| | - Stefan Oberndorfer
- Department of Neurology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Daniel Pinggera
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Scheichel
- Department of Neurosurgery, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Camillo Sherif
- Department of Neurosurgery, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | | | | | - Claudius Thomé
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Trenkler
- Institute of Neuroradiology, Kepler University Hospital, NeuromedCampus, Johannes Kepler University of Linz, Linz, Austria
| | - Tadeja Urbanic-Purkart
- Department of Neurology, Medical University of Graz, Graz, Austria
- Division of Neuroradiology, Vascular and Interventional Radiology, Medical University of Graz, Graz, Austria
| | - Serge Weis
- Division of Neuropathology, Kepler University Hospital, NeuromedCampus, Johannes Kepler University, Linz, Austria
| | - Georg Widhalm
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Franz Wuertz
- Institute of Pathology, State Hospital Klagenfurt, Klagenfurt, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Internal Medicine 1, Medical University of Vienna, Vienna, Austria
| | - Bernhard Baumann
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | | | - Do-Hyun Nam
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Georg Langs
- Department of Biomedical Imaging and Image-guided Therapy, Computational Imaging Research Laboratory, Medical University of Vienna, Vienna, Austria
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Adelheid Woehrer
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|