1
|
Sridharan N, Salem A, Little RA, Tariq M, Cheung S, Dubec MJ, Faivre-Finn C, Parker GJM, Porta N, O'Connor JPB. Measuring repeatability of dynamic contrast-enhanced MRI biomarkers improves evaluation of biological response to radiotherapy in lung cancer. Eur Radiol 2025; 35:664-673. [PMID: 39122855 PMCID: PMC11782379 DOI: 10.1007/s00330-024-10970-7] [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: 03/19/2024] [Revised: 05/09/2024] [Accepted: 07/01/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVES To measure dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) biomarker repeatability in patients with non-small cell lung cancer (NSCLC). To use these statistics to identify which individual target lesions show early biological response. MATERIALS AND METHODS A single-centre, prospective DCE-MRI study was performed between September 2015 and April 2017. Patients with NSCLC were scanned before standard-of-care radiotherapy to evaluate biomarker repeatability and two weeks into therapy to evaluate biological response. Volume transfer constant (Ktrans), extravascular extracellular space volume fraction (ve) and plasma volume fraction (vp) were measured at each timepoint along with tumour volume. Repeatability was assessed using a within-subject coefficient of variation (wCV) and repeatability coefficient (RC). Cohort treatment effects on biomarkers were estimated using mixed-effects models. RC limits of agreement revealed which individual target lesions changed beyond that expected with biomarker daily variation. RESULTS Fourteen patients (mean age, 67 years +/- 12, 8 men) had 22 evaluable lesions (12 primary tumours, 8 nodal metastases, 2 distant metastases). The wCV (in 8/14 patients) was between 9.16% to 17.02% for all biomarkers except for vp, which was 42.44%. Cohort-level changes were significant for Ktrans and ve (p < 0.001) and tumour volume (p = 0.002). Ktrans and tumour volume consistently showed the greatest number of individual lesions showing biological response. In distinction, no individual lesions had a real change in ve despite the cohort-level change. CONCLUSION Identifying individual early biological responders provided additional information to that derived from conventional cohort cohort-level statistics, helping to prioritise which parameters would be best taken forward into future studies. CLINICAL RELEVANCE STATEMENT Dynamic contrast-enhanced magnetic resonance imaging biomarkers Ktrans and tumour volume are repeatable and detect early treatment-induced changes at both cohort and individual lesion levels, supporting their use in further evaluation of radiotherapy and targeted therapeutics. KEY POINTS Few literature studies report quantitative imaging biomarker precision, by measuring repeatability or reproducibility. Several DCE-MRI biomarkers of lung cancer tumour microenvironment were highly repeatable. Repeatability coefficient measurements enabled lesion-specific evaluation of early biological response to therapy, improving conventional assessment.
Collapse
Affiliation(s)
- Nivetha Sridharan
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK.
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK.
| | - Ahmed Salem
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Ross A Little
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Maira Tariq
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Susan Cheung
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Michael J Dubec
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Geoffrey J M Parker
- Bioxydyn Ltd, Manchester, UK
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Nuria Porta
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - James P B O'Connor
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK.
- Division of Cancer Sciences, University of Manchester, Manchester, UK.
- Radiology Department, The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
2
|
Huang M, Song C, Zhou X, Wang H, Lin Y, Wang J, Cai H, Wang M, Peng Z, Dong Z, Feng ST. Tissue-matched analysis of MRI evaluating the tumor infiltrating lymphocytes in hepatocellular carcinoma. Int J Cancer 2024. [PMID: 39635936 DOI: 10.1002/ijc.35281] [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: 05/24/2024] [Revised: 10/14/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024]
Abstract
Tumor-infiltrating lymphocytes (TILs) play critical roles in the tumor microenvironment and immunotherapy response. This study aims to explore the feasibility of multi-parametric magnetic resonance imaging (MRI) in evaluating TILs and to develop an evaluation model that considers spatial heterogeneity. Multi-parametric MRI was performed on hepatocellular carcinoma (HCC) mice (N = 28). Three-dimensional (3D) printing was employed for tissue sampling, to match the multi-parametric MRI data with tumor tissues, followed by flow cytometry analysis and next-generation RNA-sequencing. Pearson's correlation, multivariate logistic regression, and receiver operating characteristic (ROC) curve analyses were utilized to model TIL-related MRI parameters. MRI quantitative parameters, including T1 relaxation times and perfusion, were correlated with the infiltration of leukocytes, T-cells, CD4+ T-cells, CD8+ T-cells, PD1 + CD8+ T-cells, B-cells, macrophages, and regulatory T-cells (correlation coefficients ranged from -0.656 to 0.482, p <.05) in tumor tissues. TILs were clustered into inflamed and non-inflamed subclasses, with the proportion of T-cells, CD8+ T-cells, and PD1 + CD8+ T-cells significantly higher in the inflamed group compared to the non-inflamed group (43.37% vs. 25.45%, 50.83% vs. 34.90%, 40.45% vs. 29.47%, respectively; p <.001). The TIL evaluation model, based on the Z-score combining Kep and T1post, was able to distinguish between these subgroups, yielding an area under the curve of 0.816 (95% confidence interval 0.721-0.910) and a cut-off value of -0.03 (sensitivity 68.4%, specificity 91.3%). Additionally, the Z-score was related to the gene expression of T-cell activation, chemokine production, and cell adhesion. The tissue-matched analysis of multi-parametric MRI offers a feasible method of regional evaluation and can distinguish between TIL subclasses.
Collapse
Affiliation(s)
- Mengqi Huang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenyu Song
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoqi Zhou
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huanjun Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingyu Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jifei Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huasong Cai
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meng Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhenpeng Peng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhi Dong
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
3
|
Baboli M, Wang F, Dong Z, Dietrich J, Uhlmann EJ, Batchelor TT, Cahill DP, Andronesi OC. Absolute Metabolite Quantification in Individuals with Glioma and Healthy Individuals Using Whole-Brain Three-dimensional MR Spectroscopic and Echo-planar Time-resolved Imaging. Radiology 2024; 312:e232401. [PMID: 39315894 PMCID: PMC11449233 DOI: 10.1148/radiol.232401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
BACKGROUND MR spectroscopic imaging (MRSI) can be used to quantify an extended brain metabolic profile but is confounded by changes in tissue water levels due to disease. PURPOSE To develop a fast absolute quantification method for metabolite concentrations combining whole-brain MRSI with echo-planar time-resolved imaging (EPTI) relaxometry in individuals with glioma and healthy individuals. MATERIALS AND METHODS In this prospective study performed from August 2022 to August 2023, using internal water as concentration reference, the MRSI-EPTI quantification method was compared with the conventional method using population-average literature relaxation values. Healthy participants and participants with mutant IDH1 gliomas underwent imaging at 3 T with a 32-channel coil. Real-time navigated adiabatic spiral three-dimensional MRSI scans were acquired in approximately 8 minutes and reconstructed with a super-resolution pipeline to obtain brain metabolic images at 2.4-mm isotropic resolution. High-spatial-resolution multiparametric EPTI was performed in 3 minutes, with 1-mm isotropic resolution, to correct the relaxation and proton density of the water reference signal. Bland-Altman analysis and the Wilcoxon signed rank test were used to compare absolute quantifications from the proposed and conventional methods. RESULTS Six healthy participants (four male; mean age, 37 years ± 11 [SD]) and nine participants with glioma (six male; mean age, 41 years ± 15; one with wild-type IDH1 and eight with mutant IDH1) were included. In healthy participants, there was good agreement (+4% bias) between metabolic concentrations derived using the two methods, with a CI of plus or minus 26%. In participants with glioma, there was large disagreement between the two methods (+39% bias) and a CI of plus or minus 55%. The proposed quantification method improved tumor contrast-to-noise ratio (median values) for total N-acetyl-aspartate (EPTI: 0.541 [95% CI: 0.217, 0.910]; conventional: 0.484 [95% CI: 0.199, 0.823]), total choline (EPTI: 1.053 [95% CI: 0.681, 1.713]; conventional: 0.940 [95% CI: 0.617, 1.295]), and total creatine (EPTI: 0.745 [95% CI: 0.628, 0.909]; conventional: 0.553 [95% CI: 0.444, 0.828]) (P = .03 for all). CONCLUSION The whole-brain MRSI-EPTI method provided fast absolute quantification of metabolic concentrations with individual-specific corrections at 2.4-mm isotropic resolution, yielding concentrations closer to the true value in disease than the conventional literature-based corrections. © RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Mehran Baboli
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Fuyixue Wang
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Zijing Dong
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Jorg Dietrich
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Erik J Uhlmann
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Tracy T Batchelor
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Daniel P Cahill
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| | - Ovidiu C Andronesi
- From the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 149 Thirteenth St, Ste 2301, Charlestown, MA 02129 (M.B., F.W., Z.D., O.C.A.); Harvard Medical School, Boston, Mass (M.B., F.W., Z.D., J.D., E.J.U., T.T.B., D.P.C., O.C.A.); Department of Neurology, Papas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Mass (J.D.); Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Mass (E.J.U.); Department of Neurology, Brigham and Women's Hospital, Boston, Mass (T.T.B.); Dana Farber Cancer Institute, Boston, Mass (T.T.B.); and Department of Neurosurgery, Massachusetts General Hospital, Boston, Mass (D.P.C.)
| |
Collapse
|
4
|
Wan M, Zhang Y, Li J, Qian Z, Gao F, Yang Y, Li W. Optical-resolution photoacoustic microelastography system for elasticity mapping: Phantom study and practical application. JOURNAL OF BIOPHOTONICS 2024; 17:e202400032. [PMID: 38894573 DOI: 10.1002/jbio.202400032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/26/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024]
Abstract
Elastography is a noninvasive technique for characterizing the mechanical properties of biological tissues. Conventional methods have limitations in resolution and sensitivity, hindering disease detection in clinical diagnostics. To address these issues, this study developed an optical-resolution photoacoustic microelastography (OR-PAME) system. Using an agar tissue phantom with varying agar concentrations and contrast agents, PAME evaluated elasticity distribution under compression in both lateral and axial dimensions. It indirectly measured elastic properties by correlating photoacoustic responses, temporal lags, and induced displacement. We also applied the system to the study of the distribution of elastic characteristics of the liver tissue after ablation, which confirmed the potential of OR-PAME in the study of elastic characteristics. Quantitative analysis showed greater lateral displacement in regions with reduced agar concentrations, indicating decreased stiffness. PAME also detected vertical displacement along the axial plane, validating its efficacy in elastographic imaging. By improving resolution and penetration, PAME provides superior visualization of elasticity distribution. Its methodology correlates microstructural alterations with tissue biomechanics, holding potential implications in medical diagnostics.
Collapse
Affiliation(s)
- Min Wan
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| | - Yameng Zhang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
- Department of Computer Engineering, Nanjing Institute of Technology, Nanjing, Jiangsu, China
| | - Jiani Li
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| | - Zhiyu Qian
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| | - Fan Gao
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| | - Yamin Yang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| | - Weitao Li
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Wang C, Zhang Z, Dou Y, Liu Y, Chen B, Liu Q, Wang S. Development of clinical and magnetic resonance imaging-based radiomics nomograms for the differentiation of nodular fasciitis from soft tissue sarcoma. Acta Radiol 2023; 64:2578-2589. [PMID: 37593946 DOI: 10.1177/02841851231188473] [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: 08/19/2023]
Abstract
BACKGROUND Accurate differentiation of nodular fasciitis (NF) from soft tissue sarcoma (STS) before surgery is essential for the subsequent diagnosis and treatment of patients. PURPOSE To develop and evaluate radiomics nomograms based on clinical factors and magnetic resonance imaging (MRI) for the preoperative differentiation of NF from STS. MATERIAL AND METHODS This retrospective study analyzed the MRI data of 27 patients with pathologically diagnosed NF and 58 patients with STS who were randomly divided into training (n = 62) and validation (n = 23) groups. Univariate and multivariate analyses were performed to identify the clinical factors and semantic features of MRI. Radiomics analysis was applied to fat-suppressed T1-weighted (T1W-FS) images, fat-suppressed T2-weighted (T2W-FS) images, and contrast-enhanced T1-weighted (CE-T1W) images. The radiomics nomograms incorporating the radiomics signatures, clinical factors, and semantic features of MRI were developed. ROC curves and AUCs were carried out to compare the performance of the clinical factors, radiomics signatures, and clinical radiomics nomograms. RESULTS Tumor location, size, heterogeneous signal intensity on T2W-FS imaging, heterogeneous signal intensity on CE-T1W imaging, margin definitions on CE-T1W imaging, and septa were independent predictors for differentiating NF from STS (P < 0.05). The performance of the radiomics signatures based on T2W-FS imaging (AUC = 0.961) and CE-T1W imaging (AUC = 0.938) was better than that based on T1W-FS imaging (AUC = 0.833). The radiomics nomograms had AUCs of 0.949, which demonstrated good clinical utility and calibration. CONCLUSION The non-invasive clinical radiomics nomograms exhibited good performance in the differentiation of NF from STS, and they have clinical application in the preoperative diagnosis of diseases.
Collapse
Affiliation(s)
- Chunjie Wang
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Zhengyang Zhang
- Department of Radiology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, PR China
| | - Yanping Dou
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Yajie Liu
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Bo Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, PR China
| | - Qing Liu
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Shaowu Wang
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, PR China
| |
Collapse
|
6
|
Erdem F, Tamsel İ, Demirpolat G. The use of radiomics and machine learning for the differentiation of chondrosarcoma from enchondroma. JOURNAL OF CLINICAL ULTRASOUND : JCU 2023. [PMID: 37009697 DOI: 10.1002/jcu.23461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/18/2023] [Accepted: 03/25/2023] [Indexed: 06/19/2023]
Abstract
PURPOSE To construct and compare machine learning models for differentiating chondrosarcoma from enchondroma using radiomic features from T1 and fat suppressed Proton density (PD) magnetic resonance imaging (MRI). METHODS Eighty-eight patients (57 with enchondroma, 31 with chondrosarcoma) were retrospectively included. Histogram matching and N4ITK MRI bias correction filters were applied. An experienced musculoskeletal radiologist and a senior resident in radiology performed manual segmentation. Voxel sizes were resampled. Laplacian of Gaussian filter and wavelet-based features were used. One thousand eight hundred eighty-eight features were obtained for each patient, with 944 from T1 and 944 from PD images. Sixty-four unstable features were removed. Seven machine learning models were used for classification. RESULTS Classification with all features showed neural network was the best model for both readers' datasets with area under the curve (AUC), classification accuracy (CA), and F1 score of 0.979, 0.984; 0.920, 0.932; and 0.889, 0.903, respectively. Four features, including one common to both readers, were selected using fast correlation based filter. The best performing models with selected features were gradient boosting for Fatih Erdem's dataset and neural network for Gülen Demirpolat's dataset with AUC, CA, and F1 score of 0.990, 0.979; 0.943, 0.955; 0.921, 0.933, respectively. Neural Network was the second-best model for FE's dataset based on AUC (0.984). CONCLUSION Using pathology as a gold standard, this study defined and compared seven well-performing models to distinguish enchondromas from chondrosarcomas and provided radiomic feature stability and reproducibility among the readers.
Collapse
Affiliation(s)
- Fatih Erdem
- Department of Radiology, Balikesir University Hospital, Paşaköy, Bigadiç yolu üzeri, 10145 Balıkesir Merkez, Altıeylül, Balıkesir, Turkey
| | - İpek Tamsel
- Department of Radiology, Ege University Hospital, 35100, Bornova, Izmir, Turkey
| | - Gülen Demirpolat
- Department of Radiology, Balikesir University Hospital, Paşaköy, Bigadiç yolu üzeri, 10145 Balıkesir Merkez, Altıeylül, Balıkesir, Turkey
| |
Collapse
|
7
|
Slavkova KP, Patel SH, Cacini Z, Kazerouni AS, Gardner AL, Yankeelov TE, Hormuth DA. Mathematical modelling of the dynamics of image-informed tumor habitats in a murine model of glioma. Sci Rep 2023; 13:2916. [PMID: 36804605 PMCID: PMC9941120 DOI: 10.1038/s41598-023-30010-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Tumors exhibit high molecular, phenotypic, and physiological heterogeneity. In this effort, we employ quantitative magnetic resonance imaging (MRI) data to capture this heterogeneity through imaging-based subregions or "habitats" in a murine model of glioma. We then demonstrate the ability to model and predict the growth of the habitats using coupled ordinary differential equations (ODEs) in the presence and absence of radiotherapy. Female Wistar rats (N = 21) were inoculated intracranially with 106 C6 glioma cells, a subset of which received 20 Gy (N = 5) or 40 Gy (N = 8) of radiation. All rats underwent diffusion-weighted and dynamic contrast-enhanced MRI at up to seven time points. All MRI data at each visit were subsequently clustered using k-means to identify physiological tumor habitats. A family of four models consisting of three coupled ODEs were developed and calibrated to the habitat time series of control and treated rats and evaluated for predictive capability. The Akaike Information Criterion was used for model selection, and the normalized sum-of-square-error (SSE) was used to evaluate goodness-of-fit in model calibration and prediction. Three tumor habitats with significantly different imaging data characteristics (p < 0.05) were identified: high-vascularity high-cellularity, low-vascularity high-cellularity, and low-vascularity low-cellularity. Model selection resulted in a five-parameter model whose predictions of habitat dynamics yielded SSEs that were similar to the SSEs from the calibrated model. It is thus feasible to mathematically describe habitat dynamics in a preclinical model of glioma using biology-based ODEs, showing promise for forecasting heterogeneous tumor behavior.
Collapse
Affiliation(s)
- Kalina P. Slavkova
- grid.89336.370000 0004 1936 9924Department of Physics, The University of Texas at Austin, Austin, TX USA
| | - Sahil H. Patel
- grid.67105.350000 0001 2164 3847 Department of Computer Science, Case Western Reserve University, Cleveland, OH USA
| | - Zachary Cacini
- grid.35403.310000 0004 1936 9991 Department of Bioengineering, University of Illinois, Urbana-Champaign, IL USA
| | - Anum S. Kazerouni
- grid.34477.330000000122986657Department of Radiology, The University of Washington, Seattle, WA USA
| | - Andrea L. Gardner
- grid.89336.370000 0004 1936 9924Department of Biomedical Engineering, The University of Texas at Austin, Austin, USA
| | - Thomas E. Yankeelov
- grid.89336.370000 0004 1936 9924Department of Biomedical Engineering, The University of Texas at Austin, Austin, USA ,grid.89336.370000 0004 1936 9924Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX USA ,grid.89336.370000 0004 1936 9924Department of Oncology, The University of Texas at Austin, Austin, TX USA ,grid.89336.370000 0004 1936 9924The Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 E 24th Street, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX USA ,grid.240145.60000 0001 2291 4776Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - David A. Hormuth
- grid.89336.370000 0004 1936 9924The Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 E 24th Street, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX USA
| |
Collapse
|
8
|
Djuričić GJ, Ahammer H, Rajković S, Kovač JD, Milošević Z, Sopta JP, Radulovic M. Directionally Sensitive Fractal Radiomics Compatible With Irregularly Shaped Magnetic Resonance Tumor Regions of Interest: Association With Osteosarcoma Chemoresistance. J Magn Reson Imaging 2023; 57:248-258. [PMID: 35561019 DOI: 10.1002/jmri.28232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Computational analysis of routinely acquired MRI has potential to improve the tumor chemoresistance prediction and to provide decision support in precision medicine, which may extend patient survival. Most radiomic analytical methods are compatible only with rectangular regions of interest (ROIs) and irregular tumor shape is therefore an important limitation. Furthermore, the currently used analytical methods are not directionally sensitive. PURPOSE To implement a tumor analysis that is directionally sensitive and compatible with irregularly shaped ROIs. STUDY TYPE Retrospective. SUBJECTS A total of 54 patients with histopathologic diagnosis of primary osteosarcoma on tubular long bones and with prechemotherapy MRI. FIELD STRENGTH/SEQUENCE A 1.5 T, T2-weighted-short-tau-inversion-recovery-fast-spin-echo. ASSESSMENT A model to explore associations with osteosarcoma chemo-responsiveness included MRI data obtained before OsteoSa MAP neoadjuvant cytotoxic chemotherapy. Osteosarcoma morphology was analyzed in the MRI data by calculation of the nondirectional two-dimensional (2D) and directional and nondirectional one-dimensional (1D) Higuchi dimensions (Dh). MAP chemotherapy response was assessed by histopathological necrosis. STATISTICAL TESTS The area under the receiver operating characteristic (ROC) curve (AUC) evaluated the association of the calculated features with the actual chemoresponsiveness, using tumor histopathological necrosis (95%) as the endpoint. Least absolute shrinkage and selection operator (LASSO) machine learning and multivariable regression were used for feature selection. Significance was set at <0.05. RESULTS The nondirectional 1D Dh reached an AUC of 0.88 in association with the 95% tumor necrosis, while the directional 1D analysis along 180 radial lines significantly improved this association according to the Hanley/McNeil test, reaching an AUC of 0.95. The model defined by variable selection using LASSO reached an AUC of 0.98. The directional analysis showed an optimal predictive range between 90° and 97° and revealed structural osteosarcoma anisotropy manifested by its directionally dependent textural properties. DATA CONCLUSION Directionally sensitive radiomics had superior predictive performance in comparison to the standard nondirectional image analysis algorithms with AUCs reaching 0.95 and full compatibility with irregularly shaped ROIs. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Goran J Djuričić
- Faculty of Medicine, Department of Radiology, University of Belgrade, University Children's Hospital, Belgrade, Serbia
| | - Helmut Ahammer
- Division of Biophysics, GSRC, Medical University of Graz, Graz, Austria
| | - Stanislav Rajković
- Faculty of Medicine, University of Belgrade, Institute for Orthopaedics "Banjica", Belgrade, Serbia
| | - Jelena Djokić Kovač
- University of Belgrade, Faculty of Medicine, Center for Radiology, Clinical Center of Serbia, Belgrade, Serbia
| | - Zorica Milošević
- University of Belgrade, Faculty of Medicine, Institute for Oncology & Radiology of Serbia, Clinic for Radiation Oncology and Radiology, Belgrade, Serbia
| | - Jelena P Sopta
- University of Belgrade, Faculty of Medicine, Institute of Pathology, Belgrade, Serbia
| | - Marko Radulovic
- Department of Experimental Oncology, Institute for Oncology & Radiology of Serbia, Belgrade, Serbia
| |
Collapse
|
9
|
Lu Y, Massicano AVF, Gallegos CA, Heinzman KA, Parish SW, Warram JM, Sorace AG. Evaluating the Accuracy of FUCCI Cell Cycle In Vivo Fluorescent Imaging to Assess Tumor Proliferation in Preclinical Oncology Models. Mol Imaging Biol 2022; 24:898-908. [PMID: 35650411 DOI: 10.1007/s11307-022-01739-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE The primary goal of this study is to evaluate the accuracy of the fluorescence ubiquitination cell cycle indicator (FUCCI) system with fluorescence in vivo imaging compared to 3'-deoxy-3'-[18F]fluorothymidine ([18F]-FLT) positron emission tomography (PET)/computed tomography (CT) and biological validation through histology. Imaging with [18F]-FLT PET/CT can be used to noninvasively assess cancer cell proliferation and has been utilized in both preclinical and clinical studies. However, a cost-effective and straightforward method for in vivo, cell cycle targeted cancer drug screening is needed prior to moving towards translational imaging methods such as PET/CT. PROCEDURES In this study, fluorescent MDA-MB-231-FUCCI tumor growth was monitored weekly with caliper measurements and fluorescent imaging. Seven weeks post-injection, [18F]-FLT PET/CT was performed with a preclinical PET/CT, and tumors samples were harvested for histological analysis. RESULTS RFP fluorescent signal significantly correlated with tumor volume (r = 0.8153, p < 0.0001). Cell proliferation measured by GFP fluorescent imaging was correlated with tumor growth rate (r = 0.6497, p < 0.001). Also, GFP+ cells and [18F]-FLT regions of high uptake were both spatially located in the tumor borders, indicating that the FUCCI-IVIS method may provide an accurate assessment of tumor heterogeneity of cell proliferation. The quantification of total GFP signal was correlated with the sum of tumor [18F]-FLT standard uptake value (SUV) (r = 0.5361, p = 0.0724). Finally, histological analysis confirmed viable cells in the tumor and the correlation of GFP + and Ki67 + cells (r = 0.6368, p = 0.0477). CONCLUSION Fluorescent imaging of the cell cycle provides a noninvasive accurate depiction of tumor progression and response to therapy, which may benefit in vivo testing of novel cancer therapeutics that target the cell cycle.
Collapse
Affiliation(s)
- Yun Lu
- Department of Radiology, University of Alabama at Birmingham, Volker Hall G082, 1670 University Boulevard, Birmingham, AL, 35233, USA
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Adriana V F Massicano
- Department of Radiology, University of Alabama at Birmingham, Volker Hall G082, 1670 University Boulevard, Birmingham, AL, 35233, USA
| | - Carlos A Gallegos
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Katherine A Heinzman
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Sean W Parish
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Jason M Warram
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Volker Hall G082, 1670 University Boulevard, Birmingham, AL, 35233, USA.
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
10
|
Zhang Z, Li X, Sun H. Development of machine learning models integrating PET/CT radiomic and immunohistochemical pathomic features for treatment strategy choice of cervical cancer with negative pelvic lymph node by mediating COX-2 expression. Front Physiol 2022; 13:994304. [PMID: 36311222 PMCID: PMC9614332 DOI: 10.3389/fphys.2022.994304] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022] Open
Abstract
Objectives: We aimed to establish machine learning models based on texture analysis predicting pelvic lymph node metastasis (PLNM) and expression of cyclooxygenase-2 (COX-2) in cervical cancer with PET/CT negative pelvic lymph node (PLN). Methods: Eight hundred and thirty-seven texture features were extracted from PET/CT images of 148 early-stage cervical cancer patients with negative PLN. The machine learning models were established by logistic regression from selected features and evaluated by the area under the curve (AUC). The correlation of selected PET/CT texture features predicting PLNM or COX-2 expression and the corresponding immunohistochemical (IHC) texture features was analyzed by the Spearman test. Results: Fourteen texture features were reserved to calculate the Rad-score for PLNM and COX-2. The PLNM model predicting PLNM showed good prediction accuracy in the training and testing dataset (AUC = 0.817, p < 0.001; AUC = 0.786, p < 0.001, respectively). The COX-2 model also behaved well for predicting COX-2 expression levels in the training and testing dataset (AUC = 0.814, p < 0.001; AUC = 0.748, p = 0.001). The wavelet-LHH-GLCM ClusterShade of the PET image selected to predict PLNM was slightly correlated with the corresponding feature of the IHC image (r = −0.165, p < 0.05). There was a weak correlation of wavelet-LLL-GLRLM LongRunEmphasis of the PET image selected to predict COX-2 correlated with the corresponding feature of the IHC image (r = 0.238, p < 0.05). The correlation between PET image selected to predict COX-2 and the corresponding feature of the IHC image based on wavelet-LLL-GLRLM LongRunEmphasis is considered weak positive (r = 0.238, p=<0.05). Conclusion: This study underlined the significant application of the machine learning models based on PET/CT texture analysis for predicting PLNM and COX-2 expression, which could be a novel tool to assist the clinical management of cervical cancer with negative PLN on PET/CT images.
Collapse
|
11
|
Furia L, Pelicci S, Perillo F, Bolognesi MM, Pelicci PG, Facciotti F, Cattoretti G, Faretta M. Automated multimodal fluorescence microscopy for hyperplex spatial-proteomics: Coupling microfluidic-based immunofluorescence to high resolution, high sensitivity, three-dimensional analysis of histological slides. Front Oncol 2022; 12:960734. [PMCID: PMC9606676 DOI: 10.3389/fonc.2022.960734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
In situ multiplexing analysis and in situ transcriptomics are now providing revolutionary tools to achieve the comprehension of the molecular basis of cancer and to progress towards personalized medicine to fight the disease. The complexity of these tasks requires a continuous interplay among different technologies during all the phases of the experimental procedures. New tools are thus needed and their characterization in terms of performances and limits is mandatory to reach the best resolution and sensitivity. We propose here a new experimental pipeline to obtain an optimized costs-to-benefits ratio thanks to the alternate employment of automated and manual procedures during all the phases of a multiplexing experiment from sample preparation to image collection and analysis. A comparison between ultra-fast and automated immunofluorescence staining and standard staining protocols has been carried out to compare the performances in terms of antigen saturation, background, signal-to-noise ratio and total duration. We then developed specific computational tools to collect data by automated analysis-driven fluorescence microscopy. Computer assisted selection of targeted areas with variable magnification and resolution allows employing confocal microscopy for a 3D high resolution analysis. Spatial resolution and sensitivity were thus maximized in a framework where the amount of stored data and the total requested time for the procedure were optimized and reduced with respect to a standard experimental approach.
Collapse
Affiliation(s)
- Laura Furia
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Simone Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Perillo
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Giorgio Cattoretti
- Department of Medicine and Surgery, Università di Milano-Bicocca, Monza, Italy
| | - Mario Faretta
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- *Correspondence: Mario Faretta,
| |
Collapse
|
12
|
Chang R, Qi S, Zuo Y, Yue Y, Zhang X, Guan Y, Qian W. Predicting chemotherapy response in non-small-cell lung cancer via computed tomography radiomic features: Peritumoral, intratumoral, or combined? Front Oncol 2022; 12:915835. [PMID: 36003781 PMCID: PMC9393703 DOI: 10.3389/fonc.2022.915835] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/18/2022] [Indexed: 11/15/2022] Open
Abstract
Purpose This study aims to evaluate the ability of peritumoral, intratumoral, or combined computed tomography (CT) radiomic features to predict chemotherapy response in non-small cell lung cancer (NSCLC). Methods After excluding subjects with incomplete data or other types of treatments, 272 (Dataset 1) and 43 (Dataset 2, external validation) NSCLC patients who were only treated with chemotherapy as the first-line treatment were enrolled between 2015 and 2019. All patients were divided into response and nonresponse based on the response evaluation criteria in solid tumors, version 1.1. By using 3D slicer and morphological operations in python, the intra- and peritumoral regions of lung tumors were segmented from pre-treatment CT images (unenhanced) and confirmed by two experienced radiologists. Then radiomic features (the first order, texture, shape, et al.) were extracted from the above regions of interest. The models were trained and tested in Dataset 1 and further validated in Dataset 2. The performance of models was compared using the area under curve (AUC), confusion matrix, accuracy, precision, recall, and F1-score. Results The radiomic model using features from the peritumoral region of 0–3 mm outperformed that using features from 3–6, 6–9, 9–12 mm peritumoral region, and intratumoral region (AUC: 0.95 versus 0.87, 0.86, 0.85, and 0.88). By the fusion of features from 0–3 and 3–6 mm peritumoral regions, the logistic regression model achieved the best performance, with an AUC of 0.97. This model achieved an AUC of 0.85 in the external cohort. Moreover, among the 20 selected features, seven features differed significantly between the two groups (p < 0.05). Conclusions CT radiomic features from both the peri- and intratumoral regions can predict chemotherapy response in NSCLC using machine learning models. Combined features from two peritumoral regions yielded better predictions.
Collapse
Affiliation(s)
- Runsheng Chang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Shouliang Qi
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, China
- *Correspondence: Shouliang Qi,
| | - Yifan Zuo
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Yong Yue
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoye Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yubao Guan
- Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Qian
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| |
Collapse
|
13
|
MRI Radiogenomics in Precision Oncology: New Diagnosis and Treatment Method. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:2703350. [PMID: 35845886 PMCID: PMC9282990 DOI: 10.1155/2022/2703350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/04/2022] [Accepted: 05/25/2022] [Indexed: 11/21/2022]
Abstract
Precision medicine for cancer affords a new way for the most accurate and effective treatment to each individual cancer. Given the high time-evolving intertumor and intratumor heterogeneity features of personal medicine, there are still several obstacles hindering its diagnosis and treatment in clinical practice regardless of extensive exploration on it over the past years. This paper is to investigate radiogenomics methods in the literature for precision medicine for cancer focusing on the heterogeneity analysis of tumors. Based on integrative analysis of multimodal (parametric) imaging and molecular data in bulk tumors, a comprehensive analysis and discussion involving the characterization of tumor heterogeneity in imaging and molecular expression are conducted. These investigations are intended to (i) fully excavate the multidimensional spatial, temporal, and semantic related information regarding high-dimensional breast magnetic resonance imaging data, with integration of the highly specific structured data of genomics and combination of the diagnosis and cognitive process of doctors, and (ii) establish a radiogenomics data representation model based on multidimensional consistency analysis with multilevel spatial-temporal correlations.
Collapse
|
14
|
Speckter H, Radulovic M, Trivodaliev K, Vranes V, Joaquin J, Hernandez W, Mota A, Bido J, Hernandez G, Rivera D, Suazo L, Valenzuela S, Stoeter P. MRI radiomics in the prediction of the volumetric response in meningiomas after gamma knife radiosurgery. J Neurooncol 2022; 159:281-291. [PMID: 35715668 DOI: 10.1007/s11060-022-04063-y] [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: 05/08/2022] [Accepted: 06/07/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE This report presents the first investigation of the radiomics value in predicting the meningioma volumetric response to gamma knife radiosurgery (GKRS). METHODS The retrospective study included 93 meningioma patients imaged by three Tesla MRI. Tumor morphology was quantified by calculating 337 shape, first- and second-order radiomic features from MRI obtained before GKRS. Analysis was performed on original 3D MR images and after their laplacian of gaussian (LoG), logarithm and exponential filtering. The prediction performance was evaluated by Pearson correlation, linear regression and ROC analysis, with meningioma volume change per month as the outcome. RESULTS Sixty calculated features significantly correlated with the outcome. The feature selection based on LASSO and multivariate regression started from all available 337 radiomic and 12 non-radiomic features. It selected LoG-sigma-1-0-mm-3D_firstorder_InterquartileRange and logarithm_ngtdm_Busyness as the predictively most robust and non-redundant features. The radiomic score based on these two features produced an AUC = 0.81. Adding the non-radiomic karnofsky performance status (KPS) to the score has increased the AUC to 0.88. Low values of the radiomic score defined a homogeneous subgroup of 50 patients with consistent absence (0%) of tumor progression. CONCLUSION This is the first report of a strong association between MRI radiomic features and volumetric meningioma response to radiosurgery. The clinical importance of the early and reliable prediction of meningioma responsiveness to radiosurgery is based on its potential to aid individualized therapy decision making.
Collapse
Affiliation(s)
- Herwin Speckter
- Centro Gamma Knife Dominicano and Department of Radiology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic.
| | - Marko Radulovic
- Department of Experimental Oncology, Institute for Oncology & Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia
| | | | - Velicko Vranes
- Instituto Tecnológico de Santo Domingo (INTEC), Santo Domingo, Dominican Republic
| | - Johanna Joaquin
- Centro Gamma Knife Dominicano and Department of Radiology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Wenceslao Hernandez
- Centro Gamma Knife Dominicano and Department of Radiology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Angel Mota
- Centro Gamma Knife Dominicano and Department of Radiology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Jose Bido
- Centro Gamma Knife Dominicano and Department of Radiology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Giancarlo Hernandez
- Centro Gamma Knife Dominicano and Department of Radiology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Diones Rivera
- Centro Gamma Knife Dominicano and Department of Radiology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Luis Suazo
- Centro Gamma Knife Dominicano and Department of Radiology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Santiago Valenzuela
- Centro Gamma Knife Dominicano and Department of Radiology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Peter Stoeter
- Centro Gamma Knife Dominicano and Department of Radiology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| |
Collapse
|
15
|
Xi Y, Ge X, Ji H, Wang L, Duan S, Chen H, Wang M, Hu H, Jiang F, Ding Z. Prediction of Response to Induction Chemotherapy Plus Concurrent Chemoradiotherapy for Nasopharyngeal Carcinoma Based on MRI Radiomics and Delta Radiomics: A Two-Center Retrospective Study. Front Oncol 2022; 12:824509. [PMID: 35530350 PMCID: PMC9074388 DOI: 10.3389/fonc.2022.824509] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/23/2022] [Indexed: 12/03/2022] Open
Abstract
Objective We aimed to establish an MRI radiomics model and a Delta radiomics model to predict tumor retraction after induction chemotherapy (IC) combined with concurrent chemoradiotherapy (CCRT) for primary nasopharyngeal carcinoma (NPC) in non-endemic areas and to validate its efficacy. Methods A total of 272 patients (155 in the training set, 66 in the internal validation set, and 51 in the external validation set) with biopsy pathologically confirmed primary NPC who were screened for pretreatment MRI were retrospectively collected. The NPC tumor was delineated as a region of interest in the two sequenced images of MRI before treatment and after IC, followed by radiomics feature extraction. With the use of maximum relevance minimum redundancy (mRMR) and least absolute shrinkage and selection operator (LASSO) algorithms, logistic regression was performed to establish pretreatment MRI radiomics and pre- and post-IC Delta radiomics models. The optimal Youden’s index was taken; the receiver operating characteristic (ROC) curve, calibration curve, and decision curve were drawn to evaluate the predictive efficacy of different models. Results Seven optimal feature subsets were selected from the pretreatment MRI radiomics model, and twelve optimal subsets were selected from the Delta radiomics model. The area under the ROC curve, accuracy, sensitivity, specificity, negative predictive value (NPV), and positive predictive value (PPV) of the MRI radiomics model were 0.865, 0.827, 0.837, 0.813, 0.776, and 0.865, respectively; the corresponding indicators of the Delta radiomics model were 0.941, 0.883, 0.793, 0.968, 0.833, and 0.958, respectively. Conclusion The pretreatment MRI radiomics model and pre- and post-IC Delta radiomics models could predict the IC-CCRT response of NPC in non-epidemic areas.
Collapse
Affiliation(s)
- Yuzhen Xi
- Department of Radiology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, China
- Department of Radiology, 903rd Hospital of PLA, Hangzhou, China
| | - Xiuhong Ge
- Department of Radiology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiming Ji
- Department of Radiology, Liangzhu Hospital, Hangzhou, China
| | - Luoyu Wang
- Department of Radiology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, China
| | - Shaofeng Duan
- GE Healthcare, Precision Health Institution, Shanghai, China
| | - Haonan Chen
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengze Wang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital Affiliated to Medical College Zhejiang University, Hangzhou, China
| | - Feng Jiang
- Department of Head and Neck Radiotherapy, Zhejiang Cancer Hospital/Zhejiang Province Key Laboratory of Radiation Oncology, Hangzhou, China
- *Correspondence: Feng Jiang, ; Zhongxiang Ding,
| | - Zhongxiang Ding
- Department of Radiology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Feng Jiang, ; Zhongxiang Ding,
| |
Collapse
|
16
|
Wang X, Li X, Chen H, Peng Y, Li Y. Pulmonary MRI Radiomics and Machine Learning: Effect of Intralesional Heterogeneity on Classification of Lesion. Acad Radiol 2022; 29 Suppl 2:S73-S81. [PMID: 33495072 DOI: 10.1016/j.acra.2020.12.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/09/2020] [Accepted: 12/30/2020] [Indexed: 12/20/2022]
Abstract
RATIONALE AND OBJECTIVES To investigate the effect of intralesional heterogeneity on differentiating benign and malignant pulmonary lesions, quantitative magnetic resonance imaging (MRI) radiomics, and machine learning methods were adopted. MATERIALS AND METHODS A total of 176 patients with multiparametric MRI were involved in this exploratory study. To investigate the effect of intralesional heterogeneity on lesion classification, a radiomics model called tumor heterogeneity model was developed and compared to the conventional radiomics model based on the entire tumor. In tumor heterogeneity model, each lesion was divided into five sublesions depending on the spatial location through clustering algorithm. From the five sublesions in multi-parametric MRI sequences, 1100 radiomics features were extracted. The recursive feature elimination method was employed to select features and support vector machine classifier was used to distinguish benign and malignant lesion. The performance of classification was evaluated with the receiver operating characteristic curve and the area under the curve (AUC) was the figure of merit. The 3-fold cross-validation (CV) with and without nesting was used to validate the model, respectively. RESULTS The tumor heterogeneity model (AUC = 0.74 ± 0.04 and 0.90 ± 0.03, CV with and without nesting, respectively) outperforms conventional model (AUC = 0.68 ± 0.04 and 0.87 ± 0.03). The difference between the two models is statistically significant (p = 0.03) for lesions greater than 18.80 cm3. CONCLUSION Intralesional heterogeneity influences the classification of pulmonary lesions. The tumor heterogeneity model tends to perform better than conventional radiomics model.
Collapse
Affiliation(s)
- Xinhui Wang
- School of Electronic and Information Engineering, Beijing Jiaotong University, Shangyuan Village No 3 in Haidian, Beijing, China
| | - Xinchun Li
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Houjin Chen
- School of Electronic and Information Engineering, Beijing Jiaotong University, Shangyuan Village No 3 in Haidian, Beijing, China.
| | - Yahui Peng
- School of Electronic and Information Engineering, Beijing Jiaotong University, Shangyuan Village No 3 in Haidian, Beijing, China
| | - Yanfeng Li
- School of Electronic and Information Engineering, Beijing Jiaotong University, Shangyuan Village No 3 in Haidian, Beijing, China
| |
Collapse
|
17
|
Jacquemin V, Antoine M, Dom G, Detours V, Maenhaut C, Dumont JE. Dynamic Cancer Cell Heterogeneity: Diagnostic and Therapeutic Implications. Cancers (Basel) 2022; 14:280. [PMID: 35053446 PMCID: PMC8773841 DOI: 10.3390/cancers14020280] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Though heterogeneity of cancers is recognized and has been much discussed in recent years, the concept often remains overlooked in different routine examinations. Indeed, in clinical or biological articles, reviews, and textbooks, cancers and cancer cells are generally presented as evolving distinct entities rather than as an independent heterogeneous cooperative cell population with its self-oriented biology. There are, therefore, conceptual gaps which can mislead the interpretations/diagnostic and therapeutic approaches. In this short review, we wish to summarize and discuss various aspects of this dynamic evolving heterogeneity and its biological, pathological, clinical, diagnostic, and therapeutic implications, using thyroid carcinoma as an illustrative example.
Collapse
Affiliation(s)
- Valerie Jacquemin
- Correspondence: (V.J.); (J.E.D.); Tel.: +32-2-555-32-26 (V.J.); +32-2-555-41-34 (J.E.D.)
| | | | | | | | | | - Jacques E. Dumont
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, 1070 Brussels, Belgium; (M.A.); (G.D.); (V.D.); (C.M.)
| |
Collapse
|
18
|
Girot C, Volk A, Walczak C, Lassau N, Pitre-Champagnat S. New method for quantification of intratumoral heterogeneity: a feasibility study on K trans maps from preclinical DCE-MRI. MAGMA (NEW YORK, N.Y.) 2021; 34:845-857. [PMID: 34091826 DOI: 10.1007/s10334-021-00930-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/31/2022]
Abstract
OBJECT To develop new imaging biomarkers of therapeutic efficacy through the quantification of intratumoral microvascular heterogeneity. MATERIALS AND METHODS The described method was a combination of non-supervised clustering and extraction of intratumoral complexity features (ICF): number of non-connected objects, volume fraction. It was applied to a set of 3D DCE-MRI Ktrans maps acquired previously on tumor bearing mice prior to and on day 4 of anti-angiogenic treatment. Evolutions of ICF were compared to conventional summary statistics (CSS) and to heterogeneity related whole tumor texture features (TF) on treated (n = 9) and control (n = 6) mice. RESULTS Computed optimal number of clusters per tumor was 4. Several intratumoral features extracted from the clusters were able to monitor a therapy effect. Whereas no feature significantly changed for the control group, 6 features significantly changed for the treated group (4 ICF, 2 CSS). Among these, 5 also significantly differentiated the two groups (3 ICF, 2 CSS). TF failed in demonstrating differences within and between the two groups. DISCUSSION ICF are potential imaging biomarkers for anti-angiogenic therapy assessment. The presented method may be expected to have advantages with respect to texture analysis-based methods regarding interpretability of results and setup of standardized image analysis protocols.
Collapse
Affiliation(s)
- Charly Girot
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France.
| | - Andreas Volk
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Christine Walczak
- Institut Curie, Inserm, Université Paris-Saclay, CNRS, 91405, Orsay, France
| | - Nathalie Lassau
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France.,Département de Radiologie, Gustave Roussy, 94805, Villejuif, France
| | - Stéphanie Pitre-Champagnat
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| |
Collapse
|
19
|
Li X, Xu C, Yu Y, Guo Y, Sun H. Prediction of lymphovascular space invasion using a combination of tenascin-C, cox-2, and PET/CT radiomics in patients with early-stage cervical squamous cell carcinoma. BMC Cancer 2021; 21:866. [PMID: 34320931 PMCID: PMC8317359 DOI: 10.1186/s12885-021-08596-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 07/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lymphovascular space invasion is an independent prognostic factor in early-stage cervical cancer. However, there is a lack of non-invasive methods to detect lymphovascular space invasion. Some researchers found that Tenascin-C and Cyclooxygenase-2 was correlated with lymphovascular space invasion. Radiomics has been studied as an emerging tool for distinguishing tumor pathology stage, evaluating treatment response, and predicting prognosis. This study aimed to establish a machine learning model that combines radiomics based on PET imaging with tenascin-C (TNC) and cyclooxygenase-2 (COX-2) for predicting lymphovascular space invasion (LVSI) in patients with early-stage cervical cancer. METHODS One hundred and twelve patients with early-stage cervical squamous cell carcinoma who underwent PET/CT examination were retrospectively analyzed. Four hundred one radiomics features based on PET/CT images were extracted and integrated into radiomics score (Rad-score). Immunohistochemical analysis was performed to evaluate TNC and COX-2 expression. Mann-Whitney U test was used to distinguish differences in the Rad-score, TNC, and COX-2 between LVSI and non-LVSI groups. The correlations of characteristics were tested by Spearman analysis. Machine learning models including radiomics model, protein model and combined model were established by logistic regression algorithm and evaluated by ROC curve. Pairwise comparisons of ROC curves were tested by DeLong test. RESULTS The Rad-score of patients with LVSI was significantly higher than those without. A significant correlation was shown between LVSI and Rad-score (r = 0.631, p < 0.001). TNC was correlated to both the Rad-score (r = 0.244, p = 0.024) and COX-2 (r = 0.227, p = 0.036). The radiomics model had the best predictive performance among all models in training and external dataset (AUCs: 0.914, 0.806, respectively, p < 0.001). However, in testing dataset, the combined model had better efficiency for predicting LVSI than other models (AUCs: 0.801 vs. 0.756 and 0.801 vs. 0.631, respectively). CONCLUSION The machine learning model of the combination of PET radiomics with COX-2 and TNC provides a new tool for detecting LVSI in patients with early-stage cervical cancer. In the future, multicentric studies on larger sample of patients will be used to test the model. TRIAL REGISTRATION This is a retrospective study and there is no experimental intervention on human participants. The Ethics Committee has confirmed that retrospectively registered is not required.
Collapse
Affiliation(s)
- Xiaoran Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning China
| | - Chen Xu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning China
| | - Yang Yu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning China
| | - Yan Guo
- GE Healthcare, Shenyang, Liaoning China
| | - Hongzan Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning China
| |
Collapse
|
20
|
Chang R, Qi S, Yue Y, Zhang X, Song J, Qian W. Predictive Radiomic Models for the Chemotherapy Response in Non-Small-Cell Lung Cancer based on Computerized-Tomography Images. Front Oncol 2021; 11:646190. [PMID: 34307127 PMCID: PMC8293296 DOI: 10.3389/fonc.2021.646190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 06/16/2021] [Indexed: 01/10/2023] Open
Abstract
The heterogeneity and complexity of non-small cell lung cancer (NSCLC) tumors mean that NSCLC patients at the same stage can have different chemotherapy prognoses. Accurate predictive models could recognize NSCLC patients likely to respond to chemotherapy so that they can be given personalized and effective treatment. We propose to identify predictive imaging biomarkers from pre-treatment CT images and construct a radiomic model that can predict the chemotherapy response in NSCLC. This single-center cohort study included 280 NSCLC patients who received first-line chemotherapy treatment. Non-contrast CT images were taken before and after the chemotherapy, and clinical information were collected. Based on the Response Evaluation Criteria in Solid Tumors and clinical criteria, the responses were classified into two categories: response (n = 145) and progression (n = 135), then all data were divided into two cohorts: training cohort (224 patients) and independent test cohort (56 patients). In total, 1629 features characterizing the tumor phenotype were extracted from a cube containing the tumor lesion cropped from the pre-chemotherapy CT images. After dimensionality reduction, predictive models of the chemotherapy response of NSCLC with different feature selection methods and different machine-learning classifiers (support vector machine, random forest, and logistic regression) were constructed. For the independent test cohort, the predictive model based on a random-forest classifier with 20 radiomic features achieved the best performance, with an accuracy of 85.7% and an area under the receiver operating characteristic curve of 0.941 (95% confidence interval, 0.898–0.982). Of the 20 selected features, four were first-order statistics of image intensity and the others were texture features. For nine features, there were significant differences between the response and progression groups (p < 0.001). In the response group, three features, indicating heterogeneity, were overrepresented and one feature indicating homogeneity was underrepresented. The proposed radiomic model with pre-chemotherapy CT features can predict the chemotherapy response of patients with non-small cell lung cancer. This radiomic model can help to stratify patients with NSCLC, thereby offering the prospect of better treatment.
Collapse
Affiliation(s)
- Runsheng Chang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Shouliang Qi
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China.,Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, China
| | - Yong Yue
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoye Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiangdian Song
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Wei Qian
- Department of Electrical and Computer Engineering, University of Texas at El Paso, El Paso, TX, United States
| |
Collapse
|
21
|
Pan J, Zhang K, Le H, Jiang Y, Li W, Geng Y, Li S, Hong G. Radiomics Nomograms Based on Non-enhanced MRI and Clinical Risk Factors for the Differentiation of Chondrosarcoma from Enchondroma. J Magn Reson Imaging 2021; 54:1314-1323. [PMID: 33949727 DOI: 10.1002/jmri.27690] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Differentiating chondrosarcoma from enchondroma using conventional MRI remains challenging. An effective method for accurate preoperative diagnosis could affect the management and prognosis of patients. PURPOSE To validate and evaluate radiomics nomograms based on non-enhanced MRI and clinical risk factors for the differentiation of chondrosarcoma from enchondroma. STUDY TYPE Retrospective. POPULATION A total of 103 patients with pathologically confirmed chondrosarcoma (n = 53) and enchondroma (n = 50) were randomly divided into training (n = 68) and validation (n = 35) groups. FIELD STRENGTH/SEQUENCE Axial non-contrast-enhanced T1-weighted images (T1WI) and fat-suppressed T2-weighted images (T2WI-FS) were acquired at 3.0 T. ASSESSMENT Clinical risk factors (sex, age, and tumor location) and diagnosis assessment based on morphologic MRI by three radiologists were recorded. Three radiomics signatures were established based on the T1WI, T2WI-FS, and T1WI + T2WI-FS sequences. Three clinical radiomics nomograms were developed based on the clinical risk factors and three radiomics signatures. STATISTICAL TESTS The area under the receiver operating characteristic curve (AUC) was used to evaluate the performance of radiomics signatures and clinical radiomics nomograms. RESULTS Tumor location was an important clinical risk factor (P < 0.05). The radiomics signature based on T1WI and T1WI + T2WI-FS features performed better than that based on T2WI-FS in the validation group (AUC in the validation group: 0.961, 0.938, and 0.833, respectively; P < 0.05). In the validation group, the three clinical radiomics nomograms (T1WI, T2WI-FS, and T1WI + T2WI-FS) achieved AUCs of 0.938, 0.935, and 0.954, respectively. In all patients, the clinical radiomics nomogram based on T2WI-FS (AUC = 0.967) performed better than that based on T2WI-FS (AUC = 0.901, P < 0.05). DATA CONCLUSION The proposed clinical radiomics nomogram showed promising performance in differentiating chondrosarcoma from enchondroma. LEVEL OF EVIDENCE 4 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jielin Pan
- Department of Radiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China.,Department of Radiology, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Ke Zhang
- Department of Radiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Hongbo Le
- Department of Radiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Yunping Jiang
- Department of Radiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Wenjuan Li
- Department of Radiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Yayuan Geng
- Scientific Research Department, HY Medical Technology, Beijing, China
| | - Shaolin Li
- Department of Radiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Guobin Hong
- Department of Radiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| |
Collapse
|
22
|
Chiloiro G, Rodriguez-Carnero P, Lenkowicz J, Casà C, Masciocchi C, Boldrini L, Cusumano D, Dinapoli N, Meldolesi E, Carano D, Damiani A, Barbaro B, Manfredi R, Valentini V, Gambacorta MA. Delta Radiomics Can Predict Distant Metastasis in Locally Advanced Rectal Cancer: The Challenge to Personalize the Cure. Front Oncol 2020; 10:595012. [PMID: 33344243 PMCID: PMC7744725 DOI: 10.3389/fonc.2020.595012] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/02/2020] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Distant metastases are currently the main cause of treatment failure in locally advanced rectal cancer (LARC) patients. The aim of this research is to investigate a correlation between the variation of radiomics features using pre- and post-neoadjuvant chemoradiation (nCRT) magnetic resonance imaging (MRI) with 2 years distant metastasis (2yDM) rate in LARC patients. METHODS AND MATERIALS Diagnostic pre- and post- nCRT MRI of LARC patients, treated in a single institution from May 2008 to June 2015 with an adequate follow-up time, were retrospectively collected. Gross tumor volumes (GTV) were contoured by an abdominal radiologist and blindly reviewed by a radiation oncologist expert in rectal cancer. The dataset was firstly randomly split into 90% training data, for features selection, and 10% testing data, for the validation. The final set of features after the selection was used to train 15 different classifiers using accuracy as target metric. The models' performance was then assessed on the testing data and the best performing classifier was then selected, maximising the confusion matrix balanced accuracy (BA). RESULTS Data regarding 213 LARC patients (36% female, 64% male) were collected. Overall 2yDM was 17%. A total of 2,606 features extracted from the pre- and post- nCRT GTV were tested and 4 features were selected after features selection process. Among the 15 tested classifiers, logistic regression proved to be the best performing one with a testing set BA, sensitivity and specificity of 78.5%, 71.4% and 85.7%, respectively. CONCLUSIONS This study supports a possible role of delta radiomics in predicting following occurrence of distant metastasis. Further studies including a consistent external validation are needed to confirm these results and allows to translate radiomics model in clinical practice. Future integration with clinical and molecular data will be mandatory to fully personalized treatment and follow-up approaches.
Collapse
Affiliation(s)
- Giuditta Chiloiro
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | - Jacopo Lenkowicz
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Calogero Casà
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carlotta Masciocchi
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luca Boldrini
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Davide Cusumano
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Nicola Dinapoli
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Elisa Meldolesi
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Davide Carano
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Damiani
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Brunella Barbaro
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Riccardo Manfredi
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Vincenzo Valentini
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Antonietta Gambacorta
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
23
|
Liang P, Xu C, Tan F, Li S, Chen M, Hu D, Kamel I, Duan Y, Li Z. Prediction of the World Health Organization Grade of rectal neuroendocrine tumors based on CT histogram analysis. Cancer Med 2020; 10:595-604. [PMID: 33263225 PMCID: PMC7877354 DOI: 10.1002/cam4.3628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/19/2020] [Accepted: 11/10/2020] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES To investigate the diagnostic value of contrast-enhanced computed tomography (CECT) histogram analysis in predicting the World Health Organization (WHO) grade of rectal neuroendocrine tumors (R-NETs). MATERIALS AND METHODS A total of 61 (35 G1, 12 G2, 10 G3, and 4 NECs) patients who underwent preoperative CECT and treated with surgery to be confirmed as R-NETs were included in this study from January 2014 to May 2019. We depicted ROIs and measured the CECT texture parameters (mean, median, 10th, 25th, 75th, 90th percentiles, skewness, kurtosis, and entropy) from arterial phase (AP) and venous phase (VP) images by two radiologists. We calculated intraclass correlation coefficient (ICC) and compared the histogram parameters between low-grade (G1) and higher grade (HG) (G2/G3/NECs) by applying appropriate statistical method. We obtained the optimal parameters to identify G1 from HG using receiver operating characteristic (ROC) curves. RESULTS The capability of AP and VP histogram parameters for differentiating G1 from HG was similar in several histogram parameters (mean, median, 10th, 25th, 75th, and 90th percentiles) (all p < 0.001). Skewness, kurtosis, and entropy on AP images showed no significant differences between G1 and HG (p = 0.853, 0.512, 0.557, respectively). Entropy on VP images was significantly different (p = 0.017) between G1 and HG, however, skewness and kurtosis showed no significant differences (p = 0.654, 0.172, respectively). ROC analysis showed a good predictive performance between G1 and HG, and the 75th (AP) generated the highest area under the curve (AUC = 0.871), followed by the 25th (AP), mean (VP), and median (VP) (AUC = 0.864). Combined the size of tumor and the 75th (AP) generated the highest AUC. CONCLUSIONS CECT histogram parameters, including arterial and venous phases, can be used as excellent indicators for predicting G1 and HG of rectal neuroendocrine tumors, and the size of the tumor is also an important independent predictor.
Collapse
Affiliation(s)
- Ping Liang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chuou Xu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fangqin Tan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shichao Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mingzhen Chen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Daoyu Hu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ihab Kamel
- Russell H. Morgan Department of Radiology and Radiological Science, the Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Yaqi Duan
- Department of Pathology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
24
|
Syed AK, Whisenant JG, Barnes SL, Sorace AG, Yankeelov TE. Multiparametric Analysis of Longitudinal Quantitative MRI data to Identify Distinct Tumor Habitats in Preclinical Models of Breast Cancer. Cancers (Basel) 2020; 12:cancers12061682. [PMID: 32599906 PMCID: PMC7352623 DOI: 10.3390/cancers12061682] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
This study identifies physiological tumor habitats from quantitative magnetic resonance imaging (MRI) data and evaluates their alterations in response to therapy. Two models of breast cancer (BT-474 and MDA-MB-231) were imaged longitudinally with diffusion-weighted MRI and dynamic contrast-enhanced MRI to quantify tumor cellularity and vascularity, respectively, during treatment with trastuzumab or albumin-bound paclitaxel. Tumors were stained for anti-CD31, anti-Ki-67, and H&E. Imaging and histology data were clustered to identify tumor habitats and percent tumor volume (MRI) or area (histology) of each habitat was quantified. Histological habitats were correlated with MRI habitats. Clustering of both the MRI and histology data yielded three clusters: high-vascularity high-cellularity (HV-HC), low-vascularity high-cellularity (LV-HC), and low-vascularity low-cellularity (LV-LC). At day 4, BT-474 tumors treated with trastuzumab showed a decrease in LV-HC (p = 0.03) and increase in HV-HC (p = 0.03) percent tumor volume compared to control. MDA-MB-231 tumors treated with low-dose albumin-bound paclitaxel showed a longitudinal decrease in LV-HC percent tumor volume at day 3 (p = 0.01). Positive correlations were found between histological and imaging-derived habitats: HV-HC (BT-474: p = 0.03), LV-HC (MDA-MB-231: p = 0.04), LV-LC (BT-474: p = 0.04; MDA-MB-231: p < 0.01). Physiologically distinct tumor habitats associated with therapeutic response were identified with MRI and histology data in preclinical models of breast cancer.
Collapse
Affiliation(s)
- Anum K Syed
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jennifer G Whisenant
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Stephanie L Barnes
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Anna G Sorace
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Oncology, The University of Texas at Austin, Austin, TX 78712, USA
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
25
|
[Radiomics models based on non-enhanced MRI can differentiate chondrosarcoma from enchondroma]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:483-490. [PMID: 32895139 PMCID: PMC7225098 DOI: 10.12122/j.issn.1673-4254.2020.04.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To develop and validate radiomics models based on non-enhanced magnetic resonance (MR) imaging for differentiating chondrosarcoma from enchondroma. METHODS We retrospectively evaluated a total of 68 patients (including 27 with chondrosarcoma and 41 with enchondroma), who were randomly divided into training group (n=46) and validation group (n=22). Radiomics features were extracted from T1WI and T2WI-FS sequences of the whole tumor by two radiologists independently and selected by Low Variance, Univariate feature selection, and least absolute shrinkage and selection operator (LASSO). Radiomics models were constructed by multivariate logistic regression analysis based on the features from T1WI and T2WI-FS sequences. The receiver-operating characteristics (ROC) curve and intraclass correlation coefficient (ICC) analyses of the radiomics models and conventional MR imaging were performed to determine their diagnostic accuracy. RESULTS The ICC value for interreader agreement of the radiomics features ranged from 0.779 to 0.923, which indicated good agreement. Ten and 11 features were selected from the T1WI and T2WI-FS sequences to construct radiomics models, respectively. The areas under the curve (AUCs) of T1WI and T2WI-FS models were 0.990 and 0.925 in training group and 0.915 and 0.855 in the validation group, respectively, showing no significant differences between the two sequence-based models (P>0.05). In all the cases, the AUCs of the two radiomics models based on T1WI and T2WI-FS sequences and conventional MR imaging were 0.955, 0.901 and 0.569, respectively, demonstrating a significantly higher diagnostic accuracy of the two sequence-based radiomics models than conventional MR imaging (P<0.01). CONCLUSIONS The radiomics models based on T1WI and T2WI-FS non-enhanced MR imaging can be used for the differentiation of chondrosarcoma from enchondroma.
Collapse
|
26
|
PET/MRI and genetic intrapatient heterogeneity in head and neck cancers. Strahlenther Onkol 2020; 196:542-551. [PMID: 32211941 DOI: 10.1007/s00066-020-01606-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/09/2020] [Indexed: 01/24/2023]
Abstract
PURPOSE The relation between functional imaging and intrapatient genetic heterogeneity remains poorly understood. The aim of our study was to investigate spatial sampling and functional imaging by FDG-PET/MRI to describe intrapatient tumour heterogeneity. METHODS Six patients with oropharyngeal cancer were included in this pilot study. Two tumour samples per patient were taken and sequenced by next-generation sequencing covering 327 genes relevant in head and neck cancer. Corresponding regions were delineated on pretherapeutic FDG-PET/MRI images to extract apparent diffusion coefficients and standardized uptake values. RESULTS Samples were collected within the primary tumour (n = 3), within the primary tumour and the involved lymph node (n = 2) as well as within two independent primary tumours (n = 1). Genetic heterogeneity of the primary tumours was limited and most driver gene mutations were found ubiquitously. Slightly increasing heterogeneity was found between primary tumours and lymph node metastases. One private predicted driver mutation within a primary tumour and one in a lymph node were found. However, the two independent primary tumours did not show any shared mutations in spite of a clinically suspected field cancerosis. No conclusive correlation between genetic heterogeneity and heterogeneity of PET/MRI-derived parameters was observed. CONCLUSION Our limited data suggest that single sampling might be sufficient in some patients with oropharyngeal cancer. However, few driver mutations might be missed and, if feasible, spatial sampling should be considered. In two independent primary tumours, both lesions should be sequenced. Our data with a limited number of patients do not support the concept that multiparametric PET/MRI features are useful to guide biopsies for genetic tumour characterization.
Collapse
|
27
|
Crombé A, Saut O, Guigui J, Italiano A, Buy X, Kind M. Influence of temporal parameters of DCE‐MRI on the quantification of heterogeneity in tumor vascularization. J Magn Reson Imaging 2019; 50:1773-1788. [DOI: 10.1002/jmri.26753] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/31/2019] [Accepted: 04/02/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Amandine Crombé
- Department of RadiologyInstitut Bergonié, Comprehensive Cancer Center Bordeaux France
- University of BordeauxIMB, UMR CNRS 5251, INRIA Project Team Monc Talence France
| | - Olivier Saut
- University of BordeauxIMB, UMR CNRS 5251, INRIA Project Team Monc Talence France
| | - Jerome Guigui
- Department of RadiologyInstitut Bergonié, Comprehensive Cancer Center Bordeaux France
| | - Antoine Italiano
- Department of Medical OncologyInstitut Bergonié, Comprehensive Cancer Center Bordeaux France
| | - Xavier Buy
- Department of RadiologyInstitut Bergonié, Comprehensive Cancer Center Bordeaux France
| | - Michèle Kind
- Department of RadiologyInstitut Bergonié, Comprehensive Cancer Center Bordeaux France
| |
Collapse
|
28
|
Blocker SJ, Mowery YM, Holbrook MD, Qi Y, Kirsch DG, Johnson GA, Badea CT. Bridging the translational gap: Implementation of multimodal small animal imaging strategies for tumor burden assessment in a co-clinical trial. PLoS One 2019; 14:e0207555. [PMID: 30958825 PMCID: PMC6453461 DOI: 10.1371/journal.pone.0207555] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/13/2019] [Indexed: 12/17/2022] Open
Abstract
In designing co-clinical cancer studies, preclinical imaging brings unique challenges that emphasize the gap between man and mouse. Our group is developing quantitative imaging methods for the preclinical arm of a co-clinical trial studying immunotherapy and radiotherapy in a soft tissue sarcoma model. In line with treatment for patients enrolled in the clinical trial SU2C-SARC032, primary mouse sarcomas are imaged with multi-contrast micro-MRI (T1 weighted, T2 weighted, and T1 with contrast) before and after immune checkpoint inhibition and pre-operative radiation therapy. Similar to the patients, after surgery the mice will be screened for lung metastases with micro-CT using respiratory gating. A systems evaluation was undertaken to establish a quantitative baseline for both the MR and micro-CT systems against which others systems might be compared. We have constructed imaging protocols which provide clinically-relevant resolution and contrast in a genetically engineered mouse model of sarcoma. We have employed tools in 3D Slicer for semi-automated segmentation of both MR and micro-CT images to measure tumor volumes efficiently and reliably in a large number of animals. Assessment of tumor burden in the resulting images was precise, repeatable, and reproducible. Furthermore, we have implemented a publicly accessible platform for sharing imaging data collected during the study, as well as protocols, supporting information, and data analyses. In doing so, we aim to improve the clinical relevance of small animal imaging and begin establishing standards for preclinical imaging of tumors from the perspective of a co-clinical trial.
Collapse
Affiliation(s)
- S. J. Blocker
- Center for In Vivo Microscopy, Duke University Medical Center, Durham, NC, United States of America
| | - Y. M. Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, United States of America
| | - M. D. Holbrook
- Center for In Vivo Microscopy, Duke University Medical Center, Durham, NC, United States of America
| | - Y. Qi
- Center for In Vivo Microscopy, Duke University Medical Center, Durham, NC, United States of America
| | - D. G. Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, United States of America
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, United States of America
| | - G. A. Johnson
- Center for In Vivo Microscopy, Duke University Medical Center, Durham, NC, United States of America
| | - C. T. Badea
- Center for In Vivo Microscopy, Duke University Medical Center, Durham, NC, United States of America
| |
Collapse
|
29
|
Tumor Ensemble-Based Modeling and Visualization of Emergent Angiogenic Heterogeneity in Breast Cancer. Sci Rep 2019; 9:5276. [PMID: 30918274 PMCID: PMC6437174 DOI: 10.1038/s41598-019-40888-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/19/2019] [Indexed: 01/29/2023] Open
Abstract
There is a critical need for new tools to investigate the spatio-temporal heterogeneity and phenotypic alterations that arise in the tumor microenvironment. However, computational investigations of emergent inter- and intra-tumor angiogenic heterogeneity necessitate 3D microvascular data from 'whole-tumors' as well as "ensembles" of tumors. Until recently, technical limitations such as 3D imaging capabilities, computational power and cost precluded the incorporation of whole-tumor microvascular data in computational models. Here, we describe a novel computational approach based on multimodality, 3D whole-tumor imaging data acquired from eight orthotopic breast tumor xenografts (i.e. a tumor 'ensemble'). We assessed the heterogeneous angiogenic landscape from the microvascular to tumor ensemble scale in terms of vascular morphology, emergent hemodynamics and intravascular oxygenation. We demonstrate how the abnormal organization and hemodynamics of the tumor microvasculature give rise to unique microvascular niches within the tumor and contribute to inter- and intra-tumor heterogeneity. These tumor ensemble-based simulations together with unique data visualization approaches establish the foundation of a novel 'cancer atlas' for investigators to develop their own in silico systems biology applications. We expect this hybrid image-based modeling framework to be adaptable for the study of other tissues (e.g. brain, heart) and other vasculature-dependent diseases (e.g. stroke, myocardial infarction).
Collapse
|
30
|
Noninvasive evaluation of 18F-FDG/ 18F-FMISO-based Micro PET in monitoring hepatic metastasis of colorectal cancer. Sci Rep 2018; 8:17832. [PMID: 30546057 PMCID: PMC6292879 DOI: 10.1038/s41598-018-36238-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 11/15/2018] [Indexed: 12/19/2022] Open
Abstract
This study aimed to explore the application of two radiotracers (18F-fluorodeoxyglucose (FDG) and 18F-fluoromisonidazole (FMISO)) in monitoring hepatic metastases of human colorectal cancer (CRC). Mouse models of CRC hepatic metastases were established by implantation of the human CRC cell lines LoVo and HT29 by intrasplenic injection. Wound healing and Transwell assays were performed to examine cell migration and invasion abilities. Radiotracer-based cellular uptake in vitro and micro-positron emission tomography imaging of liver metastases in vivo were performed. The incidence of liver metastases in LoVo-xenografted mice was significantly higher than that in HT29-xenografted ones. The SUVmax/mean values of 18F-FMISO, but not 18F-FDG, in LoVo xenografts were significantly greater than in HT29 xenografts. In vitro, LoVo cells exhibited stronger metastatic potential and higher radiotracer uptake than HT29 cells. Mechanistically, the expression of HIF-1α and GLUT-1 in LoVo cells and LoVo tumor tissues was remarkably higher than in HT29 cells and tissues. Linear regression analysis demonstrated correlations between cellular 18F-FDG/18F-FMISO uptake and HIF-1α/GLUT-1 expression in vitro, as well as between 18F-FMISO SUVmax and GLUT-1 expression in vivo. 18F-FMISO uptake may serve as a potential biomarker for the detection of liver metastases in CRC, whereas its clinical use warrants validation.
Collapse
|
31
|
Tar PD, Thacker NA, Babur M, Watson Y, Cheung S, Little RA, Gieling RG, Williams KJ, O’Connor JPB. A new method for the high-precision assessment of tumor changes in response to treatment. Bioinformatics 2018; 34:2625-2633. [PMID: 29547950 PMCID: PMC6061877 DOI: 10.1093/bioinformatics/bty115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 02/05/2018] [Accepted: 03/12/2018] [Indexed: 11/12/2022] Open
Abstract
Motivation Imaging demonstrates that preclinical and human tumors are heterogeneous, i.e. a single tumor can exhibit multiple regions that behave differently during both development and also in response to treatment. The large variations observed in control group, tumors can obscure detection of significant therapeutic effects due to the ambiguity in attributing causes of change. This can hinder development of effective therapies due to limitations in experimental design rather than due to therapeutic failure. An improved method to model biological variation and heterogeneity in imaging signals is described. Specifically, linear Poisson modeling (LPM) evaluates changes in apparent diffusion co-efficient between baseline and 72 h after radiotherapy, in two xenograft models of colorectal cancer. The statistical significance of measured changes is compared to those attainable using a conventional t-test analysis on basic apparent diffusion co-efficient distribution parameters. Results When LPMs were applied to treated tumors, the LPMs detected highly significant changes. The analyses were significant for all tumors, equating to a gain in power of 4-fold (i.e. equivalent to having a sample size 16 times larger), compared with the conventional approach. In contrast, highly significant changes are only detected at a cohort level using t-tests, restricting their potential use within personalized medicine and increasing the number of animals required during testing. Furthermore, LPM enabled the relative volumes of responding and non-responding tissue to be estimated for each xenograft model. Leave-one-out analysis of the treated xenografts provided quality control and identified potential outliers, raising confidence in LPM data at clinically relevant sample sizes. Availability and implementation TINA Vision open source software is available from www.tina-vision.net. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- P D Tar
- Division of Informatics, Imaging and Data Science, Manchester Pharmacy School, Manchester, UK
| | - N A Thacker
- Division of Informatics, Imaging and Data Science, Manchester Pharmacy School, Manchester, UK
| | - M Babur
- Division of Pharmacy and Optometry, Manchester Pharmacy School, Manchester, UK
| | - Y Watson
- Division of Informatics, Imaging and Data Science, Manchester Pharmacy School, Manchester, UK
| | - S Cheung
- Division of Informatics, Imaging and Data Science, Manchester Pharmacy School, Manchester, UK
| | - R A Little
- Division of Informatics, Imaging and Data Science, Manchester Pharmacy School, Manchester, UK
| | - R G Gieling
- Division of Pharmacy and Optometry, Manchester Pharmacy School, Manchester, UK
| | - K J Williams
- Division of Pharmacy and Optometry, Manchester Pharmacy School, Manchester, UK
- Division of Cancer Sciences, University of Manchester
| | - J P B O’Connor
- Division of Cancer Sciences, University of Manchester
- Department of Radiology, The Christie Hospital NHS Trust, Manchester, UK
| |
Collapse
|
32
|
Wu J, Tha KK, Xing L, Li R. Radiomics and radiogenomics for precision radiotherapy. JOURNAL OF RADIATION RESEARCH 2018; 59:i25-i31. [PMID: 29385618 PMCID: PMC5868194 DOI: 10.1093/jrr/rrx102] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/14/2017] [Indexed: 06/07/2023]
Abstract
Imaging plays an important role in the diagnosis and staging of cancer, as well as in radiation treatment planning and evaluation of therapeutic response. Recently, there has been significant interest in extracting quantitative information from clinical standard-of-care images, i.e. radiomics, in order to provide a more comprehensive characterization of image phenotypes of the tumor. A number of studies have demonstrated that a deeper radiomic analysis can reveal novel image features that could provide useful diagnostic, prognostic or predictive information, improving upon currently used imaging metrics such as tumor size and volume. Furthermore, these imaging-derived phenotypes can be linked with genomic data, i.e. radiogenomics, in order to understand their biological underpinnings or further improve the prediction accuracy of clinical outcomes. In this article, we will provide an overview of radiomics and radiogenomics, including their rationale, technical and clinical aspects. We will also present some examples of the current results and some emerging paradigms in radiomics and radiogenomics for clinical oncology, with a focus on potential applications in radiotherapy. Finally, we will highlight the challenges in the field and suggest possible future directions in radiomics to maximize its potential impact on precision radiotherapy.
Collapse
Affiliation(s)
- Jia Wu
- Department of Radiation Oncology, Stanford University School of Medicine, 875 Blake Wilbur Drive, Stanford, CA 94305-5847, USA
| | - Khin Khin Tha
- Global Station for Quantum Biomedical Science and Engineering, Global Institute for Cooperative Research and Education, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo 060-8638, Japan
| | - Lei Xing
- Department of Radiation Oncology, Stanford University School of Medicine, 875 Blake Wilbur Drive, Stanford, CA 94305-5847, USA
- Global Station for Quantum Biomedical Science and Engineering, Global Institute for Cooperative Research and Education, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo 060-8638, Japan
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, 875 Blake Wilbur Drive, Stanford, CA 94305-5847, USA
- Global Station for Quantum Biomedical Science and Engineering, Global Institute for Cooperative Research and Education, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo 060-8638, Japan
| |
Collapse
|
33
|
Xie T, Chen X, Fang J, Kang H, Xue W, Tong H, Cao P, Wang S, Yang Y, Zhang W. Textural features of dynamic contrast-enhanced MRI derived model-free and model-based parameter maps in glioma grading. J Magn Reson Imaging 2017; 47:1099-1111. [PMID: 28845594 DOI: 10.1002/jmri.25835] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/25/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Presurgical glioma grading by dynamic contrast-enhanced MRI (DCE-MRI) has unresolved issues. PURPOSE The aim of this study was to investigate the ability of textural features derived from pharmacokinetic model-based or model-free parameter maps of DCE-MRI in discriminating between different grades of gliomas, and their correlation with pathological index. STUDY TYPE Retrospective. SUBJECTS Forty-two adults with brain gliomas. FIELD STRENGTH/SEQUENCE 3.0T, including conventional anatomic sequences and DCE-MRI sequences (variable flip angle T1-weighted imaging and three-dimensional gradient echo volumetric imaging). ASSESSMENT Regions of interest on the cross-sectional images with maximal tumor lesion. Five commonly used textural features, including Energy, Entropy, Inertia, Correlation, and Inverse Difference Moment (IDM), were generated. RESULTS All textural features of model-free parameters (initial area under curve [IAUC], maximal signal intensity [Max SI], maximal up-slope [Max Slope]) could effectively differentiate between grade II (n = 15), grade III (n = 13), and grade IV (n = 14) gliomas (P < 0.05). Two textural features, Entropy and IDM, of four DCE-MRI parameters, including Max SI, Max Slope (model-free parameters), vp (Extended Tofts), and vp (Patlak) could differentiate grade III and IV gliomas (P < 0.01) in four measurements. Both Entropy and IDM of Patlak-based Ktrans and vp could differentiate grade II (n = 15) from III (n = 13) gliomas (P < 0.01) in four measurements. No textural features of any DCE-MRI parameter maps could discriminate between subtypes of grade II and III gliomas (P < 0.05). Both Entropy and IDM of Extended Tofts- and Patlak-based vp showed highest area under curve in discriminating between grade III and IV gliomas. However, intraclass correlation coefficient (ICC) of these features revealed relatively lower inter-observer agreement. No significant correlation was found between microvascular density and textural features, compared with a moderate correlation found between cellular proliferation index and those features. DATA CONCLUSION Textural features of DCE-MRI parameter maps displayed a good ability in glioma grading. LEVEL OF EVIDENCE 3 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2018;47:1099-1111.
Collapse
Affiliation(s)
- Tian Xie
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiao Chen
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jingqin Fang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Houyi Kang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Wei Xue
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Haipeng Tong
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Peng Cao
- GE HealthCare (China), Pudong, Shanghai, China
| | - Sumei Wang
- Department of Radiology, Division of Neuroradiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yizeng Yang
- Department of Medicine, Gastroenterology Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Weiguo Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, China
| |
Collapse
|
34
|
Wang DC, Wang X. Systems heterogeneity: An integrative way to understand cancer heterogeneity. Semin Cell Dev Biol 2016; 64:1-4. [PMID: 27552921 DOI: 10.1016/j.semcdb.2016.08.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 12/14/2022]
Abstract
The concept of systems heterogeneity was firstly coined and explained in the Special Issue, as a new alternative to understand the importance and complexity of heterogeneity in cancer. Systems heterogeneity can offer a full image of heterogeneity at multi-dimensional functions and multi-omics by integrating gene or protein expression, epigenetics, sequencing, phosphorylation, transcription, pathway, or interaction. The Special Issue starts with the roles of epigenetics in the initiation and development of cancer heterogeneity through the interaction between permanent genetic mutations and dynamic epigenetic alterations. Cell heterogeneity was defined as the difference in biological function and phenotypes between cells in the same organ/tissue or in different organs, as well as various challenges, as exampled in telocytes. The single cell heterogeneity has the value of identifying diagnostic biomarkers and therapeutic targets and clinical potential of single cell systems heterogeneity in clinical oncology. A number of signaling pathways and factors contribute to the development of systems heterogeneity. Proteomic heterogeneity can change the strategy and thinking of drug discovery and development by understanding the interactions between proteins or proteins with drugs in order to optimize drug efficacy and safety. The association of cancer heterogeneity with cancer cell evolution and metastasis was also overviewed as a new alternative for diagnostic biomarkers and therapeutic targets in clinical application.
Collapse
Affiliation(s)
- Diane Catherine Wang
- Minghang Hospital of Fudan University, Shanghai Medical College, Shanghai, China
| | - Xiangdong Wang
- Minghang Hospital of Fudan University, Shanghai Medical College, Shanghai, China.
| |
Collapse
|