51
|
Katiyar P, Schwenck J, Frauenfeld L, Divine MR, Agrawal V, Kohlhofer U, Gatidis S, Kontermann R, Königsrainer A, Quintanilla-Martinez L, la Fougère C, Schölkopf B, Pichler BJ, Disselhorst JA. Quantification of intratumoural heterogeneity in mice and patients via machine-learning models trained on PET-MRI data. Nat Biomed Eng 2023; 7:1014-1027. [PMID: 37277483 DOI: 10.1038/s41551-023-01047-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/26/2023] [Indexed: 06/07/2023]
Abstract
In oncology, intratumoural heterogeneity is closely linked with the efficacy of therapy, and can be partially characterized via tumour biopsies. Here we show that intratumoural heterogeneity can be characterized spatially via phenotype-specific, multi-view learning classifiers trained with data from dynamic positron emission tomography (PET) and multiparametric magnetic resonance imaging (MRI). Classifiers trained with PET-MRI data from mice with subcutaneous colon cancer quantified phenotypic changes resulting from an apoptosis-inducing targeted therapeutic and provided biologically relevant probability maps of tumour-tissue subtypes. When applied to retrospective PET-MRI data of patients with liver metastases from colorectal cancer, the trained classifiers characterized intratumoural tissue subregions in agreement with tumour histology. The spatial characterization of intratumoural heterogeneity in mice and patients via multimodal, multiparametric imaging aided by machine-learning may facilitate applications in precision oncology.
Collapse
Affiliation(s)
- Prateek Katiyar
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Max Planck Institute for Intelligent Systems, Tübingen, Germany
| | - Johannes Schwenck
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Leonie Frauenfeld
- Institute of Pathology and Neuropathology, Eberhard Karls University Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Mathew R Divine
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Vaibhav Agrawal
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Max Planck Institute for Intelligent Systems, Tübingen, Germany
| | - Ursula Kohlhofer
- Institute of Pathology and Neuropathology, Eberhard Karls University Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Sergios Gatidis
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Radiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Roland Kontermann
- Institute of Cell Biology and Immunology, SRCSB, University of Stuttgart, Stuttgart, Germany
| | - Alfred Königsrainer
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
- Institute of Pathology and Neuropathology, Eberhard Karls University Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernhard Schölkopf
- Max Planck Institute for Intelligent Systems, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jonathan A Disselhorst
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
52
|
Wang H, Zha H, Du Y, Li C, Zhang J, Ye X. An integrated radiomics nomogram based on conventional ultrasound improves discriminability between fibroadenoma and pure mucinous carcinoma in breast. Front Oncol 2023; 13:1170729. [PMID: 37427125 PMCID: PMC10324567 DOI: 10.3389/fonc.2023.1170729] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/14/2023] [Indexed: 07/11/2023] Open
Abstract
Objective To evaluate the ability of integrated radiomics nomogram based on ultrasound images to distinguish between breast fibroadenoma (FA) and pure mucinous carcinoma (P-MC). Methods One hundred seventy patients with FA or P-MC (120 in the training set and 50 in the test set) with definite pathological confirmation were retrospectively enrolled. Four hundred sixty-four radiomics features were extracted from conventional ultrasound (CUS) images, and radiomics score (Radscore) was constructed using the Least Absolute Shrinkage and Selection Operator (LASSO) algorithm. Different models were developed by a support vector machine (SVM), and the diagnostic performance of the different models was assessed and validated. A comparison of the receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA) was performed to evaluate the incremental value of the different models. Results Finally, 11 radiomics features were selected, and then Radscore was developed based on them, which was higher in P-MC in both cohorts. In the test group, the clinic + CUS + radiomics (Clin + CUS + Radscore) model achieved a significantly higher area under the curve (AUC) value (AUC = 0.86, 95% CI, 0.733-0.942) when compared with the clinic + radiomics (Clin + Radscore) (AUC = 0.76, 95% CI, 0.618-0.869, P > 0.05), clinic + CUS (Clin + CUS) (AUC = 0.76, 95% CI, 0.618-0.869, P< 0.05), Clin (AUC = 0.74, 95% CI, 0.600-0.854, P< 0.05), and Radscore (AUC = 0.64, 95% CI, 0.492-0.771, P< 0.05) models, respectively. The calibration curve and DCA also suggested excellent clinical value of the combined nomogram. Conclusion The combined Clin + CUS + Radscore model may help improve the differentiation of FA from P-MC.
Collapse
Affiliation(s)
- Hui Wang
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hailing Zha
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Du
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cuiying Li
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiulou Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinhua Ye
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
53
|
Masud MA, Kim JY, Kim E. Effective dose window for containing tumor burden under tolerable level. NPJ Syst Biol Appl 2023; 9:17. [PMID: 37221258 DOI: 10.1038/s41540-023-00279-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/05/2023] [Indexed: 05/25/2023] Open
Abstract
A maximum-tolerated dose (MTD) reduces the drug-sensitive cell population, though it may result in the competitive release of drug resistance. Alternative treatment strategies such as adaptive therapy (AT) or dose modulation aim to impose competitive stress on drug-resistant cell populations by maintaining a sufficient number of drug-sensitive cells. However, given the heterogeneous treatment response and tolerable tumor burden level of individual patients, determining an effective dose that can fine-tune competitive stress remains challenging. This study presents a mathematical model-driven approach that determines the plausible existence of an effective dose window (EDW) as a range of doses that conserve sufficient sensitive cells while maintaining the tumor volume below a threshold tolerable tumor volume (TTV). We use a mathematical model that explains intratumor cell competition. Analyzing the model, we derive an EDW determined by TTV and the competitive strength. By applying a fixed endpoint optimal control model, we determine the minimal dose to contain cancer at a TTV. As a proof of concept, we study the existence of EDW for a small cohort of melanoma patients by fitting the model to longitudinal tumor response data. We performed identifiability analysis, and for the patients with uniquely identifiable parameters, we deduced patient-specific EDW and minimal dose. The tumor volume for a patient could be theoretically contained at the TTV either using continuous dose or AT strategy with doses belonging to EDW. Further, we conclude that the lower bound of the EDW approximates the minimum effective dose (MED) for containing tumor volume at the TTV.
Collapse
Affiliation(s)
- M A Masud
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Eunjung Kim
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea.
| |
Collapse
|
54
|
Panico A, Gatta G, Salvia A, Grezia GD, Fico N, Cuccurullo V. Radiomics in Breast Imaging: Future Development. J Pers Med 2023; 13:jpm13050862. [PMID: 37241032 DOI: 10.3390/jpm13050862] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/02/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Breast cancer is the most common and most commonly diagnosed non-skin cancer in women. There are several risk factors related to habits and heredity, and screening is essential to reduce the incidence of mortality. Thanks to screening and increased awareness among women, most breast cancers are diagnosed at an early stage, increasing the chances of cure and survival. Regular screening is essential. Mammography is currently the gold standard for breast cancer diagnosis. In mammography, we can encounter problems with the sensitivity of the instrument; in fact, in the case of a high density of glands, the ability to detect small masses is reduced. In fact, in some cases, the lesion may not be particularly evident, it may be hidden, and it is possible to incur false negatives as partial details that may escape the radiologist's eye. The problem is, therefore, substantial, and it makes sense to look for techniques that can increase the quality of diagnosis. In recent years, innovative techniques based on artificial intelligence have been used in this regard, which are able to see where the human eye cannot reach. In this paper, we can see the application of radiomics in mammography.
Collapse
Affiliation(s)
- Alessandra Panico
- Radiology Division, Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Gianluca Gatta
- Radiology Division, Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Antonio Salvia
- Radiology Division, Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | | | - Noemi Fico
- Department of Physics "Ettore Pancini", Università di Napoli Federico II, 80126 Naples, Italy
| | - Vincenzo Cuccurullo
- Nuclear Medicine Unit, Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80138 Naples, Italy
| |
Collapse
|
55
|
Ma Y, Shi F, Sun T, Chen H, Cheng H, Liu X, Wu S, Lu J, Zou Y, Zhang J, Jin L, Shen D, Wu J. Histopathological auxiliary system for brain tumour (HAS-Bt) based on weakly supervised learning using a WHO CNS5-style pipeline. J Neurooncol 2023; 163:71-82. [PMID: 37173511 DOI: 10.1007/s11060-023-04306-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 03/31/2023] [Indexed: 05/15/2023]
Abstract
PURPOSE Classification and grading of central nervous system (CNS) tumours play a critical role in the clinic. When WHO CNS5 simplifies the histopathology diagnosis and places greater emphasis on molecular pathology, artificial intelligence (AI) has been widely used to meet the increased need for an automatic histopathology scheme that could liberate pathologists from laborious work. This study was to explore the diagnosis scope and practicality of AI. METHODS A one-stop Histopathology Auxiliary System for Brain tumours (HAS-Bt) is introduced based on a pipeline-structured multiple instance learning (pMIL) framework developed with 1,385,163 patches from 1038 hematoxylin and eosin (H&E) slides. The system provides a streamlined service including slide scanning, whole-slide image (WSI) analysis and information management. A logical algorithm is used when molecular profiles are available. RESULTS The pMIL achieved an accuracy of 0.94 in a 9-type classification task on an independent dataset composed of 268 H&E slides. Three auxiliary functions are developed and a built-in decision tree with multiple molecular markers is used to automatically formed integrated diagnosis. The processing efficiency was 443.0 s per slide. CONCLUSION HAS-Bt shows outstanding performance and provides a novel aid for the integrated neuropathological diagnostic workflow of brain tumours using CNS 5 pipeline.
Collapse
Affiliation(s)
- Yixin Ma
- Glioma Surgery Division, Neurologic Surgery Department, Huashan Hospital Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital Fudan University, Shanghai, China
| | - Feng Shi
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Tianyang Sun
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Hong Chen
- National Center for Neurological Disorders, Huashan Hospital Fudan University, Shanghai, China
- Department of Pathology, Huashan Hospital Fudan University, Shanghai, China
| | - Haixia Cheng
- National Center for Neurological Disorders, Huashan Hospital Fudan University, Shanghai, China
- Department of Pathology, Huashan Hospital Fudan University, Shanghai, China
| | - Xiaojia Liu
- National Center for Neurological Disorders, Huashan Hospital Fudan University, Shanghai, China
- Department of Pathology, Huashan Hospital Fudan University, Shanghai, China
| | - Shuai Wu
- Glioma Surgery Division, Neurologic Surgery Department, Huashan Hospital Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital Fudan University, Shanghai, China
| | - Junfeng Lu
- Glioma Surgery Division, Neurologic Surgery Department, Huashan Hospital Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital Fudan University, Shanghai, China
| | - Yaping Zou
- Wuhan Zhongji Biotechnology Co., Ltd, Wuhan, China
| | - Jun Zhang
- Wuhan Zhongji Biotechnology Co., Ltd, Wuhan, China
| | - Lei Jin
- Glioma Surgery Division, Neurologic Surgery Department, Huashan Hospital Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Huashan Hospital Fudan University, Shanghai, China.
| | - Dinggang Shen
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China.
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| | - Jinsong Wu
- Glioma Surgery Division, Neurologic Surgery Department, Huashan Hospital Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Huashan Hospital Fudan University, Shanghai, China.
| |
Collapse
|
56
|
Uysal E, Topaloğlu ÖF, Arı A, Özer H, Koplay M. Can magnetic resonance imaging texture analysis change the breast imaging reporting and data system category of breast lesions? Clin Imaging 2023; 97:44-49. [PMID: 36889114 DOI: 10.1016/j.clinimag.2023.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 02/19/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
PURPOSE This study aimed to reveal magnetic resonance imaging (MRI) texture analysis (TA)'s contribution to categorizing breast lesions according to the Breast Imaging-Reporting and Data System (BI-RADS) lexicon. METHOD Two hundred and seventeen women with BI-RADS category 3, 4, and 5 lesions on breast MRI were included in the study. For TA, the region of interest was drawn manually to encompass the entire lesion on the fat-suppressed T2W and the first post-contrast T1W images. To identify the independent predictors of breast cancer, multivariate logistic regression analyses were performed using texture parameters. Estimated benign and malignant groups were formed according to the TA regression model. RESULTS Texture parameters extracted from T2WI, including median, gray-level co-occurrence matrix (GLCM) contrast, GLCM correlation, GLCM joint entropy, GLCM sum entropy, and GLCM sum of squares, and parameters extracted from T1WI, including maximum, GLCM contrast, GLCM joint entropy, GLCM sum entropy, were independent predictors of breast cancer. In the estimated new groups according to the TA regression model, 19 (91%) of the benign 4a lesions were downgraded to BI-RADS category 3. CONCLUSIONS The addition of quantitative parameters obtained by MRI TA to BI-RADS criteria significantly increased the accuracy rate in differentiating benign and malignant breast lesions. When categorizing BI-RADS 4a lesions, the use of MRI TA in addition to conventional imaging findings may reduce unnecessary biopsy rates.
Collapse
Affiliation(s)
- Emine Uysal
- Department of Radiology, Faculty of Medicine, Selçuk University, Selçuklu, Konya, Turkey.
| | - Ömer Faruk Topaloğlu
- Department of Radiology, Faculty of Medicine, Selçuk University, Selçuklu, Konya, Turkey
| | - Ayşe Arı
- Department of Radiology, Faculty of Medicine, Selçuk University, Selçuklu, Konya, Turkey
| | - Halil Özer
- Department of Radiology, Faculty of Medicine, Selçuk University, Selçuklu, Konya, Turkey
| | - Mustafa Koplay
- Department of Radiology, Faculty of Medicine, Selçuk University, Selçuklu, Konya, Turkey
| |
Collapse
|
57
|
Pan Z, Men K, Liang B, Song Z, Wu R, Dai J. A subregion-based prediction model for local-regional recurrence risk in head and neck squamous cell carcinoma. Radiother Oncol 2023; 184:109684. [PMID: 37120101 DOI: 10.1016/j.radonc.2023.109684] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND AND PURPOSE Given that the intratumoral heterogeneity of head and neck squamous cell carcinoma may be related to the local control rate of radiotherapy, the aim of this study was to construct a subregion-based model that can predict the risk of local-regional recurrence, and to quantitatively assess the relative contribution of subregions. MATERIALS AND METHODS The CT images, PET images, dose images and GTVs of 228 patients with head and neck squamous cell carcinoma from four different institutions of the The Cancer Imaging Archive(TCIA) were included in the study. Using a supervoxel segmentation algorithm called maskSLIC to generate individual-level subregions. After extracting 1781 radiomics and 1767 dosiomics features from subregions, an attention-based multiple instance risk prediction model (MIR) was established. The GTV model was developed based on the whole tumour area and was used to compare the prediction performance with the MIR model. Furthermore, the MIR-Clinical model was constructed by integrating the MIR model with clinical factors. Subregional analysis was carried out through the Wilcoxon test to find the differential radiomic features between the highest and lowest weighted subregions. RESULTS Compared with the GTV model, the C-index of MIR model was significantly increased from 0.624 to 0.721(Wilcoxon test, p value< 0.0001). When MIR model was combined with clinical factors, the C-index was further increased to 0.766. Subregional analysis showed that for LR patients, the top three differential radiomic features between the highest and lowest weighted subregions were GLRLM_ShortRunHighGrayLevelEmphasis, GRLM_HghGrayLevelRunEmphasis and GLRLM_LongRunHighGrayLevelEmphasis. CONCLUSION This study developed a subregion-based model that can predict the risk of local-regional recurrence and quantitatively assess relevant subregions, which may provide technical support for the precision radiotherapy in head and neck squamous cell carcinoma.
Collapse
Affiliation(s)
- Ziqi Pan
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Kuo Men
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bin Liang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhiyue Song
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Runye Wu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jianrong Dai
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
58
|
Fang Z, Pu H, Chen XL, Yuan Y, Zhang F, Li H. MRI radiomics signature to predict lymph node metastasis after neoadjuvant chemoradiation therapy in locally advanced rectal cancer. Abdom Radiol (NY) 2023; 48:2270-2283. [PMID: 37085730 DOI: 10.1007/s00261-023-03910-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/01/2023] [Accepted: 04/05/2023] [Indexed: 04/23/2023]
Abstract
PURPOSE To investigative the performance of MRI-radiomics analysis derived from T2WI and apparent diffusion coefficients (ADC) images before and after neoadjuvant chemoradiation therapy (nCRT) separately or simultaneously for predicting post-nCRT lymph node status in patients with locally advanced rectal cancer (LARC). MATERIALS AND METHODS: Eighty-three patients (training cohort, n = 57; validation cohort, n = 26) with LARC between June 2017 and December 2022 were retrospectively enrolled. All the radiomics features were extracted from volume of interest on T2WI and ADC images from baseline and post-nCRT MRI. Delta-radiomics features were defined as the difference between radiomics features before and after nCRT. Seven clinical-radiomics models were constructed by combining the most predictive radiomics signatures and clinical parameters selected from support vector machine. Receiver operating characteristic curve (ROC) was used to evaluate the performance of models. The optimum model-based LNM was applied to assess 5-years disease-free survival (DFS) using Kaplan-Meier analysis. The end point was clinical or radiological locoregional recurrence or distant metastasis during postoperative follow-up. RESULTS Clinical-deltaADC radiomics combined model presented good performance for predicting post-CRT LNM in the training (AUC = 0.895,95%CI:0.838-0.953) and validation cohort (AUC = 0.900,95%CI:0.771-1.000). Clinical-deltaADC radiomics-postT2WI radiomics combined model also showed good performances (AUC = 0.913,95%CI:0.838-0.953) in the training and (AUC = 0.912,95%CI:0.771-1.000) validation cohort. As for subgroup analysis, clinical-deltaADC radiomics combined model showed good performance predicting LNM in ypT0-T2 (AUC = 0.827;95%CI:0.649-1.000) and ypT3-T4 stage (AUC = 0.934;95%CI:0.864-1.000). In ypT0-T2 stage, clinical-deltaADC radiomics combined model-based LNM could assess 5-years DFS (P = 0.030). CONCLUSION Clinical-deltaADC radiomics combined model could predict post-nCRT LNM, and this combined model-based LNM was associated with 5-years DFS in ypT0-T2 stage.
Collapse
Affiliation(s)
- Zhu Fang
- Department of Radiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, 32# Second Section of First Ring Road, Qingyang District, Chengdu, 610070, Sichuan, China
| | - Hong Pu
- Department of Radiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, 32# Second Section of First Ring Road, Qingyang District, Chengdu, 610070, Sichuan, China
| | - Xiao-Li Chen
- Department of Radiology, Affiliated Cancer Hospital of Medical School, University of Electronic Science and Technology of China, Sichuan Cancer Hospital, 55#Four Section of South Renmin Road, Wuhou District, Chengdu, 610000, China
| | - Yi Yuan
- Department of Radiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, 32# Second Section of First Ring Road, Qingyang District, Chengdu, 610070, Sichuan, China
| | - Feng Zhang
- Department of Radiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, 32# Second Section of First Ring Road, Qingyang District, Chengdu, 610070, Sichuan, China
| | - Hang Li
- Department of Radiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, 32# Second Section of First Ring Road, Qingyang District, Chengdu, 610070, Sichuan, China.
| |
Collapse
|
59
|
Deng S, Zhu Y. Prediction of Glioma Grade by Tumor Heterogeneity Radiomic Analysis Based on Multiparametric MRI. INT J COMPUT INT SYS 2023. [DOI: 10.1007/s44196-023-00230-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
AbstractPredicting glioma grade plays a pivotal role in treatment and prognosis. However, several current methods for grading depend on the characteristics of the whole tumor. Predicting grade by analyzing tumor subregions has not been thoroughly investigated, which aims to improve the prediction performance. To predict glioma grade via analysis of tumor heterogeneity with features extracted from tumor subregions, it is mainly divided into four magnetic resonance imaging (MRI) sequences, including T2-weighted (T2), fluid-attenuated inversion recovery (FLAIR), pre-gadolinium T1-weighted (T1), and post-gadolinium T1-weighted methods. This study included the data of 97 patients with glioblastomas and 42 patients with low-grade gliomas before surgery. Three subregions, including enhanced tumor (ET), non-enhanced tumor, and peritumoral edema, were obtained based on segmentation labels generated by the GLISTRBoost algorithm. One hundred radiomic features were extracted from each subregion. Feature selection was performed using the cross-validated recursive feature elimination with a support vector machine (SVM) algorithm. SVM classifiers with grid search were established to predict glioma grade based on unparametric and multiparametric MRI. The area under the receiver operating characteristic curve (AUC) was used to evaluate the performance of the classifiers, and the performance of the subregions was compared with the results of the whole tumor. In uniparametric analysis, the features from the ET subregion yielded a higher AUC value of 0.8697, 0.8474, and 0.8474 than those of the whole tumor of FLAIR, T1, and T2. In multiparametric analysis, the ET subregion achieved the best performance (AUC = 0.8755), which was higher than the uniparametric results. Radiomic features from the tumor subregion can potentially be used as clinical markers to improve the predictive accuracy of glioma grades.
Collapse
|
60
|
Walentynowicz KA, Engelhardt D, Cristea S, Yadav S, Onubogu U, Salatino R, Maerken M, Vincentelli C, Jhaveri A, Geisberg J, McDonald TO, Michor F, Janiszewska M. Single-cell heterogeneity of EGFR and CDK4 co-amplification is linked to immune infiltration in glioblastoma. Cell Rep 2023; 42:112235. [PMID: 36920905 PMCID: PMC10114292 DOI: 10.1016/j.celrep.2023.112235] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/20/2022] [Accepted: 02/23/2023] [Indexed: 03/15/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor, with a median survival of ∼15 months. Targeted approaches have not been successful in this tumor type due to the large extent of intratumor heterogeneity. Mosaic amplification of oncogenes suggests that multiple genetically distinct clones are present in each tumor. To uncover the relationships between genetically diverse subpopulations of GBM cells and their native tumor microenvironment, we employ highly multiplexed spatial protein profiling coupled with single-cell spatial mapping of fluorescence in situ hybridization (FISH) for EGFR, CDK4, and PDGFRA. Single-cell FISH analysis of a total of 35,843 single nuclei reveals that tumors in which amplifications of EGFR and CDK4 more frequently co-occur in the same cell exhibit higher infiltration of CD163+ immunosuppressive macrophages. Our results suggest that high-throughput assessment of genomic alterations at the single-cell level could provide a measure for predicting the immune state of GBM.
Collapse
Affiliation(s)
- Kacper A Walentynowicz
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technologies, Jupiter, FL, USA; Department of Molecular Medicine, Scripps Research, Jupiter, FL, USA
| | - Dalit Engelhardt
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Simona Cristea
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Department of Medical Oncology, Harvard Medical School, Boston, MA, USA
| | - Shreya Yadav
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technologies, Jupiter, FL, USA; Department of Molecular Medicine, Scripps Research, Jupiter, FL, USA
| | - Ugoma Onubogu
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technologies, Jupiter, FL, USA; Department of Molecular Medicine, Scripps Research, Jupiter, FL, USA; The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Roberto Salatino
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technologies, Jupiter, FL, USA; Department of Molecular Medicine, Scripps Research, Jupiter, FL, USA; The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Melanie Maerken
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technologies, Jupiter, FL, USA; Department of Molecular Medicine, Scripps Research, Jupiter, FL, USA
| | | | - Aashna Jhaveri
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jacob Geisberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thomas O McDonald
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Franziska Michor
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; The Broad Institute of MIT and Harvard, Cambridge, MA, USA; The Ludwig Center at Harvard, Boston, MA, USA.
| | - Michalina Janiszewska
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technologies, Jupiter, FL, USA; Department of Molecular Medicine, Scripps Research, Jupiter, FL, USA; The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
61
|
Lee DH, Park JE, Kim N, Park SY, Kim YH, Cho YH, Kim JH, Kim HS. Tumor Habitat Analysis Using Longitudinal Physiological MRI to Predict Tumor Recurrence After Stereotactic Radiosurgery for Brain Metastasis. Korean J Radiol 2023; 24:235-246. [PMID: 36788768 PMCID: PMC9971843 DOI: 10.3348/kjr.2022.0492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/08/2022] [Accepted: 12/11/2022] [Indexed: 02/16/2023] Open
Abstract
OBJECTIVE It is difficult to predict the treatment response of tissue after stereotactic radiosurgery (SRS) because radiation necrosis (RN) and tumor recurrence can coexist. Our study aimed to predict tumor recurrence, including the recurrence site, after SRS of brain metastasis by performing a longitudinal tumor habitat analysis. MATERIALS AND METHODS Two consecutive multiparametric MRI examinations were performed for 83 adults (mean age, 59.0 years; range, 27-82 years; 44 male and 39 female) with 103 SRS-treated brain metastases. Tumor habitats based on contrast-enhanced T1- and T2-weighted images (structural habitats) and those based on the apparent diffusion coefficient (ADC) and cerebral blood volume (CBV) images (physiological habitats) were defined using k-means voxel-wise clustering. The reference standard was based on the pathology or Response Assessment in Neuro-Oncologycriteria for brain metastases (RANO-BM). The association between parameters of single-time or longitudinal tumor habitat and the time to recurrence and the site of recurrence were evaluated using the Cox proportional hazards regression analysis and Dice similarity coefficient, respectively. RESULTS The mean interval between the two MRI examinations was 99 days. The longitudinal analysis showed that an increase in the hypovascular cellular habitat (low ADC and low CBV) was associated with the risk of recurrence (hazard ratio [HR], 2.68; 95% confidence interval [CI], 1.46-4.91; P = 0.001). During the single-time analysis, a solid low-enhancing habitat (low T2 and low contrast-enhanced T1 signal) was associated with the risk of recurrence (HR, 1.54; 95% CI, 1.01-2.35; P = 0.045). A hypovascular cellular habitat was indicative of the future recurrence site (Dice similarity coefficient = 0.423). CONCLUSION After SRS of brain metastases, an increased hypovascular cellular habitat observed using a longitudinal MRI analysis was associated with the risk of recurrence (i.e., treatment resistance) and was indicative of recurrence site. A tumor habitat analysis may help guide future treatments for patients with brain metastases.
Collapse
Affiliation(s)
- Da Hyun Lee
- Department of Radiology, Ajou University School of Medicine, Suwon, Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| | | | - Seo Young Park
- Department of Statistics and Data Science, Korea National Open University, Seoul, Korea
| | - Young-Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Young Hyun Cho
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jeong Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
62
|
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: 7.0] [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
|
63
|
Identification of an Autophagy-Related Signature for Prognosis and Immunotherapy Response Prediction in Ovarian Cancer. Biomolecules 2023; 13:biom13020339. [PMID: 36830707 PMCID: PMC9953331 DOI: 10.3390/biom13020339] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/08/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is one of the most malignant tumors in the female reproductive system, with a poor prognosis. Various responses to treatments including chemotherapy and immunotherapy are observed among patients due to their individual characteristics. Applicable prognostic markers could make it easier to refine risk stratification for OC patients. Autophagy is closely implicated in the occurrence and development of tumors, including OC. Whether autophagy -related genes can be used as prognostic markers for OC patients remains unclear. METHODS The gene transcriptome data of 374 OC patients were downloaded from The Cancer Genome Atlas (TCGA) database. The correlation between the autophagy levels and outcomes of OC patients was identified through the single sample gene set enrichment analysis (ssGSEA). Recognized molecular markers of autophagy in different clinical specimens were detected by immunohistochemistry (IHC) assay. The gene set enrichment analysis (GSEA), ESTIMATE, and CIBERSORT analysis were applied to explore the correlation of autophagy with the tumor immune microenvironment (TIME). Single-cell RNA-sequencing (scRNA-seq) data from seven OC patients were included for characterizing cell-cell interaction patterns of autophagy-high or low tumor cells. Machine learning, Stepwise Cox regression and LASSO-Cox analysis were used to screen autophagy hub genes, which were used to establish an autophagy-related signature for prognosis evaluation. Four tumor immunotherapy cohorts were obtained from the GEO (Gene Expression Omnibus) database and the literature for autophagy risk score validation. RESULTS The autophagy levels were closely related to the prognosis of the OC patients. Additionally, the autophagy levels were correlated with TIME status including immune score, and immune-cell infiltration. The scRNA-seq analysis found that tumor cells with high or low autophagy levels had different interactions with immune cells, especially macrophages. Eight autophagy-hub genes (ZFYVE1, AMBRA1, LAMP2, TRAF6, PDPK1, ATG2B, DAPK1 and TP53INP2) were screened for an autophagy-related signature. According to this signature, higher risk score was correlated with poor prognosis and better immunotherapy response in the OC patients. CONCLUSIONS The autophagy-related signature is applicable to predict the prognosis and immune checkpoint inhibitors (ICIs) therapy efficiency in OC patients. It is possible to identify OC patients who will respond to ICIs therapy and have a favorable prognosis, although more verification is needed.
Collapse
|
64
|
Bosque JJ, Calvo GF, Molina-García D, Pérez-Beteta J, García Vicente AM, Pérez-García VM. Metabolic activity grows in human cancers pushed by phenotypic variability. iScience 2023; 26:106118. [PMID: 36843844 PMCID: PMC9950952 DOI: 10.1016/j.isci.2023.106118] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/30/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
Different evolutionary processes push cancers to increasingly aggressive behaviors, energetically sustained by metabolic reprogramming. The collective signature emerging from this transition is macroscopically displayed by positron emission tomography (PET). In fact, the most readily PET measure, the maximum standardized uptake value (SUVmax), has been found to have prognostic value in different cancers. However, few works have linked the properties of this metabolic hotspot to cancer evolutionary dynamics. Here, by analyzing diagnostic PET images from 512 patients with cancer, we found that SUVmax scales superlinearly with the mean metabolic activity (SUVmean), reflecting a dynamic preferential accumulation of activity on the hotspot. Additionally, SUVmax increased with metabolic tumor volume (MTV) following a power law. The behavior from the patients data was accurately captured by a mechanistic evolutionary dynamics model of tumor growth accounting for phenotypic transitions. This suggests that non-genetic changes may suffice to fuel the observed sustained increases in tumor metabolic activity.
Collapse
Affiliation(s)
- Jesús J. Bosque
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), University of Castilla-La Mancha, Ciudad Real, Spain,Corresponding author
| | - Gabriel F. Calvo
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), University of Castilla-La Mancha, Ciudad Real, Spain
| | - David Molina-García
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), University of Castilla-La Mancha, Ciudad Real, Spain
| | - Julián Pérez-Beteta
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), University of Castilla-La Mancha, Ciudad Real, Spain
| | - Ana M. García Vicente
- Nuclear Medicine Unit, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Víctor M. Pérez-García
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), University of Castilla-La Mancha, Ciudad Real, Spain
| |
Collapse
|
65
|
Li J, Qiu Z, Zhang C, Chen S, Wang M, Meng Q, Lu H, Wei L, Lv H, Zhong W, Zhang X. ITHscore: comprehensive quantification of intra-tumor heterogeneity in NSCLC by multi-scale radiomic features. Eur Radiol 2023; 33:893-903. [PMID: 36001124 DOI: 10.1007/s00330-022-09055-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/15/2022] [Accepted: 07/24/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVES To quantify intra-tumor heterogeneity (ITH) in non-small cell lung cancer (NSCLC) from computed tomography (CT) images. METHODS We developed a quantitative ITH measurement-ITHscore-by integrating local radiomic features and global pixel distribution patterns. The associations of ITHscore with tumor phenotypes, genotypes, and patient's prognosis were examined on six patient cohorts (n = 1399) to validate its effectiveness in characterizing ITH. RESULTS For stage I NSCLC, ITHscore was consistent with tumor progression from stage IA1 to IA3 (p < 0.001) and captured key pathological change in terms of malignancy (p < 0.001). ITHscore distinguished the presence of lymphovascular invasion (p = 0.003) and pleural invasion (p = 0.001) in tumors. ITHscore also separated patient groups with different overall survival (p = 0.004) and disease-free survival conditions (p = 0.005). Radiogenomic analysis showed that the level of ITHscore in stage I and stage II NSCLC is correlated with heterogeneity-related pathways. In addition, ITHscore was proved to be a stable measurement and can be applied to ITH quantification in head-and-neck cancer (HNC). CONCLUSIONS ITH in NSCLC can be quantified from CT images by ITHscore, which is an indicator for tumor phenotypes and patient's prognosis. KEY POINTS • ITHscore provides a radiomic quantification of intra-tumor heterogeneity in NSCLC. • ITHscore is an indicator for tumor phenotypes and patient's prognosis. • ITHscore has the potential to be generalized to other cancer types such as HNC.
Collapse
Affiliation(s)
- Jiaqi Li
- Bioinformatics Division, BNRIST and MOE Key Lab of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China
| | - Zhenbin Qiu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Sijie Chen
- Bioinformatics Division, BNRIST and MOE Key Lab of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China
| | - Mengmin Wang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qiuchen Meng
- Bioinformatics Division, BNRIST and MOE Key Lab of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China
| | - Haiming Lu
- Bioinformatics Division, BNRIST and MOE Key Lab of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China
| | - Lei Wei
- Bioinformatics Division, BNRIST and MOE Key Lab of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China
| | - Hairong Lv
- Bioinformatics Division, BNRIST and MOE Key Lab of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China
- Fuzhou Institute of Data Technology, Fuzhou, China
| | - Wenzhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Xuegong Zhang
- Bioinformatics Division, BNRIST and MOE Key Lab of Bioinformatics, Department of Automation, Tsinghua University, Beijing, China.
- School of Medicine, Tsinghua University, Beijing, China.
| |
Collapse
|
66
|
Liu J, Wang X, Sahin IH, Imanirad I, Felder SI, Kim RD, Xie H. Tumor Response-speed Heterogeneity as a Novel Prognostic Factor in Patients With Metastatic Colorectal Cancer. Am J Clin Oncol 2023; 46:50-57. [PMID: 36606664 DOI: 10.1097/coc.0000000000000972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE Differential tumor response to therapy is partially attributed to tumor heterogeneity. Additional efforts are needed to identify tumor heterogeneity parameters in response to therapy that is easily applicable in clinical practice. We aimed to describe tumor response-speed heterogeneity and evaluate its prognostic value in patients with metastatic colorectal cancer. PATIENTS AND METHODS Individual patient data from Amgen (NCT00364013) and Sanofi (NCT00305188; NCT00272051) trials were retrieved from Project Data Sphere. Patients in the Amgen 5-fluorouracil, leucovorin, oxaliplatin (FOLFOX) arm were used to establish response-speed heterogeneity. Its prognostic value was subsequently validated in the Sanofi FOLFOX arms and the Amgen panitumumab+FOLFOX arm. Kaplan-Meier method and Cox proportional hazards models were used for survival analyses. RESULTS Patients with high response-speed heterogeneity in the Amgen FOLFOX cohort had significantly shorter ( P <0.001) median progression-free survival (PFS) of 7.27 months (95% CI, 6.12-7.96 mo) and overall survival (OS) of 16.0 months (95% CI, 13.8-18.2 mo) than patients with low response-speed heterogeneity with median PFS of 9.41 months (95% CI, 8.75-10.89 mo) and OS of 22.4 months (95% CI, 20.1-26.7 mo), respectively. Tumor response-speed heterogeneity was a poor prognostic factor of shorter PFS (hazard ratio, 4.17; 95% CI, 2.49-6.99; P <0.001) and shorter OS (hazard ratio, 2.57; 95% CI, 1.64-4.01; P <0.001), after adjustment for other common prognostic factors. Comparable findings were found in the external validation cohorts. CONCLUSION Tumor response-speed heterogeneity to first-line chemotherapy was a novel prognostic factor associated with early disease progression and shorter survival in patients with metastatic colorectal cancer.
Collapse
Affiliation(s)
- Junjia Liu
- Albert Einstein College of Medicine, Bronx, New York
| | | | - Ibrahim H Sahin
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Iman Imanirad
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Seth I Felder
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Richard D Kim
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Hao Xie
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
67
|
Chen M, Wan Y, Li X, Xiang J, Chen X, Jiang J, Han X, Zhong L, Xiao F, Liu J, Huang H, Li H, Liu J, Hou J. Dynamic single-cell RNA-seq analysis reveals distinct tumor program associated with microenvironmental remodeling and drug sensitivity in multiple myeloma. Cell Biosci 2023; 13:19. [PMID: 36717896 PMCID: PMC9887807 DOI: 10.1186/s13578-023-00971-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a hematological malignancy characterized by clonal proliferation of malignant plasma cells. Despite extensive research, molecular mechanisms in MM that drive drug sensitivity and clinic outcome remain elusive. RESULTS Single-cell RNA sequencing was applied to study tumor heterogeneity and molecular dynamics in 10 MM individuals before and after 2 cycles of bortezomib-cyclophosphamide-dexamethasone (VCD) treatment, with 3 healthy volunteers as controls. We identified that unfolded protein response and metabolic-related program were decreased, whereas stress-associated and immune reactive programs were increased after 2 cycles of VCD treatment. Interestingly, low expression of the immune reactive program by tumor cells was associated with unfavorable drug response and poor survival in MM, which probably due to downregulation of MHC class I mediated antigen presentation and immune surveillance, and upregulation of markers related to immune escape. Furthermore, combined with immune cells profiling, we uncovered a link between tumor intrinsic immune reactive program and immunosuppressive phenotype in microenvironment, evidenced by exhausted states and expression of checkpoint molecules and suppressive genes in T cells, NK cells and monocytes. Notably, expression of YBX1 was associated with downregulation of immune activation signaling in myeloma and reduced immune cells infiltration, thereby contributed to poor prognosis. CONCLUSIONS We dissected the tumor and immune reprogramming in MM during targeted therapy at the single-cell resolution, and identified a tumor program that integrated tumoral signaling and changes in immune microenvironment, which provided insights into understanding drug sensitivity in MM.
Collapse
Affiliation(s)
- Mengping Chen
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Yike Wan
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Xin Li
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Jing Xiang
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Xiaotong Chen
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Jinxing Jiang
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Xiaofeng Han
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Lu Zhong
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Fei Xiao
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Jia Liu
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Honghui Huang
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Hua Li
- grid.16821.3c0000 0004 0368 8293Bio-ID Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, 200240 China
| | - Junling Liu
- grid.16821.3c0000 0004 0368 8293Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Jian Hou
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| |
Collapse
|
68
|
Xu H, Abdallah N, Marion JM, Chauvet P, Tauber C, Carlier T, Lu L, Hatt M. Radiomics prognostic analysis of PET/CT images in a multicenter head and neck cancer cohort: investigating ComBat strategies, sub-volume characterization, and automatic segmentation. Eur J Nucl Med Mol Imaging 2023; 50:1720-1734. [PMID: 36690882 DOI: 10.1007/s00259-023-06118-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023]
Abstract
PURPOSE This study aimed to investigate the impact of several ComBat harmonization strategies, intra-tumoral sub-volume characterization, and automatic segmentations for progression-free survival (PFS) prediction through radiomics modeling for patients with head and neck cancer (HNC) in PET/CT images. METHODS The HECKTOR MICCAI 2021 challenge set containing PET/CT images and clinical data of 325 oropharynx HNC patients was exploited. A total of 346 IBSI-compliant radiomic features were extracted for each patient's primary tumor volume defined by the reference manual contours. Modeling relied on least absolute shrinkage Cox regression (Lasso-Cox) for feature selection (FS) and Cox proportional-hazards (CoxPH) models were built to predict PFS. Within this methodological framework, 8 different strategies for ComBat harmonization were compared, including before or after FS, in feature groups separately or all features directly, and with center or clustering-determined labels. Features extracted from tumor sub-volume clustering were also investigated for their prognostic additional value. Finally, 3 automatic segmentations (2 threshold-based and a 3D U-Net) were also compared. All results were evaluated with the concordance index (C-index). RESULTS Radiomics features without harmonization, combined with clinical factors, led to models with C-index values of 0.69 in the testing set. The best version of ComBat harmonization, i.e., after FS, for feature groups separately and relying on clustering-determined labels, achieved a C-index of 0.71. The use of features extracted from tumor sub-volumes further improved the C-index to 0.72. Models that relied on the automatic segmentations yielded close but slightly lower prognostic performance (0.67-0.70) compared to reference contours. CONCLUSION A standard radiomics pipeline allowed for prediction of PFS in a multicenter HNC cohort. Applying a specific strategy of ComBat harmonization improved the performance. The extraction of intra-tumoral sub-volume features and automatic segmentation could contribute to the improvement and automation of prognosis modeling, respectively.
Collapse
Affiliation(s)
- Hui Xu
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, Guangdong, China.,LaTIM, INSERM, UMR 1101, University Brest, Brest, France
| | | | | | | | - Clovis Tauber
- INSERM U930, Université François Rabelais de Tours, Tours, France
| | - Thomas Carlier
- Nuclear Medicine Department, CHU and CRCINA, INSERM, CNRS, Univ Angers, Univ Nantes, Nantes, France
| | - Lijun Lu
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou, 510515, Guangdong, China. .,Pazhou Lab, Guangzhou, 510330, China.
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, University Brest, Brest, France
| |
Collapse
|
69
|
Krishnan SN, Barua S, Frankel TL, Rao A. Towards the characterization of the tumor microenvironment through dictionary learning-based interpretable classification of multiplexed immunofluorescence images. Phys Med Biol 2023; 68:014002. [PMID: 36541756 PMCID: PMC9903331 DOI: 10.1088/1361-6560/aca86a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 12/02/2022] [Indexed: 12/03/2022]
Abstract
Objective.Histology image analysis is a crucial diagnostic step in staging and treatment planning, especially for cancerous lesions. With the increasing adoption of computational methods for image analysis, significant strides are being made to improve the performance metrics of image segmentation and classification frameworks. However, many developed frameworks effectively function as black boxes, granting minimal context to the decision-making process. Thus, there is a need to develop methods that offer reasonable discriminatory power and a biologically-informed intuition to the decision-making process.Approach.In this study, we utilized and modified a discriminative feature-based dictionary learning (DFDL) paradigm to generate a classification framework that allows for discrimination between two distinct clinical histologies. This framework allows us (i) to discriminate between 2 clinically distinct diseases or histologies and (ii) provides interpretable group-specific representative dictionary image patches, or 'atoms', generated during classifier training. This implementation is performed on multiplexed immunofluorescence images from two separate patient cohorts- a pancreatic cohort consisting of cancerous and non-cancerous tissues and a metastatic non-small cell lung cancer (mNSCLC) cohort of responders and non-responders to an immunotherapeutic treatment regimen. The analysis was done at both the image-level and subject-level. Five cell types were selected, namely, epithelial cells, cytotoxic lymphocytes, antigen presenting cells, HelperT cells, and T-regulatory cells, as our phenotypes of interest.Results.We showed that DFDL had significant discriminant capabilities for both the pancreatic pathologies cohort (subject-level AUC-0.8878) and the mNSCLC immunotherapy response cohort (subject-level AUC-0.7221). The secondary analysis also showed that more than 50% of the obtained dictionary atoms from the classifier contained biologically relevant information.Significance.Our method shows that the generated dictionary features can help distinguish patients presenting two different histologies with strong sensitivity and specificity metrics. These features allow for an additional layer of model interpretability, a highly desirable element in clinical applications for identifying novel biological phenomena.
Collapse
Affiliation(s)
- Santhoshi N Krishnan
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas-77005, United States of America,
Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan-48109, United States of America
| | - Souptik Barua
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas-77005, United States of America
| | - Timothy L Frankel
- Department of Surgery, University of Michigan, Ann Arbor, Michigan-48109, United States of America
| | - Arvind Rao
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas-77005, United States of America,
Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan-48109, United States of America,
Department of Biostatistics, University of Michigan, Ann Arbor, Michigan-48109, United States of America,
Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan-48109, United States of America,
Department of Radiology, University of Michigan, Ann Arbor, Michigan-48109, United States of America
| |
Collapse
|
70
|
Liu S, Du S, Gao S, Teng Y, Jin F, Zhang L. A delta-radiomic lymph node model using dynamic contrast enhanced MRI for the early prediction of axillary response after neoadjuvant chemotherapy in breast cancer patients. BMC Cancer 2023; 23:15. [PMID: 36604679 PMCID: PMC9817310 DOI: 10.1186/s12885-022-10496-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/29/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The objective of this paper is to explore the value of a delta-radiomic model of the axillary lymph node (ALN) using dynamic contrast-enhanced (DCE) MRI for early prediction of the axillary pathological complete response (pCR) of breast cancer patients after neoadjuvant chemotherapy (NAC). METHODS A total of 120 patients with ALN-positive breast cancer who underwent breast MRI before and after the first cycle of NAC between October 2018 and May 2021 were prospectively included in this study. Patients were divided into a training (n = 84) and validation (n = 36) cohort based on the temporal order of their treatments. Radiomic features were extracted from the largest slice of targeted ALN on DCE-MRI at pretreatment and after one cycle of NAC, and their changes (delta-) were calculated and recorded. Logistic regression was then applied to build radiomic models using the pretreatment (pre-), first-cycle(1st-), and changes (delta-) radiomic features separately. A clinical model was also built and combined with the radiomic models. The models were evaluated by discrimination, calibration, and clinical application and compared using DeLong test. RESULTS Among the three radiomic models, the ALN delta-radiomic model performed the best with AUCs of 0.851 (95% CI: 0.770-0.932) and 0.822 (95% CI: 0.685-0.958) in the training and validation cohorts, respectively. The clinical model yielded moderate AUCs of 0.742 (95% CI: 0.637-0.846) and 0.723 (95% CI: 0.550-0.896), respectively. After combining clinical features to the delta-radiomics model, the efficacy of the combined model (AUC = 0.932) in the training cohort was significantly higher than that of both the delta-radiomic model (Delong p = 0.017) and the clinical model (Delong p < 0.001) individually. Additionally, in the validation cohort, the combined model had the highest AUC (0.859) of any of the models we tested although this was not statistically different from any other individual model's validation AUC. Calibration and decision curves showed a good agreement and a high clinical benefit for the combined model. CONCLUSION This preliminary study indicates that ALN-based delta-radiomic model combined with clinical features is a promising strategy for the early prediction of downstaging ALN status after NAC. Future axillary MRI applications need to be further explored.
Collapse
Affiliation(s)
- Shasha Liu
- grid.412636.40000 0004 1757 9485Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001 China
| | - Siyao Du
- grid.412636.40000 0004 1757 9485Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001 China
| | - Si Gao
- grid.412636.40000 0004 1757 9485Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001 China
| | - Yuee Teng
- grid.412636.40000 0004 1757 9485Departments of Medical Oncology and Thoracic Surgery, The First Hospital of China Medical University, Shenyang, 110001 China
| | - Feng Jin
- grid.412636.40000 0004 1757 9485Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, 110001 China
| | - Lina Zhang
- grid.412636.40000 0004 1757 9485Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001 China
| |
Collapse
|
71
|
Ruan Y, He L, Chen J, Wang J, Zhao S, Guo X, Xie Y, Cai Z, Shen X, Li C. Three-dimensional core-shell alginate microsphere for cancer hypoxia simulation in vitro. Front Bioeng Biotechnol 2023; 11:1174206. [PMID: 37113672 PMCID: PMC10126516 DOI: 10.3389/fbioe.2023.1174206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
Hypoxia is one of the major causes of cancer resistance and metastasis. Currently, it is still lack of convenient ways to simulate the in vivo hypoxic tumor microenvironment (TME) under normoxia in vitro. In this study, based on multi-polymerized alginate, we established a three-dimensional culture system with a core-shell structure (3d-ACS), which prevents oxygen diffusion to a certain extent, thereby simulating the hypoxic TME in vivo. The cell activity, hypoxia inducible factor (HIF) expression, drug resistance, and the related gene and protein changes of the gastric cancer (GC) cells were investigated in vitro and in vivo. The results demonstrated that the GC cells formed organoid-like structures in the 3d-ACS and manifested more aggressive growth and decreased drug responses. Our study provides an accessible hypoxia platform in the laboratory with moderate configuration and it may be applied in studies of the hypoxia-induced drug resistances and other preclinical fields.
Collapse
Affiliation(s)
- Yejiao Ruan
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lingyun He
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiamin Chen
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jinfeng Wang
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shujing Zhao
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaoling Guo
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yao Xie
- Beijing Automation Control Equipment Institute, Beijing, China
| | - Zhenzhai Cai
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Zhenzhai Cai, ; Xian Shen, ; Chao Li,
| | - Xian Shen
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Zhenzhai Cai, ; Xian Shen, ; Chao Li,
| | - Chao Li
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Zhenzhai Cai, ; Xian Shen, ; Chao Li,
| |
Collapse
|
72
|
Bilal Masokano I, Pei Y, Chen J, Liu W, Xie S, Liu H, Feng D, He Q, Li W. Development and validation of MRI-based model for the preoperative prediction of macrotrabecular hepatocellular carcinoma subtype. Insights Imaging 2022; 13:201. [PMID: 36544029 PMCID: PMC9772375 DOI: 10.1186/s13244-022-01333-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/20/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Macrotrabecular hepatocellular carcinoma (MTHCC) has a poor prognosis and is difficult to diagnose preoperatively. The purpose is to build and validate MRI-based models to predict the MTHCC subtype. METHODS Two hundred eight patients with confirmed HCC were enrolled. Three models (model 1: clinicoradiologic model; model 2: fusion radiomics signature; model 3: combined model 1 and model 2) were built based on their clinical data and MR images to predict MTHCC in training and validation cohorts. The performance of the models was assessed using the area under the curve (AUC). The clinical utility of the models was estimated by decision curve analysis (DCA). A nomogram was constructed, and its calibration was evaluated. RESULTS Model 1 is easier to build than models 2 and 3, with a good AUC of 0.773 (95% CI 0.696-0.838) and 0.801 (95% CI 0.681-0.891) in predicting MTHCC in training and validation cohorts, respectively. It performed slightly superior to model 2 in both training (AUC 0.747; 95% CI 0.689-0.806; p = 0.548) and validation (AUC 0.718; 95% CI 0.618-0.810; p = 0.089) cohorts and was similar to model 3 in the validation (AUC 0.866; 95% CI 0.801-0.928; p = 0.321) but inferior in the training (AUC 0.889; 95% CI 0.851-0.926; p = 0.001) cohorts. The DCA of model 1 had a higher net benefit than the treat-all and treat-none strategy at a threshold probability of 10%. The calibration curves of model 1 closely aligned with the true MTHCC rates in the training (p = 0.355) and validation sets (p = 0.364). CONCLUSION The clinicoradiologic model has a good performance in diagnosing MTHCC, and it is simpler and easier to implement, making it a valuable tool for pretherapeutic decision-making in patients.
Collapse
Affiliation(s)
- Ismail Bilal Masokano
- grid.216417.70000 0001 0379 7164Department of Radiology, Xiangya Hospital, Central South University, No. 168 Xiangya Road, Kaifu District, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan China
| | - Yigang Pei
- grid.216417.70000 0001 0379 7164Department of Radiology, Xiangya Hospital, Central South University, No. 168 Xiangya Road, Kaifu District, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Juan Chen
- grid.216417.70000 0001 0379 7164Department of Radiology, Xiangya Hospital, Central South University, No. 168 Xiangya Road, Kaifu District, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Wenguang Liu
- grid.216417.70000 0001 0379 7164Department of Radiology, Xiangya Hospital, Central South University, No. 168 Xiangya Road, Kaifu District, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Simin Xie
- grid.216417.70000 0001 0379 7164Department of Radiology, Xiangya Hospital, Central South University, No. 168 Xiangya Road, Kaifu District, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Huaping Liu
- grid.216417.70000 0001 0379 7164Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan China
| | - Deyun Feng
- grid.216417.70000 0001 0379 7164Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Qiongqiong He
- grid.216417.70000 0001 0379 7164Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Wenzheng Li
- grid.216417.70000 0001 0379 7164Department of Radiology, Xiangya Hospital, Central South University, No. 168 Xiangya Road, Kaifu District, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| |
Collapse
|
73
|
Shen J, Du H, Wang Y, Du L, Yang D, Wang L, Zhu R, Zhang X, Wu J. A novel nomogram model combining CT texture features and urine energy metabolism to differentiate single benign from malignant pulmonary nodule. Front Oncol 2022; 12:1035307. [PMID: 36591441 PMCID: PMC9798090 DOI: 10.3389/fonc.2022.1035307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
Objective To investigate a novel diagnostic model for benign and malignant pulmonary nodule diagnosis based on radiomic and clinical features, including urine energy metabolism index. Methods A total of 107 pulmonary nodules were prospectively recruited and pathologically confirmed as malignant in 86 cases and benign in 21 cases. A chest CT scan and urine energy metabolism test were performed in all cases. A nomogram model was established in combination with radiomic and clinical features, including urine energy metabolism levels. The nomogram model was compared with the radiomic model and the clinical feature model alone to test its diagnostic validity, and receiver operating characteristic (ROC) curves were plotted to assess diagnostic validity. Results The nomogram was established using a logistic regression algorithm to combine radiomic features and clinical characteristics including urine energy metabolism results. The predictive performance of the nomogram was evaluated using the area under the ROC and calibration curve, which showed the best performance, area under the curve (AUC) = 0.982, 95% CI = 0.940-1.000, compared to clinical and radiomic models in the testing cohort. The clinical benefit of the model was assessed using the decision curve analysis (DCA) and using the nomogram for benign and malignant pulmonary nodules, and preoperative prediction of benign and malignant pulmonary nodules using nomograms showed better clinical benefit. Conclusion This study shows that a coupled model combining CT imaging features and clinical features (including urine energy metabolism) in combination with the nomogram model has higher diagnostic performance than the radiomic and clinical models alone, suggesting that the combination of both methods is more advantageous in identifying benign and malignant pulmonary nodules.
Collapse
Affiliation(s)
- Jing Shen
- Graduate School, Tianjin Medical University, Tianjin, China,Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Hai Du
- Graduate School, Tianjin Medical University, Tianjin, China,Department of Radiology, Ordos Central Hospital, Ordos Inner Mongolia, China
| | - Yadong Wang
- School of Medicine, Dalian University, Dalian, China,Department of Research, Dalian Detecsen Biomedical Co., LTD, Dalian, China
| | - Lina Du
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China,Graduate School, Dalian Medical University, Dalian, China
| | - Dong Yang
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China,Graduate School, Dalian University, Dalian, China
| | - Lingwei Wang
- Department of Cardio-Thoracic Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ruiping Zhu
- Department of Pathology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Xiaohui Zhang
- College of Environment and Chemical Engineering, Dalian University, Dalian, China,*Correspondence: Jianlin Wu, ; Xiaohui Zhang,
| | - Jianlin Wu
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China,*Correspondence: Jianlin Wu, ; Xiaohui Zhang,
| |
Collapse
|
74
|
Makihara K, Yamaguchi M, Ito K, Sakaguchi K, Hori Y, Semba T, Funahashi Y, Fujii H, Terada Y. New Cluster Analysis Method for Quantitative Dynamic Contrast-Enhanced MRI Assessing Tumor Heterogeneity Induced by a Tumor-Microenvironmental Ameliorator (E7130) Treatment to a Breast Cancer Mouse Model. J Magn Reson Imaging 2022; 56:1820-1831. [PMID: 35524730 DOI: 10.1002/jmri.28226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) can provide insight into tumor perfusion. However, a method that can quantitatively measure the intra-tumor distribution of tumor voxel clusters with a distinct range of Ktrans and ve values remains insufficiently explored. HYPOTHESIS Two-dimensional cluster analysis may quantify the distribution of a tumor voxel subregion with a distinct range of Ktrans and ve values in human breast cancer xenografts. STUDY TYPE Prospective longitudinal study. ANIMAL MODEL Twenty-two female athymic nude mice with MCF-7 xenograft, treated with E7130, a tumor-microenvironmental ameliorator, or saline. FIELD STRENGTH/SEQUENCE 9.4 Tesla, turbo rapid acquisition with relaxation enhancement, and spoiled gradient-echo sequences. ASSESSMENT We performed two-dimensional k-means clustering to identify tumor voxel clusters with a distinct range of Ktrans and ve values on Days 0, 2, and 5 after treatment, calculated the ratio of the number of tumor voxels in each cluster to the total number of tumor voxels, and measured the normalized distances defined as the ratio of the distance between each tumor voxel and the nearest tumor margin to a tumor radius. STATISTICAL TESTS Unpaired t-tests, Dunnett's multiple comparison tests, and Chi-squared test were used. RESULTS The largest and second largest clusters constituted 44.4% and 27.5% of all tumor voxels with cluster centroid values of Ktrans at 0.040 min-1 and 0.116 min-1 , and ve at 0.131 and 0.201, respectively. At baseline (Day 0), the average normalized distances for the largest and second largest clusters were 0.33 and 0.24, respectively. E7130-treated group showed the normalized distance of the initial largest cluster decreasing to 0.25, while that of the second largest cluster increasing to 0.31. Saline-treated group showed no change. DATA CONCLUSION A two-dimensional cluster analysis might quantify the spatial distribution of a tumor subregion with a distinct range of Ktrans and ve values. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Kazuyuki Makihara
- Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan.,Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Masayuki Yamaguchi
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Ken Ito
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Oncology Tsukuba Research Development, Discovery, Medicine Creation, Eisai Co., Ltd., Tsukuba-shi, Japan
| | - Kazuya Sakaguchi
- Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan.,Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Yusaku Hori
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Oncology Tsukuba Research Development, Discovery, Medicine Creation, Eisai Co., Ltd., Tsukuba-shi, Japan
| | - Taro Semba
- Oncology Tsukuba Research Development, Discovery, Medicine Creation, Eisai Co., Ltd., Tsukuba-shi, Japan
| | - Yasuhiro Funahashi
- Lenvima Co-Global Lead, Oncology Business Group, Eisai Co., Ltd., Woodcliff Lake, New Jersey, USA
| | - Hirofumi Fujii
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Yasuhiko Terada
- Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan.,Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
75
|
Waqar M, Van Houdt PJ, Hessen E, Li KL, Zhu X, Jackson A, Iqbal M, O’Connor J, Djoukhadar I, van der Heide UA, Coope DJ, Borst GR. Visualising spatial heterogeneity in glioblastoma using imaging habitats. Front Oncol 2022; 12:1037896. [PMID: 36505856 PMCID: PMC9731157 DOI: 10.3389/fonc.2022.1037896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/31/2022] [Indexed: 11/26/2022] Open
Abstract
Glioblastoma is a high-grade aggressive neoplasm characterised by significant intra-tumoral spatial heterogeneity. Personalising therapy for this tumour requires non-invasive tools to visualise its heterogeneity to monitor treatment response on a regional level. To date, efforts to characterise glioblastoma's imaging features and heterogeneity have focussed on individual imaging biomarkers, or high-throughput radiomic approaches that consider a vast number of imaging variables across the tumour as a whole. Habitat imaging is a novel approach to cancer imaging that identifies tumour regions or 'habitats' based on shared imaging characteristics, usually defined using multiple imaging biomarkers. Habitat imaging reflects the evolution of imaging biomarkers and offers spatially preserved assessment of tumour physiological processes such perfusion and cellularity. This allows for regional assessment of treatment response to facilitate personalised therapy. In this review, we explore different methodologies to derive imaging habitats in glioblastoma, strategies to overcome its technical challenges, contrast experiences to other cancers, and describe potential clinical applications.
Collapse
Affiliation(s)
- Mueez Waqar
- Department of Neurosurgery, Geoffrey Jefferson Brain Research Centre, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - Petra J. Van Houdt
- Department of Radiation Oncology, the Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Eline Hessen
- Department of Radiation Oncology, the Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ka-Loh Li
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - Xiaoping Zhu
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - Alan Jackson
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
- Department of Neuroradiology, Geoffrey Jefferson Brain Research Centre, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Mudassar Iqbal
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - James O’Connor
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
- Department of Radiology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Ibrahim Djoukhadar
- Department of Neuroradiology, Geoffrey Jefferson Brain Research Centre, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Uulke A. van der Heide
- Department of Radiation Oncology, the Netherlands Cancer Institute, Amsterdam, Netherlands
| | - David J. Coope
- Department of Neurosurgery, Geoffrey Jefferson Brain Research Centre, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - Gerben R. Borst
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health and Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
76
|
Mohammadi F, Visagan S, Gross SM, Karginov L, Lagarde JC, Heiser LM, Meyer AS. A lineage tree-based hidden Markov model quantifies cellular heterogeneity and plasticity. Commun Biol 2022; 5:1258. [PMID: 36396800 PMCID: PMC9671968 DOI: 10.1038/s42003-022-04208-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Individual cells can assume a variety of molecular and phenotypic states and recent studies indicate that cells can rapidly adapt in response to therapeutic stress. Such phenotypic plasticity may confer resistance, but also presents opportunities to identify molecular programs that could be targeted for therapeutic benefit. Approaches to quantify tumor-drug responses typically focus on snapshot, population-level measurements. While informative, these methods lack lineage and temporal information, which are particularly critical for understanding dynamic processes such as cell state switching. As new technologies have become available to measure lineage relationships, modeling approaches will be needed to identify the forms of cell-to-cell heterogeneity present in these data. Here we apply a lineage tree-based adaptation of a hidden Markov model that employs single cell lineages as input to learn the characteristic patterns of phenotypic heterogeneity and state transitions. In benchmarking studies, we demonstrated that the model successfully classifies cells within experimentally-tractable dataset sizes. As an application, we analyzed experimental measurements in cancer and non-cancer cell populations under various treatments. We find evidence of multiple phenotypically distinct states, with considerable heterogeneity and unique drug responses. In total, this framework allows for the flexible modeling of single cell heterogeneity across lineages to quantify, understand, and control cell state switching.
Collapse
Affiliation(s)
- Farnaz Mohammadi
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Shakthi Visagan
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Sean M Gross
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Luka Karginov
- Department of Bioengineering, University of Illinois, Urbana Champaign, IL, USA
| | - J C Lagarde
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Aaron S Meyer
- Department of Bioengineering, University of California, Los Angeles, CA, USA.
- Department of Bioinformatics, University of California, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA.
| |
Collapse
|
77
|
Incremental benefits of size-zone matrix-based radiomics features for the prognosis of lung adenocarcinoma: advantage of spatial partitioning on tumor evaluation. Eur Radiol 2022; 32:7691-7699. [PMID: 35554645 DOI: 10.1007/s00330-022-08818-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/04/2022] [Accepted: 04/13/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Prognostic models of lung adenocarcinoma (ADC) can be built using radiomics features from various categories. The size-zone matrix (SZM) features have a strong biological basis related to tumor partitioning, but their incremental benefits have not been fully explored. In our study, we aimed to evaluate the incremental benefits of SZM features for the prognosis of lung ADC. METHODS A total of 298 patients were included and their pretreatment computed tomography images were analyzed in fivefold cross-validation. We built a risk model of overall survival using SZM features and compared it with a conventional radiomics risk model and a clinical variable-based risk model. We also compared it with other models incorporating various combinations of SZM features, other radiomics features, and clinical variables. A total of seven risk models were compared and evaluated using the hazard ratio (HR) on the left-out test fold. RESULTS As a baseline, the clinical variable risk model showed an HR of 2.739. Combining the radiomics signature with SZM feature was better (HR 4.034) than using radiomics signature alone (HR 3.439). Combining radiomics signature, SZM feature, and clinical variable (HR 6.524) fared better than just combining radiomics signature and clinical variables (HR 4.202). These results confirmed the added benefits of SZM features for prognosis in lung ADC. CONCLUSION Combining SZM feature with the radiomics signature was better than using the radiomics signature alone and the benefits of SZM features were maintained when clinical variables were added confirming the incremental benefits of SZM features for lung ADC prognosis. KEY POINTS • Size-zone matrix (SZM) features provide incremental benefits for the prognosis of lung adenocarcinoma. • Combining the radiomics signature with SZM features performed better than using a radiomics signature alone.
Collapse
|
78
|
Prognostic Value of [18F]-FDG PET/CT Radiomics Combined with Sarcopenia Status among Patients with Advanced Gastroesophageal Cancer. Cancers (Basel) 2022; 14:cancers14215314. [PMID: 36358733 PMCID: PMC9658937 DOI: 10.3390/cancers14215314] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/17/2022] Open
Abstract
We investigated, whether 18[18F]-FDG PET/CT-derived radiomics combined with sarcopenia measurements improves survival prognostication among patients with advanced, metastatic gastroesophageal cancer. In our study, 128 consecutive patients with advanced, metastatic esophageal and gastroesophageal cancer (n = 128; 26 females; 102 males; mean age 63.5 ± 11.7 years; age range: 29−91 years) undergoing 18[18F]-FDG PET/CT for staging between November 2008 and December 2019 were included. Segmentation of the primary tumor and radiomics analysis derived from PET and CT images was performed semi-automatically with a commonly used open-source software platform (LIFEX, Version 6.30, lifexsoft.org). Patients’ nutritional status was determined by measuring the skeletal muscle index (SMI) at the level of L3 on the CT component. Univariable and multivariable analyses were performed to establish a survival prediction model including radiomics, clinical data, and SMI score. Univariable Cox proportional hazards model revealed ECOG (<0.001) and bone metastasis (p = 0.028) to be significant clinical parameters for overall survival (OS) and progression free survival (PFS). Age (p = 0.017) was an additional prognostic factor for OS. Multivariable analysis showed improved prognostication for overall and progression free survival when adding sarcopenic status, PET and CT radiomics to the model with clinical parameters only. PET and CT radiomics derived from hybrid 18[18F]-FDG PET/CT combined with sarcopenia measurements and clinical parameters may improve survival prediction among patients with advanced, metastatic gastroesophageal cancer.
Collapse
|
79
|
Wang X, Li S, Lin X, Lu Y, Mao C, Ye Z, Li X, Koh TS, Liu J, Liu J, Ma X, Cheng J, Ning G, Yan Z, Hou Z. Evaluation of tracer kinetic parameters in cervical cancer using dynamic contrast-enhanced MRI as biomarkers in terms of biological relevance, diagnostic performance and inter-center variability. Front Oncol 2022; 12:958219. [PMID: 36324571 PMCID: PMC9620719 DOI: 10.3389/fonc.2022.958219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/04/2022] [Indexed: 11/18/2022] Open
Abstract
Objectives This study assessed the clinical value of parameters derived from dynamic contrast-enhanced (DCE) MRI with respect to correlation with angiogenesis and proliferation of cervical cancer, performance of diagnosis and reproducibility of DCE-MRI parameters across MRI scanners. Materials and Methods A total of 113 patients with cervical carcinoma from two centers were included in this retrospective study. The DCE data were centralized and processed using five tracer kinetic models (TKMs) (Tofts, Ex-Tofts, ATH, SC, and DP), yielding the following parameters: volume transfer constant (Ktrans), extravascular extracellular volume (Ve), fractional volume of vascular space (Vp), blood flow (Fp), and permeability surface area product (PS). CD34 counts and Ki-67 PI (proliferation index) of cervical cancer and normal cervix tissue were obtained using immunohistochemical staining in Center 1. Results CD34 count and Ki-67 PI in cervical cancer were significantly higher than in normal cervix tissue (p<0.05). Parameter Ve from each TKM was significantly smaller in cervical cancer tissue than in normal cervix tissue (p<0.05), indicating the higher proliferation of cervical cancer cells. Ve of each TKM attained the largest AUC to diagnose cervical cancer. The distributions of DCE parameters for both cervical cancer and normal cervix tissue were not significantly different between two centers (P>0.05). Conclusion Parameter Ve was similar to the expression of Ki-67 in revealing the proliferation of tissue cells, attained good performance in diagnosis of cervical cancer, and demonstrated consistent findings on measured values across centers.
Collapse
Affiliation(s)
- Xue Wang
- Department of Radiology, The Second Affiliated Hospital and Yuying Children′s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shujian Li
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianhui Lin
- Department of Pathology, The Second Affiliated Hospital and Yuying Children′s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Lu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children′s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chuanwan Mao
- Department of Radiology, The Second Affiliated Hospital and Yuying Children′s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhijun Ye
- Department of Radiology, The Second Affiliated Hospital of Sichuan University, Chengdu, China
| | - Xuesheng Li
- Department of Radiology, The Second Affiliated Hospital of Sichuan University, Chengdu, China
| | - Tong-San Koh
- Department of Oncologic Imaging, National Cancer Center, Singapore, Singapore
- The department of Jiangsu Key Laboratory of Medical Optics, Duke-National University of Singapore (NUS) Graduate Medical School, Singapore, Singapore
| | - Jie Liu
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingjing Liu
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyue Ma
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gang Ning
- Department of Radiology, The Second Affiliated Hospital of Sichuan University, Chengdu, China
| | - Zhihan Yan
- Department of Radiology, The Second Affiliated Hospital and Yuying Children′s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zujun Hou
- Department of Radiology, The Second Affiliated Hospital and Yuying Children′s Hospital of Wenzhou Medical University, Wenzhou, China
- Jiangsu Key Laboratory of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
- *Correspondence: Zujun Hou,
| |
Collapse
|
80
|
A Machine Learning Model Based on Unsupervised Clustering Multihabitat to Predict the Pathological Grading of Meningiomas. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8955227. [PMID: 36132071 PMCID: PMC9484898 DOI: 10.1155/2022/8955227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022]
Abstract
Purpose We aim to develop and validate a machine learning model by enhanced MRI to determine the pathological grading of meningiomas with unsupervised clustering image analysis method, which are multihabitat to reflect the inherent heterogeneity of tumors. Materials and Methods A total of 120 patients with meningiomas confirmed by postoperative pathology were included in the study, including 60 patients with low-grade meningiomas (WHO grade I) and 60 patients with high-grade meningiomas (WHO grade II and WHO grade III). All patients underwent complete head enhanced magnetic resonance scans before surgery or any anti-tumor treatment. Enrolled patients in the group received surgical resection and obtained postoperative pathological data. The patients in the training group (84 people) and the test group (36 people) were randomly divided into two groups according to the ratio of 7 to 3. Multi-habitat features were extracted from MRI images based on enhanced T1. Machine learning method was used to model, which was used to distinguish high-grade meningioma from low-grade meningioma. At the same time, the obtained machine learning model was calibrated and evaluated. Results In patients with low-grade meningioma and high-grade meningioma, we found significant differences in Silhouette coefficient (P<0.05). In the machine learning model, the area under the curve was 0.838 in the training group (sensitivity, 67.65%; specificity, 88.82%) and 0.73 in the test group (sensitivity, 69.05%; specificity, 71.43%). After the analysis of calibration curve and decision curve analysis, the model had shown the potential of great application value. Conclusions Multi-habitat analysis based on enhanced MRI (T1) could accurately predict the pathological grading of meningiomas. This unsupervised image-based method could reflect the direct heterogeneity between high-grade meningiomas and low-grade meningiomas, which is of great significance for patients' treatment and prevention of recurrence.
Collapse
|
81
|
Somasundaram A, Vállez García D, Pfaehler E, van Sluis J, Dierckx RAJO, de Vries EGE, Boellaard R. Mitigation of noise-induced bias of PET radiomic features. PLoS One 2022; 17:e0272643. [PMID: 36006959 PMCID: PMC9409510 DOI: 10.1371/journal.pone.0272643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/22/2022] [Indexed: 11/20/2022] Open
Abstract
Introduction One major challenge in PET radiomics is its sensitivity to noise. Low signal-to-noise ratio (SNR) affects not only the precision but also the accuracy of quantitative metrics extracted from the images resulting in noise-induced bias. This phantom study aims to identify the radiomic features that are robust to noise in terms of precision and accuracy and to explore some methods that might help to correct noise-induced bias. Methods A phantom containing three 18F-FDG filled 3D printed inserts, reflecting heterogeneous tracer uptake and realistic tumor shapes, was used in the study. The three different phantom inserts were filled and scanned with three different tumor-to-background ratios, simulating a total of nine different tumors. From the 40-minute list-mode data, ten frames each for 5 s, 10 s, 30 s, and 120 s frame duration were reconstructed to generate images with different noise levels. Under these noise conditions, the precision and accuracy of the radiomic features were analyzed using intraclass correlation coefficient (ICC) and similarity distance metric (SDM) respectively. Based on the ICC and SDM values, the radiomic features were categorized into four groups: poor, moderate, good, and excellent precision and accuracy. A “difference image” created by subtracting two statistically equivalent replicate images was used to develop a model to correct the noise-induced bias. Several regression methods (e.g., linear, exponential, sigmoid, and power-law) were tested. The best fitting model was chosen based on Akaike information criteria. Results Several radiomic features derived from low SNR images have high repeatability, with 68% of radiomic features having ICC ≥ 0.9 for images with a frame duration of 5 s. However, most features show a systematic bias that correlates with the increase in noise level. Out of 143 features with noise-induced bias, the SDM values were improved based on a regression model (53 features to excellent and 67 to good) indicating that the noise-induced bias of these features can be, at least partially, corrected. Conclusion To have a predictive value, radiomic features should reflect tumor characteristics and be minimally affected by noise. The present study has shown that it is possible to correct for noise-induced bias, at least in a subset of the features, using a regression model based on the local image noise estimates.
Collapse
Affiliation(s)
- Ananthi Somasundaram
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Groningen, The Netherlands
- * E-mail:
| | - David Vállez García
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Groningen, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC–Location VU University Medical Center, Amsterdam, The Netherlands
| | - Elisabeth Pfaehler
- Department of Nuclear Medicine, University Hospital Juelich, Aachen, Germany
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudi A. J. O. Dierckx
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth G. E. de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Groningen, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC–Location VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
82
|
Deng PZ, Zhao BG, Huang XH, Xu TF, Chen ZJ, Wei QF, Liu XY, Guo YQ, Yuan SG, Liao WJ. Preoperative contrast-enhanced computed tomography-based radiomics model for overall survival prediction in hepatocellular carcinoma. World J Gastroenterol 2022; 28:4376-4389. [PMID: 36159012 PMCID: PMC9453776 DOI: 10.3748/wjg.v28.i31.4376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/14/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common primary liver malignancy with a rising incidence worldwide. The prognosis of HCC patients after radical resection remains poor. Radiomics is a novel machine learning method that extracts quantitative features from medical images and provides predictive information of cancer, which can assist with cancer diagnosis, therapeutic decision-making and prognosis improvement.
AIM To develop and validate a contrast-enhanced computed tomography-based radiomics model for predicting the overall survival (OS) of HCC patients after radical hepatectomy.
METHODS A total of 150 HCC patients were randomly divided into a training cohort (n = 107) and a validation cohort (n = 43). Radiomics features were extracted from the entire tumour lesion. The least absolute shrinkage and selection operator algorithm was applied for the selection of radiomics features and the construction of the radiomics signature. Univariate and multivariate Cox regression analyses were used to identify the independent prognostic factors and develop the predictive nomogram, incorporating clinicopathological characteristics and the radiomics signature. The accuracy of the nomogram was assessed with the concordance index, receiver operating characteristic (ROC) curve and calibration curve. The clinical utility was evaluated by decision curve analysis (DCA). Kaplan–Meier methodology was used to compare the survival between the low- and high-risk subgroups.
RESULTS In total, seven radiomics features were selected to construct the radiomics signature. According to the results of univariate and multivariate Cox regression analyses, alpha-fetoprotein (AFP), neutrophil-to-lymphocyte ratio (NLR) and radiomics signature were included to build the nomogram. The C-indices of the nomogram in the training and validation cohorts were 0.736 and 0.774, respectively. ROC curve analysis for predicting 1-, 3-, and 5-year OS confirmed satisfactory accuracy [training cohort, area under the curve (AUC) = 0.850, 0.791 and 0.823, respectively; validation cohort, AUC = 0.905, 0.884 and 0.911, respectively]. The calibration curve analysis indicated a good agreement between the nomogram-prediction and actual survival. DCA curves suggested that the nomogram had more benefit than traditional staging system models. Kaplan–Meier survival analysis indicated that patients in the low-risk group had longer OS and disease-free survival (all P < 0.0001).
CONCLUSION The nomogram containing the radiomics signature, NLR and AFP is a reliable tool for predicting the OS of HCC patients.
Collapse
Affiliation(s)
- Peng-Zhan Deng
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Bi-Geng Zhao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Xian-Hui Huang
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Ting-Feng Xu
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Zi-Jun Chen
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Qiu-Feng Wei
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Xiao-Yi Liu
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Yu-Qi Guo
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Sheng-Guang Yuan
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Wei-Jia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
83
|
Li G, Wu X, Ma X. Artificial intelligence in radiotherapy. Semin Cancer Biol 2022; 86:160-171. [PMID: 35998809 DOI: 10.1016/j.semcancer.2022.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/18/2022] [Indexed: 11/19/2022]
Abstract
Radiotherapy is a discipline closely integrated with computer science. Artificial intelligence (AI) has developed rapidly over the past few years. With the explosive growth of medical big data, AI promises to revolutionize the field of radiotherapy through highly automated workflow, enhanced quality assurance, improved regional balances of expert experiences, and individualized treatment guided by multi-omics. In addition to independent researchers, the increasing number of large databases, biobanks, and open challenges significantly facilitated AI studies on radiation oncology. This article reviews the latest research, clinical applications, and challenges of AI in each part of radiotherapy including image processing, contouring, planning, quality assurance, motion management, and outcome prediction. By summarizing cutting-edge findings and challenges, we aim to inspire researchers to explore more future possibilities and accelerate the arrival of AI radiotherapy.
Collapse
Affiliation(s)
- Guangqi Li
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Xin Wu
- Head & Neck Oncology ward, Division of Radiotherapy Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Xuelei Ma
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China.
| |
Collapse
|
84
|
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: 5.5] [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
|
85
|
Shi J, Zhao Z, Jiang T, Ai H, Liu J, Chen X, Luo Y, Fan H, Jiang X. A deep learning approach with subregion partition in MRI image analysis for metastatic brain tumor. Front Neuroinform 2022; 16:973698. [PMID: 35991287 PMCID: PMC9382021 DOI: 10.3389/fninf.2022.973698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeTo propose a deep learning network with subregion partition for predicting metastatic origins and EGFR/HER2 status in patients with brain metastasis.MethodsWe retrospectively enrolled 140 patients with clinico-pathologically confirmed brain metastasis originated from primary NSCLC (n = 60), breast cancer (BC, n = 60) and other tumor types (n = 20). All patients underwent contrast-enhanced brain MRI scans. The brain metastasis was subdivided into phenotypically consistent subregions using patient-level and population-level clustering. A residual network with a global average pooling layer (RN-GAP) was proposed to calculate deep learning-based features. Features from each subregion were selected with least absolute shrinkage and selection operator (LASSO) to build logistic regression models (LRs) for predicting primary tumor types (LR-NSCLC for the NSCLC origin and LR-BC for the BC origin), EGFR mutation status (LR-EGFR) and HER2 status (LR-HER2).ResultsThe brain metastasis can be partitioned into a marginal subregion (S1) and an inner subregion (S2) in the MRI image. The developed models showed good predictive performance in the training (AUCs, LR-NSCLC vs. LR-BC vs. LR-EGFR vs. LR-HER2, 0.860 vs. 0.909 vs. 0.850 vs. 0.900) and validation (AUCs, LR-NSCLC vs. LR-BC vs. LR-EGFR vs. LR-HER2, 0.819 vs. 0.872 vs. 0.750 vs. 0.830) set.ConclusionOur proposed deep learning network with subregion partitions can accurately predict metastatic origins and EGFR/HER2 status of brain metastasis, and hence may have the potential to be non-invasive and preoperative new markers for guiding personalized treatment plans in patients with brain metastasis.
Collapse
Affiliation(s)
- Jiaxin Shi
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, China
| | - Zilong Zhao
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Tao Jiang
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, China
| | - Hua Ai
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, China
| | - Jiani Liu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Xinpu Chen
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, China
| | - Yahong Luo
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Huijie Fan
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
- *Correspondence: Huijie Fan,
| | - Xiran Jiang
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, China
- Xiran Jiang,
| |
Collapse
|
86
|
Machine learning radiomics can predict early liver recurrence after resection of intrahepatic cholangiocarcinoma. HPB (Oxford) 2022; 24:1341-1350. [PMID: 35283010 PMCID: PMC9355916 DOI: 10.1016/j.hpb.2022.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Most patients recur after resection of intrahepatic cholangiocarcinoma (IHC). We studied whether machine-learning incorporating radiomics and tumor size could predict intrahepatic recurrence within 1-year. METHODS This was a retrospective analysis of patients with IHC resected between 2000 and 2017 who had evaluable computed tomography imaging. Texture features (TFs) were extracted from the liver, tumor, and future liver remnant (FLR). Random forest classification using training (70.3%) and validation cohorts (29.7%) was used to design a predictive model. RESULTS 138 patients were included for analysis. Patients with early recurrence had a larger tumor size (7.25 cm [IQR 5.2-8.9] vs. 5.3 cm [IQR 4.0-7.2], P = 0.011) and a higher rate of lymph node metastasis (28.6% vs. 11.6%, P = 0.041), but were not more likely to have multifocal disease (21.4% vs. 17.4%, P = 0.643). Three TFs from the tumor, FD1, FD30, and IH4 and one from the FLR, ACM15, were identified by feature selection. Incorporation of TFs and tumor size achieved the highest AUC of 0.84 (95% CI 0.73-0.95) in predicting recurrence in the validation cohort. CONCLUSION This study demonstrates that radiomics and machine-learning can reliably predict patients at risk for early intrahepatic recurrence with good discrimination accuracy.
Collapse
|
87
|
Zhang X, Zhao J, Zhang Q, Wang S, Zhang J, An J, Xie L, Yu X, Zhao X. MRI-based radiomics value for predicting the survival of patients with locally advanced cervical squamous cell cancer treated with concurrent chemoradiotherapy. Cancer Imaging 2022; 22:35. [PMID: 35842679 PMCID: PMC9287951 DOI: 10.1186/s40644-022-00474-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To investigate the magnetic resonance imaging (MRI)-based radiomics value in predicting the survival of patients with locally advanced cervical squamous cell cancer (LACSC) treated with concurrent chemoradiotherapy (CCRT). METHODS A total of 185 patients (training group: n = 128; testing group: n = 57) with LACSC treated with CCRT between January 2014 and December 2018 were retrospectively enrolled in this study. A total of 400 radiomics features were extracted from T2-weighted imaging, apparent diffusion coefficient map, arterial- and delayed-phase contrast-enhanced MRI. Univariate Cox regression and least absolute shrinkage and selection operator Cox regression was applied to select radiomics features and clinical characteristics that could independently predict progression-free survival (PFS) and overall survival (OS). The predictive capability of the prediction model was evaluated using Harrell's C-index. Nomograms and calibration curves were then generated. Survival curves were generated using the Kaplan-Meier method, and the log-rank test was used for comparison. RESULTS The radiomics score achieved significantly better predictive performance for the estimation of PFS (C-index, 0.764 for training and 0.762 for testing) and OS (C-index, 0.793 for training and 0.750 for testing), compared with the 2018 FIGO staging system (C-index for PFS, 0.657 for training and 0.677 for testing; C-index for OS, 0.665 for training and 0.633 for testing) and clinical-predicting model (C-index for PFS, 0.731 for training and 0.725 for testing; C-index for OS, 0.708 for training and 0.693 for testing) (P < 0.05). The combined model constructed with T stage, lymph node metastasis position, and radiomics score achieved the best performance for the estimation of PFS (C-index, 0.792 for training and 0.809 for testing) and OS (C-index, 0.822 for training and 0.785 for testing), which were significantly higher than those of the radiomics score (P < 0.05). CONCLUSIONS The MRI-based radiomics score could provide effective information in predicting the PFS and OS in patients with LACSC treated with CCRT. The combined model (including MRI-based radiomics score and clinical characteristics) showed the best prediction performance.
Collapse
Affiliation(s)
- Xiaomiao Zhang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingwei Zhao
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qi Zhang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | | | - Jieying Zhang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jusheng An
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lizhi Xie
- GE Healthcare, MR Research, Beijing, China
| | - Xiaoduo Yu
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Xinming Zhao
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
88
|
Wolf Y, Samuels Y. Intratumor Heterogeneity and Antitumor Immunity Shape One Another Bidirectionally. Clin Cancer Res 2022; 28:2994-3001. [PMID: 35380639 PMCID: PMC9306293 DOI: 10.1158/1078-0432.ccr-21-1355] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/10/2022] [Accepted: 03/28/2022] [Indexed: 01/07/2023]
Abstract
Over the last decade, it has become clear that the genomic landscapes of tumors profoundly impact their immunogenicity and how tumor cells interact with immune cells. Whereas past discoveries mainly focused on the interplay between tumor immunogenicity and tumor mutational burden (TMB), under the assumption that a higher mutation load would give rise to a better patient response to immune checkpoint blockade therapies, we and others have underlined intratumor heterogeneity (ITH) as an important determinant of the magnitude of the antitumor response and the nature of the tumor microenvironment. In this review, we define TMB versus ITH and how the two factors are being inferred from data, examine key findings in the cancer immunogenomics literature deciphering the complex cross-talk between TMB, ITH, and antitumor immunity in human cancers and in vivo models, and discuss the mutual influence of ITH and immunity-how the antitumor response can give rise to tumors with higher ITH, and how higher ITH can put shackles on the antitumor response.
Collapse
Affiliation(s)
- Yochai Wolf
- Ella Lemelbaum Institute for Immuno-Oncology and Skin Cancer, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Corresponding Authors: Yochai Wolf, Ella Lemelbaum Institute for Immuno-Oncology and Skin Cancer, Sheba Medical Center, Tel Hashomer, Ramat Gan 5265601, Israel. E-mail: ; and Yardena Samuels, Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 761000, Israel. E-mail:
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.,Corresponding Authors: Yochai Wolf, Ella Lemelbaum Institute for Immuno-Oncology and Skin Cancer, Sheba Medical Center, Tel Hashomer, Ramat Gan 5265601, Israel. E-mail: ; and Yardena Samuels, Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 761000, Israel. E-mail:
| |
Collapse
|
89
|
Mirón Mombiela R, Borrás C. The Usefulness of Radiomics Methodology for Developing Descriptive and Prognostic Image-Based Phenotyping in the Aging Population: Results From a Small Feasibility Study. FRONTIERS IN AGING 2022; 3:853671. [PMID: 35821818 PMCID: PMC9261370 DOI: 10.3389/fragi.2022.853671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/01/2022] [Indexed: 12/25/2022]
Abstract
Background: Radiomics is an emerging field that translates medical images into quantitative data to enable phenotypic profiling of human disease. In this retrospective study, we asked whether it is possible to use image-based phenotyping to describe and determine prognostic factors in the aging population. Methods: A radiomic frailty cohort with 101 patients was included in the analysis (65 ± 15 years, 55 men). A total of 44 texture features were extracted from the segmented muscle area of the ultrasound images of the anterior thigh. Univariate and multivariate analyses were performed to assess the image data sets and clinical data. Results: Our results showed that the heterogeneity of muscle was associated with an increased incidence of hearing impairment, stroke, myocardial infarction, dementia/memory loss, and falls in the following two years. Regression analysis revealed a muscle radiomic model with 87.1% correct predictive value with good sensitivity and moderate specificity (p = 0.001). Conclusion: It is possible to develop and identify image-based phenotypes in the elderly population. The muscle radiomic model needs to further be validated. Future studies correlated with biological data (genomics, transcriptomics, metabolomics, etc.) will give further insights into the biological basis and molecular processes of the developed radiomic model.
Collapse
Affiliation(s)
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, Institute of Health Research-INCLIVA, University of Valencia, and CIBERFES, Valencia, Spain
| |
Collapse
|
90
|
Ho TKK, Gwak J. Feature-level ensemble approach for COVID-19 detection using chest X-ray images. PLoS One 2022; 17:e0268430. [PMID: 35834442 PMCID: PMC9282557 DOI: 10.1371/journal.pone.0268430] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/01/2022] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS CoV-2), also known as the coronavirus disease 2019 (COVID-19), has threatened many human beings around the world and capsized economies at unprecedented magnitudes. Therefore, the detection of this disease using chest X-ray modalities has played a pivotal role in producing fast and accurate medical diagnoses, especially in countries that are unable to afford laboratory testing kits. However, identifying and distinguishing COVID-19 from virtually similar thoracic abnormalities utilizing medical images is challenging because it is time-consuming, demanding, and susceptible to human-based errors. Therefore, artificial-intelligence-driven automated diagnoses, which excludes direct human intervention, may potentially be used to achieve consistently accurate performances. In this study, we aimed to (i) obtain a customized dataset composed of a relatively small number of images collected from publicly available datasets; (ii) present the efficient integration of the shallow handcrafted features obtained from local descriptors, radiomics features specialized for medical images, and deep features aggregated from pre-trained deep learning architectures; and (iii) distinguish COVID-19 patients from healthy controls and pneumonia patients using a collection of conventional machine learning classifiers. By conducting extensive experiments, we demonstrated that the feature-based ensemble approach provided the best classification metrics, and this approach explicitly outperformed schemes that used only either local, radiomic, or deep features. In addition, our proposed method achieved state-of-the-art multi-class classification results compared to the baseline reference for the currently available COVID-19 datasets.
Collapse
Affiliation(s)
- Thi Kieu Khanh Ho
- Department of Software, Korea National University of Transportation, Chungju, South Korea
| | - Jeonghwan Gwak
- Department of Software, Korea National University of Transportation, Chungju, South Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, South Korea
- Department of AI Robotics Engineering, Korea National University of Transportation, Chungju, South Korea
- Department of IT & Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, South Korea
- * E-mail:
| |
Collapse
|
91
|
Bailo M, Pecco N, Callea M, Scifo P, Gagliardi F, Presotto L, Bettinardi V, Fallanca F, Mapelli P, Gianolli L, Doglioni C, Anzalone N, Picchio M, Mortini P, Falini A, Castellano A. Decoding the Heterogeneity of Malignant Gliomas by PET and MRI for Spatial Habitat Analysis of Hypoxia, Perfusion, and Diffusion Imaging: A Preliminary Study. Front Neurosci 2022; 16:885291. [PMID: 35911979 PMCID: PMC9326318 DOI: 10.3389/fnins.2022.885291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTumor heterogeneity poses major clinical challenges in high-grade gliomas (HGGs). Quantitative radiomic analysis with spatial tumor habitat clustering represents an innovative, non-invasive approach to represent and quantify tumor microenvironment heterogeneity. To date, habitat imaging has been applied mainly on conventional magnetic resonance imaging (MRI), although virtually extendible to any imaging modality, including advanced MRI techniques such as perfusion and diffusion MRI as well as positron emission tomography (PET) imaging.ObjectivesThis study aims to evaluate an innovative PET and MRI approach for assessing hypoxia, perfusion, and tissue diffusion in HGGs and derive a combined map for clustering of intra-tumor heterogeneity.Materials and MethodsSeventeen patients harboring HGGs underwent a pre-operative acquisition of MR perfusion (PWI), Diffusion (dMRI) and 18F-labeled fluoroazomycinarabinoside (18F-FAZA) PET imaging to evaluate tumor vascularization, cellularity, and hypoxia, respectively. Tumor volumes were segmented on fluid-attenuated inversion recovery (FLAIR) and T1 post-contrast images, and voxel-wise clustering of each quantitative imaging map identified eight combined PET and physiologic MRI habitats. Habitats’ spatial distribution, quantitative features and histopathological characteristics were analyzed.ResultsA highly reproducible distribution pattern of the clusters was observed among different cases, particularly with respect to morphological landmarks as the necrotic core, contrast-enhancing vital tumor, and peritumoral infiltration and edema, providing valuable supplementary information to conventional imaging. A preliminary analysis, performed on stereotactic bioptic samples where exact intracranial coordinates were available, identified a reliable correlation between the expected microenvironment of the different spatial habitats and the actual histopathological features. A trend toward a higher representation of the most aggressive clusters in WHO (World Health Organization) grade IV compared to WHO III was observed.ConclusionPreliminary findings demonstrated high reproducibility of the PET and MRI hypoxia, perfusion, and tissue diffusion spatial habitat maps and correlation with disease-specific histopathological features.
Collapse
Affiliation(s)
- Michele Bailo
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Nicolò Pecco
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Paola Scifo
- Department of Nuclear Medicine, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Filippo Gagliardi
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Luca Presotto
- Department of Nuclear Medicine, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Federico Fallanca
- Department of Nuclear Medicine, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Paola Mapelli
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Nuclear Medicine, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Luigi Gianolli
- Department of Nuclear Medicine, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Nicoletta Anzalone
- Vita-Salute San Raffaele University, Milan, Italy
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Maria Picchio
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Nuclear Medicine, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Pietro Mortini
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Falini
- Vita-Salute San Raffaele University, Milan, Italy
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Antonella Castellano
- Vita-Salute San Raffaele University, Milan, Italy
- Neuroradiology Unit and CERMAC, IRCCS Ospedale San Raffaele, Milan, Italy
- *Correspondence: Antonella Castellano,
| |
Collapse
|
92
|
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
|
93
|
Brighi C, Verburg N, Koh ES, Walker A, Chen C, Pillay S, de Witt Hamer PC, Aly F, Holloway LC, Keall PJ, Waddington DE. Repeatability of radiotherapy dose-painting prescriptions derived from a multiparametric magnetic resonance imaging model of glioblastoma infiltration. Phys Imaging Radiat Oncol 2022; 23:8-15. [PMID: 35734265 PMCID: PMC9207284 DOI: 10.1016/j.phro.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 12/03/2022] Open
Abstract
Magnetic resonance imaging was used to derive dose-painting prescriptions in glioma. Dose prescriptions derived from magnetic resonance imaging are highly repeatable. Dose-painting plans are more repeatable than their dose prescriptions.
Background and purpose Glioblastoma (GBM) patients have a dismal prognosis. Tumours typically recur within months of surgical resection and post-operative chemoradiation. Multiparametric magnetic resonance imaging (mpMRI) biomarkers promise to improve GBM outcomes by identifying likely regions of infiltrative tumour in tumour probability (TP) maps. These regions could be treated with escalated dose via dose-painting radiotherapy to achieve higher rates of tumour control. Crucial to the technical validation of dose-painting using imaging biomarkers is the repeatability of the derived dose prescriptions. Here, we quantify repeatability of dose-painting prescriptions derived from mpMRI. Materials and methods TP maps were calculated with a clinically validated model that linearly combined apparent diffusion coefficient (ADC) and relative cerebral blood volume (rBV) or ADC and relative cerebral blood flow (rBF) data. Maps were developed for 11 GBM patients who received two mpMRI scans separated by a short interval prior to chemoradiation treatment. A linear dose mapping function was applied to obtain dose-painting prescription (DP) maps for each session. Voxel-wise and group-wise repeatability metrics were calculated for parametric, TP and DP maps within radiotherapy margins. Results DP maps derived from mpMRI were repeatable between imaging sessions (ICC > 0.85). ADC maps showed higher repeatability than rBV and rBF maps (Wilcoxon test, p = 0.001). TP maps obtained from the combination of ADC and rBF were the most stable (median ICC: 0.89). Conclusions Dose-painting prescriptions derived from a mpMRI model of tumour infiltration have a good level of repeatability and can be used to generate reliable dose-painting plans for GBM patients.
Collapse
|
94
|
Jiménez de los Santos ME, Reyes-Pérez JA, Domínguez Osorio V, Villaseñor-Navarro Y, Moreno-Astudillo L, Vela-Sarmiento I, Sollozo-Dupont I. Whole lesion histogram analysis of apparent diffusion coefficient predicts therapy response in locally advanced rectal cancer. World J Gastroenterol 2022; 28:2609-2624. [PMID: 35949349 PMCID: PMC9254137 DOI: 10.3748/wjg.v28.i23.2609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/25/2021] [Accepted: 04/25/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Whole-tumor apparent diffusion coefficient (ADC) histogram analysis is relevant to predicting the neoadjuvant chemoradiation therapy (nCRT) response in patients with locally advanced rectal cancer (LARC).
AIM To evaluate the performance of ADC histogram-derived parameters for predicting the outcomes of patients with LARC.
METHODS This is a single-center, retrospective study, which included 48 patients with LARC. All patients underwent a pre-treatment magnetic resonance imaging (MRI) scan for primary tumor staging and a second restaging MRI for response evaluation. The sample was distributed as follows: 18 responder patients (R) and 30 non-responders (non-R). Eight parameters derived from the whole-lesion histogram analysis (ADCmean, skewness, kurtosis, and ADC10th, 25th, 50th, 75th, 90th percentiles), as well as the ADCmean from the hot spot region of interest (ROI), were calculated for each patient before and after treatment. Then all data were compared between R and non-R using the Mann-Whitney U test. Two measures of diagnostic accuracy were applied: the receiver operating characteristic curve and the diagnostic odds ratio (DOR). We also reported intra- and interobserver variability by calculating the intraclass correlation coefficient (ICC).
RESULTS Post-nCRT kurtosis, as well as post-nCRT skewness, were significantly lower in R than in non-R (both P < 0.001, respectively). We also found that, after treatment, R had a larger loss of both kurtosis and skewness than non-R (∆%kurtosis and ∆skewness, P < 0.001). Other parameters that demonstrated changes between groups were post-nCRT ADC10th, ∆%ADC10th, ∆%ADCmean, and ROI ∆%ADCmean. However, the best diagnostic performance was achieved by ∆%kurtosis at a threshold of 11.85% (Area under the receiver operating characteristic curve [AUC] = 0.991, DOR = 376), followed by post-nCRT kurtosis = 0.78 × 10-3 mm2/s (AUC = 0.985, DOR = 375.3), ∆skewness = 0.16 (AUC = 0.885, DOR = 192.2) and post-nCRT skewness = 1.59 × 10-3 mm2/s (AUC = 0.815, DOR = 168.6). Finally, intraclass correlation coefficient analysis showed excellent intraobserver and interobserver agreement, ensuring the implementation of histogram analysis into routine clinical practice.
CONCLUSION Whole-tumor ADC histogram parameters, particularly kurtosis and skewness, are relevant biomarkers for predicting the nCRT response in LARC. Both parameters appear to be more reliable than ADCmean from one-slice ROI.
Collapse
Affiliation(s)
| | | | | | | | | | - Itzel Vela-Sarmiento
- Department of Gastrointestinal Surgery, National Cancer Institute, Mexico 14080, Mexico
| | | |
Collapse
|
95
|
Wang W, Jiao Y, Zhang L, Fu C, Zhu X, Wang Q, Gu Y. Multiparametric MRI-based radiomics analysis: differentiation of subtypes of cervical cancer in the early stage. Acta Radiol 2022; 63:847-856. [PMID: 33975448 DOI: 10.1177/02841851211014188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND There are significant differences in outcomes for different histological subtypes of cervical cancer (CC). Yet, it is difficult to distinguish CC subtypes using non-invasive methods. PURPOSE To investigate whether multiparametric magnetic resonance imaging (MRI)-based radiomics analysis can differentiate CC subtypes and explore tumor heterogeneity. MATERIAL AND METHODS This study retrospectively analyzed 96 patients with CC (squamous cell carcinoma [SCC] = 50, adenocarcinoma [AC] = 46) who underwent pelvic MRI before surgery. Radiomics features were extracted from the tumor volumes on five sequences (sagittal T2-weighted imaging [T2SAG], transverse T2-weighted imaging [T2TRA], sagittal contrast-enhanced T1-weighted imaging [CESAG], transverse contrast-enhanced T1-weighted imaging [CETRA], and apparent diffusion coefficient [ADC]). Clustering and logistic regression were used to examine the distinguishing capabilities of radiomics features extracted from five different MR sequences. RESULTS Among the 105 extracted radiomics features, there were 51, 38, 37, and 2 features that showed intergroup differences for T2SAG, T2TRA, ADC, and CESAG, respectively (all P < 0.05). AC had greater textural heterogeneity than SCC (P < 0.05). Upon unsupervised clustering of significantly different features, T2SAG achieved the highest accuracy (0.844; sensitivity = 0.920; specificity = 0.761). The largest area under the curve (AUC) for classification ability was 0.86 for T2SAG. Hence, the radiomics model from five combined MR sequences (AUC = 0.89; accuracy = 0.81; sensitivity = 0.67; specificity = 0.94) exhibited better differentiation ability than any MR sequence alone. CONCLUSION Multiparametric MRI-based radiomics models may be a promising method to differentiate AC and SCC. AC showed more heterogeneous features than SCC.
Collapse
Affiliation(s)
- Wei Wang
- Department of Radiology, Fudan University Shanghai Cancer Center (FUSCC), Shanghai, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - YiNing Jiao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| | - LiChi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| | - Caixia Fu
- MR Applications Development, Siemens Shenzhen Magnetic Resonance Ltd., Shenzhen, PR China
| | - XiaoLi Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
- Department of Pathology, Fudan University Shanghai Cancer Center (FUSCC), Shanghai, PR China
| | - Qian Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center (FUSCC), Shanghai, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| |
Collapse
|
96
|
ADC textural features in patients with single brain metastases improve clinical risk models. Clin Exp Metastasis 2022; 39:459-466. [PMID: 35394585 PMCID: PMC9117356 DOI: 10.1007/s10585-022-10160-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 02/28/2022] [Indexed: 11/03/2022]
Abstract
AIMS In this retrospective study we performed a quantitative textural analysis of apparant diffusion coefficient (ADC) images derived from diffusion weighted MRI (DW-MRI) of single brain metastases (BM) patients from different primary tumors and tested whether these imaging parameters may improve established clinical risk models. METHODS We identified 87 patients with single BM who had a DW-MRI at initial diagnosis. Applying image segmentation, volumes of contrast-enhanced lesions in T1 sequences, hyperintense T2 lesions (peritumoral border zone (T2PZ)) and tumor-free gray and white matter compartment (GMWMC) were generated and registered to corresponding ADC maps. ADC textural parameters were generated and a linear backward regression model was applied selecting imaging features in association with survival. A cox proportional hazard model with backward regression was fitted for the clinical prognostic models (diagnosis-specific graded prognostic assessment score (DS-GPA) and the recursive partitioning analysis (RPA)) including these imaging features. RESULTS Thirty ADC textural parameters were generated and linear backward regression identified eight independent imaging parameters which in combination predicted survival. Five ADC texture features derived from T2PZ, the volume of the T2PZ, the normalized mean ADC of the GMWMC as well as the mean ADC slope of T2PZ. A cox backward regression including the DS-GPA, RPA and these eight parameters identified two MRI features which improved the two risk scores (HR = 1.14 [1.05;1.24] for normalized mean ADC GMWMC and HR = 0.87 [0.77;0.97]) for ADC 3D kurtosis of the T2PZ.) CONCLUSIONS: Textural analysis of ADC maps in patients with single brain metastases improved established clinical risk models. These findings may aid to better understand the pathogenesis of BM and may allow selection of patients for new treatment options.
Collapse
|
97
|
Role of Texture Analysis in Oropharyngeal Carcinoma: A Systematic Review of the Literature. Cancers (Basel) 2022; 14:cancers14102445. [PMID: 35626048 PMCID: PMC9139172 DOI: 10.3390/cancers14102445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The incidence of squamous cell carcinomas of the oropharynx has rapidly increased in the last two decades due to human papilloma virus infection (HPV). HPV-positive and HPV-negative squamous cell tumours differ in radiological imaging, treatment, and prognosis; therefore, differential diagnosis is mandatory. Radiomics with texture analysis is an innovative technique that has been used increasingly in recent years to characterise the tissue heterogeneity of certain structures such as neoplasms or organs by measuring the spatial distribution of pixel values on radiological imaging. This review delineates the application of texture analysis in oropharyngeal tumours and explores how radiomics may potentially improve clinical decision-making. Abstract Human papilloma virus infection (HPV) is associated with the development of lingual and palatine tonsil carcinomas. Diagnosing, differentiating HPV-positive from HPV-negative cancers, and assessing the presence of lymph node metastases or recurrences by the visual interpretation of images is not easy. Texture analysis can provide structural information not perceptible to human eyes. A systematic literature search was performed on 16 February 2022 for studies with a focus on texture analysis in oropharyngeal cancers. We conducted the research on PubMed, Scopus, and Web of Science platforms. Studies were screened for inclusion according to the preferred reporting items for systematic reviews. Twenty-six studies were included in our review. Nineteen articles related specifically to the oropharynx and seven articles analysed the head and neck area with sections dedicated to the oropharynx. Six, thirteen, and seven articles used MRI, CT, and PET, respectively, as the imaging techniques by which texture analysis was performed. Regarding oropharyngeal tumours, this review delineates the applications of texture analysis in (1) the diagnosis, prognosis, and assessment of disease recurrence or persistence after therapy, (2) early differentiation of HPV-positive versus HPV-negative cancers, (3) the detection of cancers not visualised by imaging alone, and (4) the assessment of lymph node metastases from unknown primary carcinomas.
Collapse
|
98
|
Machine Learning-Based Radiomics for Prediction of Epidermal Growth Factor Receptor Mutations in Lung Adenocarcinoma. DISEASE MARKERS 2022; 2022:2056837. [PMID: 35578691 PMCID: PMC9107363 DOI: 10.1155/2022/2056837] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/13/2022] [Accepted: 04/23/2022] [Indexed: 12/20/2022]
Abstract
Identifying an epidermal growth factor receptor (EGFR) mutation is important because EGFR tyrosine kinase inhibitors are the first-line treatment of choice for patients with EGFR mutation-positive lung adenocarcinomas (LUAC). This study is aimed at developing and validating a radiomics-based machine learning (ML) approach to identify EGFR mutations in patients with LUAC. We retrospectively collected data from 201 patients with positive EGFR mutation LUAC (140 in the training cohort and 61 in the validation cohort). We extracted 1316 radiomics features from preprocessed CT images and selected 14 radiomics features and 1 clinical feature which were most relevant to mutations through filter method. Subsequently, we built models using 7 ML approaches and established the receiver operating characteristic (ROC) curve to assess the discriminating performance of these models. In terms of predicting EGFR mutation, the model derived from radiomics features and combined models (radiomics features and relevant clinical factors) had an AUC of 0.79 (95% confidence interval (CI): 0.77-0.82), 0.86 (0.87-0.88), respectively. Our study offers a radiomics-based ML model using filter methods to detect the EGFR mutation in patients with LUAC. This convenient and low-cost method may be of help to noninvasively identify patients before obtaining tumor sample for molecule testing.
Collapse
|
99
|
Shi F, Liu Y, Zhou X, Shen P, Xue R, Zhang M. Disitamab vedotin: a novel antibody-drug conjugates for cancer therapy. Drug Deliv 2022; 29:1335-1344. [PMID: 35506447 PMCID: PMC9090390 DOI: 10.1080/10717544.2022.2069883] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) regulates cell mitosis, proliferation, and apoptosis. Trastuzumab is a HER2-targeted monoclonal antibody (mAB), which can prolong the overall survival rate of patients with HER2 overexpression in later periods of gastric cancer and breast cancer. Although anti-HER2 monoclonal antibody has a curative effect, adjuvant chemotherapy is still necessary to upgrade the curative effect maximumly. Antibody-drug conjugate (ADC) is a kind of therapeutic drug that contains antigen-specific antibody and cytotoxic payload, which can improve the survival time of tumor patients. To date, there are several HER2-ADC products on the market, for which two anti-HER2 ADC (trastuzumab emtansine and trastuzumab deruxtecan) have been authorized by the FDA for distinct types of HER2-positive carcinoma in the breast. Disitamab vedotin (RC48) is a newly developed ADC drug targeting HER2 that is comprised of hertuzumab coupling monomethyl auristatin E (MMAE) via a cleavable linker. This paper aims to offer a general insight and summary of the mechanism of action and the currently completed and ongoing clinical studies of RC-48 in HER-2 positive solid tumors.
Collapse
Affiliation(s)
- Fan Shi
- State Key Laboratory of Military Stomatology, Department of General Dentistry and Emergency, School of Stomatology, Air Force Military Medical University, Xi'an, China
| | - Yanli Liu
- State Key Laboratory of Military Stomatology, Department of General Dentistry and Emergency, School of Stomatology, Air Force Military Medical University, Xi'an, China
| | - Xuexiao Zhou
- School of Stomatology of Qingdao University, Qingdao, China
| | - Pei Shen
- School of Stomatology of Qingdao University, Qingdao, China
| | - Ran Xue
- Department of Pharmacy, The First Affiliated Hospital of Xi 'an Jiaotong University, Chang'an District Hospital, Xi 'an, China
| | - Min Zhang
- State Key Laboratory of Military Stomatology, Department of General Dentistry and Emergency, School of Stomatology, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
100
|
Tar PD, Thacker NA, Babur M, Lipowska-Bhalla G, Cheung S, Little RA, Williams KJ, O’Connor JPB. Habitat Imaging of Tumors Enables High Confidence Sub-Regional Assessment of Response to Therapy. Cancers (Basel) 2022; 14:2159. [PMID: 35565288 PMCID: PMC9101368 DOI: 10.3390/cancers14092159] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Imaging biomarkers are used in therapy development to identify and quantify therapeutic response. In oncology, use of MRI, PET and other imaging methods can be complicated by spatially complex and heterogeneous tumor micro-environments, non-Gaussian data and small sample sizes. Linear Poisson Modelling (LPM) enables analysis of complex data that is quantitative and can operate in small data domains. We performed experiments in 5 mouse models to evaluate the ability of LPM to identify responding tumor habitats across a range of radiation and targeted drug therapies. We tested if LPM could identify differential biological response rates. We calculated the theoretical sample size constraints for applying LPM to new data. We then performed a co-clinical trial using small data to test if LPM could detect multiple therapeutics with both improved power and reduced animal numbers compared to conventional t-test approaches. Our data showed that LPM greatly increased the amount of information extracted from diffusion-weighted imaging, compared to cohort t-tests. LPM distinguished biological response rates between Calu6 tumors treated with 3 different therapies and between Calu6 tumors and 4 other xenograft models treated with radiotherapy. A simulated co-clinical trial using real data detected high precision per-tumor treatment effects in as few as 3 mice per cohort, with p-values as low as 1 in 10,000. These findings provide a route to simultaneously improve the information derived from preclinical imaging while reducing and refining the use of animals in cancer research.
Collapse
Affiliation(s)
- Paul David Tar
- Division of Cancer Sciences, University of Manchester, Manchester M13 9PT, UK; (P.D.T.); (G.L.-B.); (S.C.); (R.A.L.)
| | - Neil A. Thacker
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester M13 9PT, UK;
| | - Muhammad Babur
- Manchester Pharmacy School, Division of Pharmacy and Optometry, University of Manchester, Manchester M13 9PT, UK; (M.B.); (K.J.W.)
| | - Grazyna Lipowska-Bhalla
- Division of Cancer Sciences, University of Manchester, Manchester M13 9PT, UK; (P.D.T.); (G.L.-B.); (S.C.); (R.A.L.)
| | - Susan Cheung
- Division of Cancer Sciences, University of Manchester, Manchester M13 9PT, UK; (P.D.T.); (G.L.-B.); (S.C.); (R.A.L.)
| | - Ross A. Little
- Division of Cancer Sciences, University of Manchester, Manchester M13 9PT, UK; (P.D.T.); (G.L.-B.); (S.C.); (R.A.L.)
| | - Kaye J. Williams
- Manchester Pharmacy School, Division of Pharmacy and Optometry, University of Manchester, Manchester M13 9PT, UK; (M.B.); (K.J.W.)
| | - James P. B. O’Connor
- Division of Cancer Sciences, University of Manchester, Manchester M13 9PT, UK; (P.D.T.); (G.L.-B.); (S.C.); (R.A.L.)
- Department of Radiology, The Christie Hospital NHS Trust, Manchester M20 4BX, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| |
Collapse
|