1
|
Andreadis T, Chouchos K, Courcoutsakis N, Seimenis I, Koulouriotis D. Development of an Automated CAD System for Lesion Detection in DCE-MRI. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025:10.1007/s10278-025-01445-2. [PMID: 39979761 DOI: 10.1007/s10278-025-01445-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) has been recognized as an effective tool for early detection and characterization of breast lesions. This study proposes an automated computer-aided diagnosis (CAD) system to facilitate lesion detection in DCE-MRI. The system initially identifies and crops the breast tissue reducing the processed image region and, thus, resulting in lower computational burden. Then, Otsu's multilevel thresholding method is applied to detect and segment the suspicious regions of interest (ROIs), considering the dynamic enhancement changes across two post-contrast sequential phases. After segmentation, a two-stage false positive reduction process is applied. A rule-based stage is first applied, followed by the segmentation of control ROIs in the contralateral breast. A feature vector is then extracted from all ROIs and supervised classification is implemented using two classifiers (feed-forward backpropagation neural network (FFBPN) and support vector machine (SVM)). A dataset of 52 DCE-MRI exams was used for assessing the performance of the system in terms of accuracy, sensitivity, specificity, and precision. A total of 138 enhancing lesions were identified by an experienced radiologist and corresponded to CAD-detected ROIs. The system's overall sensitivity was 83% when the FFBPN classifier was used and 92% when the SVM was applied. Moreover, the calculated area under curve for the SVM classifier was 0.95. Both employed classifiers exhibited high performance in identifying enhancing lesions and in differentiating them from healthy parenchyma. Current results suggest that the employment of a CAD system can expedite lesion detection in DCE-MRI images and, therefore, further research over larger datasets is warranted.
Collapse
Affiliation(s)
- Theofilos Andreadis
- Department of Production and Management Engineering, Democritus University of Thrace, Xanthi, Greece.
| | | | | | - Ioannis Seimenis
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Koulouriotis
- School of Mechanical Engineering, National Technical University of Athens, Athens, Greece
| |
Collapse
|
2
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408069. [PMID: 39535476 PMCID: PMC11727298 DOI: 10.1002/advs.202408069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Tianxiang Li
- Department of UltrasoundState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical. SciencesPeking Union Medical CollegeBeijing100730China
| | - Bingxin Gong
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Sichen Wang
- School of Life Science and TechnologyComputational Biology Research CenterHarbin Institute of TechnologyHarbin150001China
| | - Lian Yang
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Chuansheng Zheng
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| |
Collapse
|
3
|
Aranaz Murillo A, Cruz Ciria S, García Barrado A, García Mur C. MRI biomarkers and their correlation with the Oncotype DX test. RADIOLOGIA 2025; 67:54-60. [PMID: 39978880 DOI: 10.1016/j.rxeng.2023.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/15/2023] [Indexed: 02/22/2025]
Abstract
Breast cancer (BC) has high rates of incidence and prevalence, causing significant impact in our society. Magnetic resonance imaging (MRI) plays a crucial role in its detection and staging. The Oncotype DX Breast Recurrence Score (ODXRS) test can be used to guide decision making regarding adjuvant chemotherapy (CT) in early-stage luminal BC to allow for more tailored cancer treatment. The aim of this article is to review knowledge regarding MRI biomarkers to date according to the BI-RADS® classification and the use of artificial intelligence (AI) in this imaging technique to establish its correlation with the ODXRS test. The latest studies published on AI and MRI present promising findings, and their standardisation could mark a turning point in breast radiology.
Collapse
Affiliation(s)
- A Aranaz Murillo
- Servicio de Radiología, Hospital Universitario Miguel Servet, Zaragoza, Spain.
| | - S Cruz Ciria
- Servicio de Radiología, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - A García Barrado
- Servicio de Radiología, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - C García Mur
- Servicio de Radiología, Hospital Universitario Miguel Servet, Zaragoza, Spain
| |
Collapse
|
4
|
Luo H, Chen Z, Xu H, Ren J, Zhou P. Peritumoral edema enhances MRI-based deep learning radiomic model for axillary lymph node metastasis burden prediction in breast cancer. Sci Rep 2024; 14:18900. [PMID: 39143315 PMCID: PMC11324898 DOI: 10.1038/s41598-024-69725-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024] Open
Abstract
To investigate whether peritumoral edema (PE) could enhance deep learning radiomic (DLR) model in predicting axillary lymph node metastasis (ALNM) burden in breast cancer. Invasive breast cancer patients with preoperative MRI were retrospectively enrolled and categorized into low (< 2 lymph nodes involved (LNs+)) and high (≥ 2 LNs+) burden groups based on surgical pathology. PE was evaluated on T2WI, and intra- and peri-tumoral radiomic features were extracted from MRI-visible tumors in DCE-MRI. Deep learning models were developed for LN burden prediction in the training cohort and validated in an independent cohort. The incremental value of PE was evaluated through receiver operating characteristic (ROC) analysis, confirming the improvement in the area under the curve (AUC) using the Delong test. This was complemented by net reclassification improvement (NRI) and integrated discrimination improvement (IDI) metrics. The deep learning combined model, incorporating PE with selected radiomic features, demonstrated significantly higher AUC values compared to the MRI model and the DLR model in the training cohort (n = 177) (AUC: 0.953 vs. 0.849 and 0.867, p < 0.05) and the validation cohort (n = 111) (AUC: 0.963 vs. 0.883 and 0.882, p < 0.05). The complementary analysis demonstrated that PE significantly enhances the prediction performance of the DLR model (Categorical NRI: 0.551, p < 0.001; IDI = 0.343, p < 0.001). These findings were confirmed in the validation cohort (Categorical NRI: 0.539, p < 0.001; IDI = 0.387, p < 0.001). PE improved preoperative ALNM burden prediction of DLR model, facilitating personalized axillary management in breast cancer patients.
Collapse
Affiliation(s)
- Hongbing Luo
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, 4th Section of South Ren-Min Road, Chengdu, 610041, China.
| | - Zhe Chen
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, 4th Section of South Ren-Min Road, Chengdu, 610041, China
| | - Hao Xu
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, 4th Section of South Ren-Min Road, Chengdu, 610041, China
| | - Jing Ren
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, 4th Section of South Ren-Min Road, Chengdu, 610041, China
| | - Peng Zhou
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, 4th Section of South Ren-Min Road, Chengdu, 610041, China.
| |
Collapse
|
5
|
Xu R, Yu D, Luo P, Li X, Jiang L, Chang S, Li G. Do Habitat MRI and Fractal Analysis Help Distinguish Triple-Negative Breast Cancer From Non-Triple-Negative Breast Carcinoma. Can Assoc Radiol J 2024; 75:584-592. [PMID: 38389194 DOI: 10.1177/08465371241231573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
Purpose: To determine whether multiparametric MRI-based spatial habitats and fractal analysis can help distinguish triple-negative breast cancer (TNBC) from non-TNBC. Method: Multiparametric DWI and DCE-MRI at 3T were obtained from 142 biopsy- and surgery-proven breast cancer with 148 breast lesions (TNBC = 26 and non-TNBC = 122). The contrast-enhancing lesions were divided into 3 spatial habitats based on perfusion and diffusion patterns using K-means clustering. The fractal dimension (FD) of the tumour subregions was calculated. The accuracy of the habitat segmentation was measured using the Dice index. Inter- and intra-reader reliability were evaluated with the intraclass correlation coefficient (ICC). The ability to predict TNBC status was assessed using the receiver operating characteristic curve. Results: The Dice index for the whole tumour was 0.81 for inter-reader and 0.88 for intra-reader reliability. The inter- and intra-reader reliability were excellent for all 3 tumour habitats and fractal features (ICC > 0.9). TNBC had a lower hypervascular cellular habitat and higher FD 1 compared to non-TNBC (all P < .001). Multivariate analysis confirmed that hypervascular cellular habitat (OR = 0.88) and FD 1 (OR = 1.35) were independently associated with TNBC (all P < .001) after adjusting for rim enhancement, axillary lymph nodes status, and histological grade. The diagnostic model combining hypervascular cellular habitat and FD 1 showed excellent discriminatory ability for TNBC, with an AUC of 0.951 and an accuracy of 91.9%. Conclusions: The fraction of hypervascular cellular habitat and its FD may serve as useful imaging biomarkers for predicting TNBC status.
Collapse
Affiliation(s)
- Run Xu
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Yu
- United Imaging Research Institute of Intelligent Imaging, Beijing, China
| | - Peng Luo
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuefeng Li
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Jiang
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shixin Chang
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guanwu Li
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Jiang J, Li L, Yin G, Luo H, Li J. A Molecular Typing Method for Invasive Breast Cancer by Serum Raman Spectroscopy. Clin Breast Cancer 2024; 24:376-383. [PMID: 38492997 DOI: 10.1016/j.clbc.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/17/2024] [Accepted: 02/12/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND The incidence of breast cancer ranks highest among cancers and is exceedingly heterogeneous. Immunohistochemical staining is commonly used clinically to identify the molecular subtype for subsequent treatment and prognosis. PURPOSE Raman spectroscopy and support vector machine (SVM) learning algorithm were utilized to identify blood samples from breast cancer patients in order to investigate a novel molecular typing approach. METHOD Tumor tissue coarse needle aspiration biopsy samples, and peripheral venous blood samples were gathered from 459 invasive breast cancer patients admitted to the breast department of Sichuan Cancer Hospital between June 2021 and September 2022. Immunohistochemical staining and in situ hybridization were performed on the coarse needle aspiration biopsy tissues to obtain their molecular typing pathological labels, including: 70 cases of Luminal A, 167 cases of Luminal B (HER2-positive), 57 cases of Luminal B (HER2-negative), 84 cases of HER2-positive, and 81 cases of triple-negative. Blood samples were processed to obtained Raman spectra taken for SVM classification models establishment with machine algorithms (using 80% of the sample data as the training set), and then the performance of the SVM classification models was evaluated by the independent validation set (20% of the sample data). RESULTS The AUC values of SVM classification models remained above 0.85, demonstrating outstanding model performance and excellent subtype discrimination of breast cancer molecular subtypes. CONCLUSION Raman spectroscopy of serum samples can promptly and precisely detect the molecular subtype of invasive breast cancer, which has the potential for clinical value.
Collapse
Affiliation(s)
- Jun Jiang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Lintao Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Gang Yin
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Huaichao Luo
- Department of Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Junjie Li
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
7
|
Zheng X, Yin J. Efficacy of texture analysis in determining the gene amplification status of HER2 2+ for invasive ductal carcinoma cases. Minerva Med 2023; 114:832-838. [PMID: 32239879 DOI: 10.23736/s0026-4806.20.06536-2] [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: 01/17/2024]
Abstract
BACKGROUND Gene amplification of human epidermal growth factor receptor2 (HER2) 2+ is essential to be determined for treatment planning. A search of the PubMed database indicates that the correlation between texture features from dynamic contrast enhanced (DCE)-MRI and HER2 2+ status has not been investigated extensively in invasive ductal carcinoma cases. METHODS Seventy-one DCE-MRI cases of HER2 2+ status verified using fluorescence in-situ hybridization (FISH) were selected, including 36 positive and 35 negative cases. Overall, 279 texture features were derived from lesion regions of interest manually drawn onto the subtraction images between pre- and post-contrast agent. Fisher coefficient, mutual information, minimization of both classification error probability and average correlation coefficients as well as a combination of all three methods (MPF) were independently used to reduce the dimensionality of texture parameters. A popular machine learning algorithm, the Support Vector Machine, was further applied to determine HER2 2+ status. Receiver operating characteristic (ROC) analysis was conducted to evaluate the classification performance. RESULTS Diagnostic accuracy was optimal when the most significant discriminatory features were selected using MPF. The area under ROC curve reached 0.863 with corresponding accuracy, sensitivity and specificity rates of 81.80%, 85.71% and 77.78%, respectively. CONCLUSIONS Texture analysis based on breast MRI delivered consistently high performance with FISH detection and may serve as a useful supplementary tool for determining the gene amplification status of HER2 2+ for cases with invasive ductal carcinoma.
Collapse
Affiliation(s)
- Xu Zheng
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiandong Yin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China -
| |
Collapse
|
8
|
Kim MY, Yoen H, Ji H, Park SJ, Kim SM, Han W, Cho N. Ultrafast MRI and T1 and T2 Radiomics for Predicting Invasive Components in Ductal Carcinoma in Situ Diagnosed With Percutaneous Needle Biopsy. Korean J Radiol 2023; 24:1190-1199. [PMID: 38016679 PMCID: PMC10700996 DOI: 10.3348/kjr.2023.0208] [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: 08/18/2022] [Revised: 07/26/2023] [Accepted: 09/05/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the feasibility of ultrafast magnetic resonance imaging (MRI) and radiomic features derived from breast MRI for predicting the upstaging of ductal carcinoma in situ (DCIS) diagnosed using percutaneous needle biopsy. MATERIALS AND METHODS Between August 2018 and June 2020, 95 patients with 98 DCIS lesions who underwent preoperative breast MRI, including an ultrafast sequence, and subsequent surgery were included. Four ultrafast MRI parameters were analyzed: time-to-enhancement, maximum slope (MS), area under the curve for 60 s after enhancement, and time-to-peak enhancement. One hundred and seven radiomic features were extracted for the whole tumor on the first post-contrast T1WI and T2WI using PyRadiomics. Clinicopathological characteristics, ultrafast MRI findings, and radiomic features were compared between the pure DCIS and DCIS with invasion groups. Prediction models, incorporating clinicopathological, ultrafast MRI, and radiomic features, were developed. Receiver operating characteristic curve analysis and area under the curve (AUC) were used to evaluate model performance in distinguishing between the two groups using leave-one-out cross-validation. RESULTS Thirty-six of the 98 lesions (36.7%) were confirmed to have invasive components after surgery. Compared to the pure DCIS group, the DCIS with invasion group had a higher nuclear grade (P < 0.001), larger mean lesion size (P = 0.038), larger mean MS (P = 0.002), and different radiomic-related characteristics, including a more extensive tumor volume; higher maximum gray-level intensity; coarser, more complex, and heterogeneous texture; and a greater concentration of high gray-level intensity. No significant differences in AUCs were found between the model incorporating nuclear grade and lesion size (0.687) and the models integrating additional ultrafast MRI and radiomic features (0.680-0.732). CONCLUSION High nuclear grade, larger lesion size, larger MS, and multiple radiomic features were associated with DCIS upstaging. However, the addition of MS and radiomic features to the prediction model did not significantly improve the prediction performance.
Collapse
Affiliation(s)
- Min Young Kim
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Heera Yoen
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hye Ji
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sang Joon Park
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
- MEDICALIP Co. Ltd., Seoul, Republic of Korea
| | - Sun Mi Kim
- Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Wonshik Han
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Nariya Cho
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Zhang J, Wu J, Zhou XS, Shi F, Shen D. Recent advancements in artificial intelligence for breast cancer: Image augmentation, segmentation, diagnosis, and prognosis approaches. Semin Cancer Biol 2023; 96:11-25. [PMID: 37704183 DOI: 10.1016/j.semcancer.2023.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/03/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Breast cancer is a significant global health burden, with increasing morbidity and mortality worldwide. Early screening and accurate diagnosis are crucial for improving prognosis. Radiographic imaging modalities such as digital mammography (DM), digital breast tomosynthesis (DBT), magnetic resonance imaging (MRI), ultrasound (US), and nuclear medicine techniques, are commonly used for breast cancer assessment. And histopathology (HP) serves as the gold standard for confirming malignancy. Artificial intelligence (AI) technologies show great potential for quantitative representation of medical images to effectively assist in segmentation, diagnosis, and prognosis of breast cancer. In this review, we overview the recent advancements of AI technologies for breast cancer, including 1) improving image quality by data augmentation, 2) fast detection and segmentation of breast lesions and diagnosis of malignancy, 3) biological characterization of the cancer such as staging and subtyping by AI-based classification technologies, 4) prediction of clinical outcomes such as metastasis, treatment response, and survival by integrating multi-omics data. Then, we then summarize large-scale databases available to help train robust, generalizable, and reproducible deep learning models. Furthermore, we conclude the challenges faced by AI in real-world applications, including data curating, model interpretability, and practice regulations. Besides, we expect that clinical implementation of AI will provide important guidance for the patient-tailored management.
Collapse
Affiliation(s)
- Jiadong Zhang
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China
| | - Jiaojiao Wu
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Xiang Sean Zhou
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Feng Shi
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China.
| | - Dinggang Shen
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China; Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China; Shanghai Clinical Research and Trial Center, Shanghai, China.
| |
Collapse
|
10
|
Liu Z, Duan T, Zhang Y, Weng S, Xu H, Ren Y, Zhang Z, Han X. Radiogenomics: a key component of precision cancer medicine. Br J Cancer 2023; 129:741-753. [PMID: 37414827 PMCID: PMC10449908 DOI: 10.1038/s41416-023-02317-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 05/02/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Radiogenomics, focusing on the relationship between genomics and imaging phenotypes, has been widely applied to address tumour heterogeneity and predict immune responsiveness and progression. It is an inevitable consequence of current trends in precision medicine, as radiogenomics costs less than traditional genetic sequencing and provides access to whole-tumour information rather than limited biopsy specimens. By providing voxel-by-voxel genetic information, radiogenomics can allow tailored therapy targeting a complete, heterogeneous tumour or set of tumours. In addition to quantifying lesion characteristics, radiogenomics can also be used to distinguish benign from malignant entities, as well as patient characteristics, to better stratify patients according to disease risk, thereby enabling more precise imaging and screening. Here, we have characterised the radiogenomic application in precision medicine using a multi-omic approach. we outline the main applications of radiogenomics in diagnosis, treatment planning and evaluations in the field of oncology with the aim of developing quantitative and personalised medicine. Finally, we discuss the challenges in the field of radiogenomics and the scope and clinical applicability of these methods.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, 450052, Zhengzhou, Henan, China
| | - Tian Duan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
11
|
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: 0.5] [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
|
12
|
Chiacchiaretta P, Mastrodicasa D, Chiarelli AM, Luberti R, Croce P, Sguera M, Torrione C, Marinelli C, Marchetti C, Domenico A, Cocco G, Di Credico A, Russo A, D’Eramo C, Corvino A, Colasurdo M, Sensi SL, Muzi M, Caulo M, Delli Pizzi A. MRI-Based Radiomics Approach Predicts Tumor Recurrence in ER + /HER2 - Early Breast Cancer Patients. J Digit Imaging 2023; 36:1071-1080. [PMID: 36698037 PMCID: PMC10287859 DOI: 10.1007/s10278-023-00781-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/26/2023] Open
Abstract
Oncotype Dx Recurrence Score (RS) has been validated in patients with ER + /HER2 - invasive breast carcinoma to estimate patient risk of recurrence and guide the use of adjuvant chemotherapy. We investigated the role of MRI-based radiomics features extracted from the tumor and the peritumoral tissues to predict the risk of tumor recurrence. A total of 62 patients with biopsy-proved ER + /HER2 - breast cancer who underwent pre-treatment MRI and Oncotype Dx were included. An RS > 25 was considered discriminant between low-intermediate and high risk of tumor recurrence. Two readers segmented each tumor. Radiomics features were extracted from the tumor and the peritumoral tissues. Partial least square (PLS) regression was used as the multivariate machine learning algorithm. PLS β-weights of radiomics features included the 5% features with the largest β-weights in magnitude (top 5%). Leave-one-out nested cross-validation (nCV) was used to achieve hyperparameter optimization and evaluate the generalizable performance of the procedure. The diagnostic performance of the radiomics model was assessed through receiver operating characteristic (ROC) analysis. A null hypothesis probability threshold of 5% was chosen (p < 0.05). The exploratory analysis for the complete dataset revealed an average absolute correlation among features of 0.51. The nCV framework delivered an AUC of 0.76 (p = 1.1∙10-3). When combining "early" and "peak" DCE images of only T or TST, a tendency toward statistical significance was obtained for TST with an AUC of 0.61 (p = 0.05). The 47 features included in the top 5% were balanced between T and TST (23 and 24, respectively). Moreover, 33/47 (70%) were texture-related, and 25/47 (53%) were derived from high-resolution images (1 mm). A radiomics-based machine learning approach shows the potential to accurately predict the recurrence risk in early ER + /HER2 - breast cancer patients.
Collapse
Affiliation(s)
- Piero Chiacchiaretta
- Advanced Computing Core, Center of Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Odonoiatry, “G. d’Annunzio” University, Chieti, Italy
| | | | - Antonio Maria Chiarelli
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Riccardo Luberti
- Unit of Radiology, “Santissima Annunziata” Hospital, Chieti, Italy
| | - Pierpaolo Croce
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Mario Sguera
- Unit of Radiology, “Santissima Annunziata” Hospital, Chieti, Italy
| | | | | | - Chiara Marchetti
- Unit of Radiology, “Santissima Annunziata” Hospital, Chieti, Italy
| | | | - Giulio Cocco
- Unit of Ultrasound in Internal Medicine, Department of Medicine and Science of Aging, “G. D’Annunzio” University, Chieti, Italy
| | | | | | | | - Antonio Corvino
- Motor Science and Wellness Department, University of Naples “Parthenope”, 80133 Naples, Italy
| | - Marco Colasurdo
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Stefano L. Sensi
- Advanced Computing Core, Center of Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Marzia Muzi
- Breast Unit, “Gaetano Bernabeo” Hospital, Ortona, Italy
| | - Massimo Caulo
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University, Chieti, Italy
| | - Andrea Delli Pizzi
- Department of Innovative Technologies in Medicine and Odonoiatry, “G. d’Annunzio” University, Chieti, Italy
| |
Collapse
|
13
|
Steyaert S, Pizurica M, Nagaraj D, Khandelwal P, Hernandez-Boussard T, Gentles AJ, Gevaert O. Multimodal data fusion for cancer biomarker discovery with deep learning. NAT MACH INTELL 2023; 5:351-362. [PMID: 37693852 PMCID: PMC10484010 DOI: 10.1038/s42256-023-00633-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 02/17/2023] [Indexed: 09/12/2023]
Abstract
Technological advances now make it possible to study a patient from multiple angles with high-dimensional, high-throughput multi-scale biomedical data. In oncology, massive amounts of data are being generated ranging from molecular, histopathology, radiology to clinical records. The introduction of deep learning has significantly advanced the analysis of biomedical data. However, most approaches focus on single data modalities leading to slow progress in methods to integrate complementary data types. Development of effective multimodal fusion approaches is becoming increasingly important as a single modality might not be consistent and sufficient to capture the heterogeneity of complex diseases to tailor medical care and improve personalised medicine. Many initiatives now focus on integrating these disparate modalities to unravel the biological processes involved in multifactorial diseases such as cancer. However, many obstacles remain, including lack of usable data as well as methods for clinical validation and interpretation. Here, we cover these current challenges and reflect on opportunities through deep learning to tackle data sparsity and scarcity, multimodal interpretability, and standardisation of datasets.
Collapse
Affiliation(s)
- Sandra Steyaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
| | - Marija Pizurica
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
| | | | | | - Tina Hernandez-Boussard
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| | - Andrew J Gentles
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| |
Collapse
|
14
|
Predicting gene mutation status via artificial intelligence technologies based on multimodal integration (MMI) to advance precision oncology. Semin Cancer Biol 2023; 91:1-15. [PMID: 36801447 DOI: 10.1016/j.semcancer.2023.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/30/2023] [Accepted: 02/15/2023] [Indexed: 02/21/2023]
Abstract
Personalized treatment strategies for cancer frequently rely on the detection of genetic alterations which are determined by molecular biology assays. Historically, these processes typically required single-gene sequencing, next-generation sequencing, or visual inspection of histopathology slides by experienced pathologists in a clinical context. In the past decade, advances in artificial intelligence (AI) technologies have demonstrated remarkable potential in assisting physicians with accurate diagnosis of oncology image-recognition tasks. Meanwhile, AI techniques make it possible to integrate multimodal data such as radiology, histology, and genomics, providing critical guidance for the stratification of patients in the context of precision therapy. Given that the mutation detection is unaffordable and time-consuming for a considerable number of patients, predicting gene mutations based on routine clinical radiological scans or whole-slide images of tissue with AI-based methods has become a hot issue in actual clinical practice. In this review, we synthesized the general framework of multimodal integration (MMI) for molecular intelligent diagnostics beyond standard techniques. Then we summarized the emerging applications of AI in the prediction of mutational and molecular profiles of common cancers (lung, brain, breast, and other tumor types) pertaining to radiology and histology imaging. Furthermore, we concluded that there truly exist multiple challenges of AI techniques in the way of its real-world application in the medical field, including data curation, feature fusion, model interpretability, and practice regulations. Despite these challenges, we still prospect the clinical implementation of AI as a highly potential decision-support tool to aid oncologists in future cancer treatment management.
Collapse
|
15
|
McCague C, Ramlee S, Reinius M, Selby I, Hulse D, Piyatissa P, Bura V, Crispin-Ortuzar M, Sala E, Woitek R. Introduction to radiomics for a clinical audience. Clin Radiol 2023; 78:83-98. [PMID: 36639175 DOI: 10.1016/j.crad.2022.08.149] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/31/2022] [Indexed: 01/12/2023]
Abstract
Radiomics is a rapidly developing field of research focused on the extraction of quantitative features from medical images, thus converting these digital images into minable, high-dimensional data, which offer unique biological information that can enhance our understanding of disease processes and provide clinical decision support. To date, most radiomics research has been focused on oncological applications; however, it is increasingly being used in a raft of other diseases. This review gives an overview of radiomics for a clinical audience, including the radiomics pipeline and the common pitfalls associated with each stage. Key studies in oncology are presented with a focus on both those that use radiomics analysis alone and those that integrate its use with other multimodal data streams. Importantly, clinical applications outside oncology are also presented. Finally, we conclude by offering a vision for radiomics research in the future, including how it might impact our practice as radiologists.
Collapse
Affiliation(s)
- C McCague
- Department of Radiology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - S Ramlee
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - M Reinius
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - I Selby
- Department of Radiology, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - D Hulse
- Department of Radiology, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - P Piyatissa
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - V Bura
- Department of Radiology, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Radiology and Medical Imaging, County Clinical Emergency Hospital, Cluj-Napoca, Romania
| | - M Crispin-Ortuzar
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Department of Oncology, University of Cambridge, Cambridge, UK
| | - E Sala
- Department of Radiology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - R Woitek
- Department of Radiology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Research Centre for Medical Image Analysis and Artificial Intelligence (MIAAI), Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| |
Collapse
|
16
|
Brancato V, Brancati N, Esposito G, La Rosa M, Cavaliere C, Allarà C, Romeo V, De Pietro G, Salvatore M, Aiello M, Sangiovanni M. A Two-Step Feature Selection Radiomic Approach to Predict Molecular Outcomes in Breast Cancer. SENSORS (BASEL, SWITZERLAND) 2023; 23:1552. [PMID: 36772592 PMCID: PMC9921618 DOI: 10.3390/s23031552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
Breast Cancer (BC) is the most common cancer among women worldwide and is characterized by intra- and inter-tumor heterogeneity that strongly contributes towards its poor prognosis. The Estrogen Receptor (ER), Progesterone Receptor (PR), Human Epidermal Growth Factor Receptor 2 (HER2), and Ki67 antigen are the most examined markers depicting BC heterogeneity and have been shown to have a strong impact on BC prognosis. Radiomics can noninvasively predict BC heterogeneity through the quantitative evaluation of medical images, such as Magnetic Resonance Imaging (MRI), which has become increasingly important in the detection and characterization of BC. However, the lack of comprehensive BC datasets in terms of molecular outcomes and MRI modalities, and the absence of a general methodology to build and compare feature selection approaches and predictive models, limit the routine use of radiomics in the BC clinical practice. In this work, a new radiomic approach based on a two-step feature selection process was proposed to build predictors for ER, PR, HER2, and Ki67 markers. An in-house dataset was used, containing 92 multiparametric MRIs of patients with histologically proven BC and all four relevant biomarkers available. Thousands of radiomic features were extracted from post-contrast and subtracted Dynamic Contrast-Enanched (DCE) MRI images, Apparent Diffusion Coefficient (ADC) maps, and T2-weighted (T2) images. The two-step feature selection approach was used to identify significant radiomic features properly and then to build the final prediction models. They showed remarkable results in terms of F1-score for all the biomarkers: 84%, 63%, 90%, and 72% for ER, HER2, Ki67, and PR, respectively. When possible, the models were validated on the TCGA/TCIA Breast Cancer dataset, returning promising results (F1-score = 88% for the ER+/ER- classification task). The developed approach efficiently characterized BC heterogeneity according to the examined molecular biomarkers.
Collapse
Affiliation(s)
- Valentina Brancato
- IRCCS SYNLAB SDN, Istituto di Ricerca Diagnostica e Nucleare, Via E. Gianturco 113, 80143 Naples, Italy
| | - Nadia Brancati
- Institute for High Performance Computing and Networking, National Research Council of Italy (ICAR-CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Giusy Esposito
- Bio Check Up S.r.l., Via Riviera di Chiaia 9a, 80122 Naples, Italy
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Massimo La Rosa
- Institute for High Performance Computing and Networking, National Research Council of Italy (ICAR-CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Carlo Cavaliere
- IRCCS SYNLAB SDN, Istituto di Ricerca Diagnostica e Nucleare, Via E. Gianturco 113, 80143 Naples, Italy
| | - Ciro Allarà
- Bio Check Up S.r.l., Via Riviera di Chiaia 9a, 80122 Naples, Italy
| | - Valeria Romeo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppe De Pietro
- Institute for High Performance Computing and Networking, National Research Council of Italy (ICAR-CNR), Via P. Castellino 111, 80131 Naples, Italy
| | - Marco Salvatore
- IRCCS SYNLAB SDN, Istituto di Ricerca Diagnostica e Nucleare, Via E. Gianturco 113, 80143 Naples, Italy
| | - Marco Aiello
- IRCCS SYNLAB SDN, Istituto di Ricerca Diagnostica e Nucleare, Via E. Gianturco 113, 80143 Naples, Italy
| | - Mara Sangiovanni
- Institute for High Performance Computing and Networking, National Research Council of Italy (ICAR-CNR), Via P. Castellino 111, 80131 Naples, Italy
| |
Collapse
|
17
|
Chen Y, Tang W, Liu W, Li R, Wang Q, Shen X, Gong J, Gu Y, Peng W. Multiparametric
MR
Imaging Radiomics Signatures for Assessing the Recurrence Risk of
ER
+/
HER2
− Breast Cancer Quantified With 21‐Gene Recurrence Score. J Magn Reson Imaging 2022. [DOI: 10.1002/jmri.28547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Yang Chen
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Wei Tang
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Wei Liu
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Ruimin Li
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Qifeng Wang
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
- Department of Pathology Fudan University Shanghai Cancer Center Shanghai China
| | - Xigang Shen
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Jing Gong
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Yajia Gu
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Weijun Peng
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| |
Collapse
|
18
|
Sheng W, Xia S, Wang Y, Yan L, Ke S, Mellisa E, Gong F, Zheng Y, Tang T. Invasive ductal breast cancer molecular subtype prediction by MRI radiomic and clinical features based on machine learning. Front Oncol 2022; 12:964605. [PMID: 36172153 PMCID: PMC9510620 DOI: 10.3389/fonc.2022.964605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundMost studies of molecular subtype prediction in breast cancer were mainly based on two-dimensional MRI images, the predictive value of three-dimensional volumetric features from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) for predicting breast cancer molecular subtypes has not been thoroughly investigated. This study aimed to look into the role of features derived from DCE-MRI and how they could be combined with clinical data to predict invasive ductal breast cancer molecular subtypes.MethodsFrom January 2019 to December 2021, 190 Chinese women with invasive ductal breast cancer were studied (32 triple-negative, 59 HER2-enriched, and 99 luminal lesions) in this institutional review board-approved retrospective cohort study. The image processing software extracted 1130 quantitative radiomic features from the segmented lesion area, including shape-based, first-order statistical, texture, and wavelet features. Three binary classifications of the subtypes were performed: triple-negative vs. non-triple-negative, HER2-overexpressed vs. non-HER2-overexpressed, and luminal (A + B) vs. non-luminal. For the classification, five machine learning methods (random forest, logistic regression, support vector machine, naïve Bayes, and eXtreme Gradient Boosting) were employed. The classifiers were chosen using the least absolute shrinkage and selection operator method. The area evaluated classification performance under the receiver operating characteristic curve, sensitivity, specificity, accuracy, F1-Score, false positive rate, precision, and geometric mean.ResultsEXtreme Gradient Boosting model showed the best performance in luminal and non-luminal groups, with AUC, sensitivity, specificity, accuracy, F1-Score, false positive rate, precision, and geometric mean of 0.8282, 0.7524, 0.6542, 0.6964, 0.6086, 0.3458, 0.8524 and 0.7016, respectively. Meanwhile, the random forest model showed the best performance in HER2-overexpressed and non-HER2-overexpressed groups, with AUC, sensitivity, specificity, accuracy, F1-Score, false positive rate, precision, and geometric mean of 0.8054, 0.2941, 0.9744, 0.7679, 0.4348, 0.0256, 0.8333 and 0.5353, respectively. Furthermore, eXtreme Gradient Boosting model showed the best performance in the triple-negative and non-triple-negative groups, with AUC, sensitivity, specificity, accuracy, F1-Score, false positive rate, precision, and geometric mean of 0.9031, 0.9362, 0.4444, 0.8571, 0.9167, 0.5556, 0.8980 and 0.6450.ConclusionClinical data and three-dimension imaging features from DCE-MRI were identified as potential biomarkers for distinguishing between three molecular subtypes of invasive ductal carcinomas breast cancer. In the future, more extensive studies will be required to evaluate the findings.
Collapse
Affiliation(s)
- Weiyong Sheng
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Shouli Xia
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yaru Wang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lizhao Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songqing Ke
- Department of Science and Technology Research Management, Wuhan Blood Center, Wuhan, China
| | - Evelyn Mellisa
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Gong
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun Zheng
- Department of Radiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Yun Zheng, ; Tiansheng Tang,
| | - Tiansheng Tang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
- *Correspondence: Yun Zheng, ; Tiansheng Tang,
| |
Collapse
|
19
|
Wu J, Mayer AT, Li R. Integrated imaging and molecular analysis to decipher tumor microenvironment in the era of immunotherapy. Semin Cancer Biol 2022; 84:310-328. [PMID: 33290844 PMCID: PMC8319834 DOI: 10.1016/j.semcancer.2020.12.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/29/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023]
Abstract
Radiological imaging is an integral component of cancer care, including diagnosis, staging, and treatment response monitoring. It contains rich information about tumor phenotypes that are governed not only by cancer cellintrinsic biological processes but also by the tumor microenvironment, such as the composition and function of tumor-infiltrating immune cells. By analyzing the radiological scans using a quantitative radiomics approach, robust relations between specific imaging and molecular phenotypes can be established. Indeed, a number of studies have demonstrated the feasibility of radiogenomics for predicting intrinsic molecular subtypes and gene expression signatures in breast cancer based on MRI. In parallel, promising results have been shown for inferring the amount of tumor-infiltrating lymphocytes, a key factor for the efficacy of cancer immunotherapy, from standard-of-care radiological images. Compared with the biopsy-based approach, radiogenomics offers a unique avenue to profile the molecular makeup of the tumor and immune microenvironment as well as its evolution in a noninvasive and holistic manner through longitudinal imaging scans. Here, we provide a systematic review of the state of the art radiogenomics studies in the era of immunotherapy and discuss emerging paradigms and opportunities in AI and deep learning approaches. These technical advances are expected to transform the radiogenomics field, leading to the discovery of reliable imaging biomarkers. This will pave the way for their clinical translation to guide precision cancer therapy.
Collapse
Affiliation(s)
- Jia Wu
- Department of Imaging Physics, MD Anderson Cancer Center, Texas, 77030, USA; Department of Thoracic/Head & Neck Medical Oncology, MD Anderson Cancer Center, Texas, 77030, USA.
| | - Aaron T Mayer
- Department of Bioengineering, Stanford University, Stanford, California, 94305, USA; Department of Radiology, Stanford University, Stanford, California, 94305, USA; Molecular Imaging Program at Stanford, Stanford University, Stanford, California, 94305, USA; BioX Program at Stanford, Stanford University, Stanford, California, 94305, USA
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University, Stanford, California, 94305, USA
| |
Collapse
|
20
|
Darvish L, Bahreyni-Toossi MT, Roozbeh N, Azimian H. The role of radiogenomics in the diagnosis of breast cancer: a systematic review. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00310-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
One of the most common cancers diagnosed worldwide is breast cancer (BC), which is the leading cause of cancer death among women. The radiogenomics method is more accurate for managing and inhibiting this disease, which takes individual diagnosis on genes, environments, and lifestyles of each person. The present study aims to highlight the current state-of-the-art, the current role and limitations, and future directions of radiogenomics in breast cancer.
Method
This systematic review article was searched from databases such as Embase, PubMed, Web of Science, Google Scholar, Scopus, and Cochrane Library without any date or language limitations of databases. Searches were performed using Boolean OR and AND operators between the main terms and keywords of particular topic of the subject under investigation. All retrospective, prospective, cohort, and pilot studies were included, which were provided with more details about the topic. Articles such as letter to the editor, review, and short communications were excluded because of lack of information, discussions, or use of radiogenomics method on other cancers. For quality assessment of articles, STROBE checklist was used.
Result
For the systematic review, 18 articles were approved after assessing the full text of selected articles. In this review, 3614 patients with BC of selected articles were evaluated, and all radiogenomics were associated with more power in classification, differential diagnosis, and prognosis of BC. Among the various modalities to predict genomic indicators and molecular subtypes, DCE-MRI has the higher performance and finally the highest amount of AUC value (0.956) belonged to PI3K gene.
Conclusion
This review shows that radiogenomics can help with the diagnosis and treatment of breast cancer in patients. It has shown that recognizing and specifying radiogenomic phenotypes in the genomic signatures can be helpful in treatment and diagnosis of disease. The molecular methods used in these articles are limited to miRNAs expression, gene expression, Ki67 proliferation index, next-generation RNA sequencing, whole RNA sequencing, and molecular histopathology that can be completed in future studies by other methods such as exosomal miRNAs, specific proteins expression, DNA repair capacity, and other biomarkers that have prognostic and predictive value for cancer treatment response. Studies with control group and large sample size for evaluation of radiogenomics in diagnosis and treatment recommended.
Collapse
|
21
|
Bhardwaj D, Dasgupta A, DiCenzo D, Brade S, Fatima K, Quiaoit K, Trudeau M, Gandhi S, Eisen A, Wright F, Look-Hong N, Curpen B, Sannachi L, Czarnota GJ. Early Changes in Quantitative Ultrasound Imaging Parameters during Neoadjuvant Chemotherapy to Predict Recurrence in Patients with Locally Advanced Breast Cancer. Cancers (Basel) 2022; 14:cancers14051247. [PMID: 35267555 PMCID: PMC8909335 DOI: 10.3390/cancers14051247] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND This study was conducted to explore the use of quantitative ultrasound (QUS) in predicting recurrence for patients with locally advanced breast cancer (LABC) early during neoadjuvant chemotherapy (NAC). METHODS Eighty-three patients with LABC were scanned with 7 MHz ultrasound before starting NAC (week 0) and during treatment (week 4). Spectral parametric maps were generated corresponding to tumor volume. Twenty-four textural features (QUS-Tex1) were determined from parametric maps acquired using grey-level co-occurrence matrices (GLCM) for each patient, which were further processed to generate 64 texture derivatives (QUS-Tex1-Tex2), leading to a total of 95 features from each time point. Analysis was carried out on week 4 data and compared to baseline (week 0) data. ∆Week 4 data was obtained from the difference in QUS parameters, texture features (QUS-Tex1), and texture derivatives (QUS-Tex1-Tex2) of week 4 data and week 0 data. Patients were divided into two groups: recurrence and non-recurrence. Machine learning algorithms using k-nearest neighbor (k-NN) and support vector machines (SVMs) were used to generate radiomic models. Internal validation was undertaken using leave-one patient out cross-validation method. RESULTS With a median follow up of 69 months (range 7-118 months), 28 patients had disease recurrence. The k-NN classifier was the best performing algorithm at week 4 with sensitivity, specificity, accuracy, and area under curve (AUC) of 87%, 75%, 81%, and 0.83, respectively. The inclusion of texture derivatives (QUS-Tex1-Tex2) in week 4 QUS data analysis led to the improvement of the classifier performances. The AUC increased from 0.70 (0.59 to 0.79, 95% confidence interval) without texture derivatives to 0.83 (0.73 to 0.92) with texture derivatives. The most relevant features separating the two groups were higher-order texture derivatives obtained from scatterer diameter and acoustic concentration-related parametric images. CONCLUSIONS This is the first study highlighting the utility of QUS radiomics in the prediction of recurrence during the treatment of LABC. It reflects that the ongoing treatment-related changes can predict clinical outcomes with higher accuracy as compared to pretreatment features alone.
Collapse
Affiliation(s)
- Divya Bhardwaj
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Archya Dasgupta
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Daniel DiCenzo
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Stephen Brade
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Kashuf Fatima
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Karina Quiaoit
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Maureen Trudeau
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (M.T.); (S.G.); (A.E.)
- Department of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Sonal Gandhi
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (M.T.); (S.G.); (A.E.)
- Department of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Andrea Eisen
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (M.T.); (S.G.); (A.E.)
- Department of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Frances Wright
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.W.); (N.L.-H.)
- Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Nicole Look-Hong
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.W.); (N.L.-H.)
- Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Belinda Curpen
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Medical Imaging, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Lakshmanan Sannachi
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Gregory J. Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M4N 3M5, Canada
- Correspondence: ; Tel.: +416-480-6128
| |
Collapse
|
22
|
Fan M, Cui Y, You C, Liu L, Gu Y, Peng W, Bai Q, Gao X, Li L. Radiogenomic Signatures of Oncotype DX Recurrence Score Enable Prediction of Survival in Estrogen Receptor-Positive Breast Cancer: A Multicohort Study. Radiology 2021; 302:516-524. [PMID: 34846204 DOI: 10.1148/radiol.2021210738] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background Radiogenomics explores the association between imaging features and genomic assays to uncover relevant prognostic features; however, the prognostic implications of the derived signatures remain unclear. Purpose To identify preoperative radiogenomic signatures of estrogen receptor-positive breast cancer associated with the Oncotype DX recurrence score (RS) and to evaluate whether they are biomarkers for survival and responses to neoadjuvant chemotherapy (NACT). Materials and Methods In this retrospective multicohort study, three data sets were analyzed. The radiogenomic development data set, with preoperative dynamic contrast-enhanced MRI and RS data obtained between January 2016 and October 2019 was used to identify radiogenomic signatures. Prognostic implications of the imaging signatures were assessed by measuring overall survival and recurrence-free survival in the prognostic assessment data set using a multivariable Cox proportional hazards model. The therapeutic implication of the radiogenomic signatures was evaluated by determining their ability to predict the response to NACT using the treatment assessment data set obtained between August 2015 and March 2019. Prediction performance was estimated by using the area under the receiver operating characteristic curve (AUC). Results The final cohorts included a radiogenomic development data set with 130 women (mean age, 52 years ± 10 [standard deviation]), a prognostic assessment data set with 116 women (mean age, 48 years ± 9), and a treatment assessment data set with 135 women (mean age, 50 years ± 11). Radiogenomic signatures (n = 11) of texture and morphologic and statistical features were identified to generate the predicted RS (R2 = 0.33, P < .001). A predicted RS greater than 29.9 was associated with poor overall and recurrence-free survival (P = .001 and P = .007, respectively); predicted RS was greater in women with a good NACT response (30.51 ± 6.92 vs 27.35 ± 4.04 [responders vs nonresponders], P = .001). By combining the predicted RS and complementary features, the model achieved improved performance in prediction of the NACT response (AUC, 0.85; P < .001). Conclusion Radiogenomic signatures associated with genomic assays provide markers of prognosis and treatment in estrogen receptor-positive breast cancer. © RSNA, 2021 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ming Fan
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Yajing Cui
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Chao You
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Li Liu
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Yajia Gu
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Weijun Peng
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Qianming Bai
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Xin Gao
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Lihua Li
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| |
Collapse
|
23
|
Sun K, Jiao Z, Zhu H, Chai W, Yan X, Fu C, Cheng JZ, Yan F, Shen D. Radiomics-based machine learning analysis and characterization of breast lesions with multiparametric diffusion-weighted MR. J Transl Med 2021; 19:443. [PMID: 34689804 PMCID: PMC8543912 DOI: 10.1186/s12967-021-03117-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/13/2021] [Indexed: 12/29/2022] Open
Abstract
Background This study aimed to evaluate the utility of radiomics-based machine learning analysis with multiparametric DWI and to compare the diagnostic performance of radiomics features and mean diffusion metrics in the characterization of breast lesions. Methods This retrospective study included 542 lesions from February 2018 to November 2018. One hundred radiomics features were computed from mono-exponential (ME), biexponential (BE), stretched exponential (SE), and diffusion-kurtosis imaging (DKI). Radiomics-based analysis was performed by comparing four classifiers, including random forest (RF), principal component analysis (PCA), L1 regularization (L1R), and support vector machine (SVM). These four classifiers were trained on a training set with 271 patients via ten-fold cross-validation and tested on an independent testing set with 271 patients. The diagnostic performance of the mean diffusion metrics of ME (mADCall b, mADC0–1000), BE (mD, mD*, mf), SE (mDDC, mα), and DKI (mK, mD) were also calculated for comparison. The area under the receiver operating characteristic curve (AUC) was used to compare the diagnostic performance. Results RF attained higher AUCs than L1R, PCA and SVM. The AUCs of radiomics features for the differential diagnosis of breast lesions ranged from 0.80 (BE_D*) to 0.85 (BE_D). The AUCs of the mean diffusion metrics ranged from 0.54 (BE_mf) to 0.79 (ME_mADC0–1000). There were significant differences in the AUCs between the mean values of all diffusion metrics and radiomics features of AUCs (all P < 0.001) for the differentiation of benign and malignant breast lesions. Of the radiomics features computed, the most important sequence was BE_D (AUC: 0.85), and the most important feature was FO-10 percentile (Feature Importance: 0.04). Conclusions The radiomics-based analysis of multiparametric DWI by RF enables better differentiation of benign and malignant breast lesions than the mean diffusion metrics. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03117-5.
Collapse
Affiliation(s)
- Kun Sun
- Department of Radiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhicheng Jiao
- Department of Diagnostic Imaging, Alpert Medical School of Brown University, Providence, USA
| | - Hong Zhu
- Department of Radiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weimin Chai
- Department of Radiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xu Yan
- Scientific Marketing, Siemens Shanghai Magnetic Resonance Ltd., Shanghai, China
| | - Caixia Fu
- MR Application Development, Siemens Shenzhen Magnetic Resonance Ltd., Shenzhen, China
| | - Jie-Zhi Cheng
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Dinggang Shen
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China. .,School of BME, Shanghai Tech University, Shanghai, China.
| |
Collapse
|
24
|
Davey MG, Davey MS, Ryan ÉJ, Boland MR, McAnena PF, Lowery AJ, Kerin MJ. Is radiomic MRI a feasible alternative to OncotypeDX® recurrence score testing? A systematic review and meta-analysis. BJS Open 2021; 5:6388195. [PMID: 34633438 PMCID: PMC8504445 DOI: 10.1093/bjsopen/zrab081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/03/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND OncotypeDX® recurrence score (RS) aids therapeutic decision-making in oestrogen-receptor-positive (ER+) breast cancer. Radiomics is an evolving field that aims to examine the relationship between radiological features and the underlying genomic landscape of disease processes. The aim of this study was to perform a systematic review of current evidence evaluating the comparability of radiomics and RS. METHODS A systematic review was performed as per PRISMA guidelines. Studies comparing radiomic MRI tumour analyses and RS were identified. Sensitivity, specificity and area under curve (AUC) delineating low risk (RS less than 18) versus intermediate-high risk (equal to or greater than 18) and low-intermediate risk (RS less than 30) and high risk (RS greater than 30) were recorded. Log rate ratios (lnRR) and standard error were determined from AUC and 95 per cent confidence intervals. RESULTS Nine studies including 1216 patients met inclusion criteria; the mean age at diagnosis was 52.9 years. Mean RS was 16 (range 0-75); 401 patients with RS less than 18, 287 patients with RS 18-30 and 100 patients with RS greater than 30. Radiomic analysis and RS were comparable for differentiating RS less than 18 versus RS 18 or greater (RR 0.93 (95 per cent c.i. 0.85 to 1.01); P = 0.010, heterogeneity (I2)=0%) as well as RS less than 30 versus RS 30 or greater (RR 0.76 (95 per cent c.i. 0.70 to 0.83); P < 0.001, I2=0%). MRI sensitivity and specificity for RS less than 18 versus 18 or greater was 0.89 (95 per cent c.i. 0.85 to 0.93) and 0.72 (95 per cent c.i. 0.66 to 0.78) respectively, and 0.79 (95 per cent c.i. 0.72 to 0.86) and 0.74 (95 per cent c.i. 0.68 to 0.80) for RS less than 30 versus 30 or greater. CONCLUSION Radiomic tumour analysis is comparable to RS in differentiating patients into clinically relevant subgroups. For patients requiring MRI, radiomics may complement and enhance RS for prognostication and therapeutic decision making in ER+ breast cancer.
Collapse
Affiliation(s)
- M G Davey
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| | - M S Davey
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| | - É J Ryan
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| | - M R Boland
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| | - P F McAnena
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| | - A J Lowery
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| | - M J Kerin
- Department of Surgery, The Lambe Institute for Translational Research, National University of Ireland, Galway, Ireland
| |
Collapse
|
25
|
Bitencourt A, Daimiel Naranjo I, Lo Gullo R, Rossi Saccarelli C, Pinker K. AI-enhanced breast imaging: Where are we and where are we heading? Eur J Radiol 2021; 142:109882. [PMID: 34392105 PMCID: PMC8387447 DOI: 10.1016/j.ejrad.2021.109882] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/15/2021] [Accepted: 07/26/2021] [Indexed: 12/22/2022]
Abstract
Significant advances in imaging analysis and the development of high-throughput methods that can extract and correlate multiple imaging parameters with different clinical outcomes have led to a new direction in medical research. Radiomics and artificial intelligence (AI) studies are rapidly evolving and have many potential applications in breast imaging, such as breast cancer risk prediction, lesion detection and classification, radiogenomics, and prediction of treatment response and clinical outcomes. AI has been applied to different breast imaging modalities, including mammography, ultrasound, and magnetic resonance imaging, in different clinical scenarios. The application of AI tools in breast imaging has an unprecedented opportunity to better derive clinical value from imaging data and reshape the way we care for our patients. The aim of this study is to review the current knowledge and future applications of AI-enhanced breast imaging in clinical practice.
Collapse
Affiliation(s)
- Almir Bitencourt
- Department of Imaging, A.C.Camargo Cancer Center, Sao Paulo, SP, Brazil; Dasa, Sao Paulo, SP, Brazil
| | - Isaac Daimiel Naranjo
- Department of Radiology, Breast Imaging Service, Guy's and St. Thomas' NHS Trust, Great Maze Pond, London, UK
| | - Roberto Lo Gullo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Katja Pinker
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
26
|
Gouri A, Benarba B, Dekaken A, Aoures H, Benharkat S. Prediction of Late Recurrence and Distant Metastasis in Early-stage Breast Cancer: Overview of Current and Emerging Biomarkers. Curr Drug Targets 2021; 21:1008-1025. [PMID: 32164510 DOI: 10.2174/1389450121666200312105908] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022]
Abstract
Recently, a significant number of breast cancer (BC) patients have been diagnosed at an early stage. It is therefore critical to accurately predict the risk of recurrence and distant metastasis for better management of BC in this setting. Clinicopathologic patterns, particularly lymph node status, tumor size, and hormonal receptor status are routinely used to identify women at increased risk of recurrence. However, these factors have limitations regarding their predictive ability for late metastasis risk in patients with early BC. Emerging molecular signatures using gene expression-based approaches have improved the prognostic and predictive accuracy for this indication. However, the use of their based-scores for risk assessment has provided contradictory findings. Therefore, developing and using newly emerged alternative predictive and prognostic biomarkers for identifying patients at high- and low-risk is of great importance. The present review discusses some serum biomarkers and multigene profiling scores for predicting late recurrence and distant metastasis in early-stage BC based on recently published studies and clinical trials.
Collapse
Affiliation(s)
- A Gouri
- Laboratory of Medical Biochemistry, Faculty of Medicine, University of Annaba, Algeria
| | - B Benarba
- Laboratory Research on Biological Systems and Geomatics, Faculty of Nature and Life Sciences, University of Mascara, Algeria
| | - A Dekaken
- Department of Internal Medicine, El Okbi Public Hospital, Guelma, Algeria
| | - H Aoures
- Department of Gynecology and Obstetrics, EHS El Bouni, Annaba, Algeria
| | - S Benharkat
- Laboratory of Medical Biochemistry, Faculty of Medicine, University of Annaba, Algeria
| |
Collapse
|
27
|
Kolios C, Sannachi L, Dasgupta A, Suraweera H, DiCenzo D, Stanisz G, Sahgal A, Wright F, Look-Hong N, Curpen B, Sadeghi-Naini A, Trudeau M, Gandhi S, Kolios MC, Czarnota GJ. MRI texture features from tumor core and margin in the prediction of response to neoadjuvant chemotherapy in patients with locally advanced breast cancer. Oncotarget 2021; 12:1354-1365. [PMID: 34262646 PMCID: PMC8274727 DOI: 10.18632/oncotarget.28002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/11/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Radiomics involving quantitative analysis of imaging has shown promises in oncology to serve as non-invasive biomarkers. We investigated whether pre-treatment T2-weighted magnetic resonance imaging (MRI) can be used to predict response to neoadjuvant chemotherapy (NAC) in breast cancer. MATERIALS AND METHODS MRI scans were obtained for 102 patients with locally advanced breast cancer (LABC). All patients were treated with standard regimens of NAC as decided by the treating oncologist, followed by surgery and adjuvant treatment according to standard institutional practice. The primary tumor was segmented, and 11 texture features were extracted using the grey-level co-occurrence matrices analysis of the T2W-images from tumor cores and margins. Response assessment was done using clinical-pathological responses with patients classified into binary groups: responders and non-responders. Machine learning classifiers were used to develop a radiomics model, and a leave-one-out cross-validation technique was used to assess the performance. RESULTS 7 features were significantly (p < 0.05) different between the two response groups. The best classification accuracy was obtained using a k-nearest neighbor (kNN) model with sensitivity, specificity, accuracy, and area under curve of 63, 93, 87, and 0.78, respectively. CONCLUSIONS Pre-treatment T2-weighted MRI texture features can predict NAC response with reasonable accuracy.
Collapse
Affiliation(s)
- Christopher Kolios
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Lakshmanan Sannachi
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Archya Dasgupta
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Harini Suraweera
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Daniel DiCenzo
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Gregory Stanisz
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Frances Wright
- Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada
| | - Nicole Look-Hong
- Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada
| | - Belinda Curpen
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medical Imaging, University of Toronto, Toronto, Canada
| | - Ali Sadeghi-Naini
- Department of Electrical and Computer Engineering, York University, North York, Canada
| | - Maureen Trudeau
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Sonal Gandhi
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | | | - Gregory J Czarnota
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Department of Physics, Ryerson University, Toronto, Canada
| |
Collapse
|
28
|
Contrast-Enhanced Mammography and Radiomics Analysis for Noninvasive Breast Cancer Characterization: Initial Results. Mol Imaging Biol 2021; 22:780-787. [PMID: 31463822 DOI: 10.1007/s11307-019-01423-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
PURPOSE To investigate the potential of contrast-enhanced mammography (CEM) and radiomics analysis for the noninvasive differentiation of breast cancer invasiveness, hormone receptor status, and tumor grade. PROCEDURES This retrospective study included 100 patients with 103 breast cancers who underwent pretreatment CEM. Radiomics analysis was performed using MAZDA software. Lesions were manually segmented. Radiomic features were derived from first-order histogram (HIS), co-occurrence matrix (COM), run length matrix (RLM), absolute gradient, autoregressive model, the discrete Haar wavelet transform (WAV), and lesion geometry. Fisher, probability of error and average correlation (POE+ACC), and mutual information (MI) coefficients informed feature selection. Linear discriminant analysis followed by k-nearest neighbor classification (with leave-one-out cross-validation) was used for pairwise texture-based separation of tumor invasiveness and hormone receptor status using histopathology as the standard of reference. RESULTS Radiomics analysis achieved the highest accuracies of 87.4 % for differentiating invasive from noninvasive cancers based on COM+HIS/MI, 78.4 % for differentiating HR positive from HR negative cancers based on COM+HIS/Fisher, 97.2 % for differentiating human epidermal growth factor receptor 2 (HER2)-positive/HR-negative from HER2-negative/HR-positive cancers based on RLM+WAV/MI, 100 % for differentiating triple-negative from triple-positive breast cancers mainly based on COM+WAV+HIS/POE+ACC, and 82.1 % for differentiating triple-negative from HR-positive cancers mainly based on WAV+HIS/Fisher. Accuracies for differentiating grade 1 vs. grades 2 and 3 cancers were 90 % for invasive cancers (based on COM/MI) and 100 % for noninvasive cancers (almost entirely based on COM/MI). CONCLUSIONS Radiomics analysis with CEM has potential for noninvasive differentiation of tumors with different degrees of invasiveness, hormone receptor status, and tumor grade.
Collapse
|
29
|
Yin XX, Jin Y, Gao M, Hadjiloucas S. Artificial Intelligence in Breast MRI Radiogenomics: Towards Accurate Prediction of Neoadjuvant Chemotherapy Responses. Curr Med Imaging 2021; 17:452-458. [PMID: 32842944 DOI: 10.2174/1573405616666200825161921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/03/2020] [Accepted: 07/17/2020] [Indexed: 11/22/2022]
Abstract
Neoadjuvant Chemotherapy (NAC) in breast cancer patients has considerable prognostic and treatment potential and can be tailored to individual patients as part of precision medicine protocols. This work reviews recent advances in artificial intelligence so as to enable the use of radiogenomics for accurate NAC analysis and prediction. The work addresses a new problem in radiogenomics mining: How to combine structural radiomics information and non-structural genomics information for accurate NAC prediction. This requires the automated extraction of parameters from structural breast radiomics data, and finding non-structural feature vectors with diagnostic value, which then are combined with genomics data acquired from exocrine bodies in blood samples from a cohort of cancer patients to enable accurate NAC prediction. A self-attention-based deep learning approach, along with an effective multi-channel tumour image reconstruction algorithm of high dimensionality, is proposed. The aim was to generate non-structural feature vectors for accurate prediction of the NAC responses by combining imaging datasets with exocrine body related genomics analysis.
Collapse
Affiliation(s)
- Xiao-Xia Yin
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou 510006, China
| | - Yabin Jin
- The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan 528000, China
| | - Mingyong Gao
- The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan 528000, China
| | - Sillas Hadjiloucas
- Department of Biomedical Engineering, The University of Reading, RG6 6AY, United Kingdom
| |
Collapse
|
30
|
Zhang M, Sadinski M, Haddad D, Bae MS, Martinez D, Morris EA, Gibbs P, Sutton EJ. Background Parenchymal Enhancement on Breast MRI as a Prognostic Surrogate: Correlation With Breast Cancer Oncotype Dx Score. Front Oncol 2021; 10:595820. [PMID: 33614481 PMCID: PMC7890019 DOI: 10.3389/fonc.2020.595820] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose Breast MRI background parenchymal enhancement (BPE) can potentially serve as a prognostic marker, by possible correlation with molecular subtype. Oncotype Dx, a gene assay, is a prognostic and predictive surrogate for tumor aggressiveness and treatment response. The purpose of this study was to investigate the association between contralateral non-tumor breast magnetic resonance imaging (MRI) background parenchymal enhancement and tumor oncotype score. Methods In this retrospective study, patients with ER+ and HER2− early stage invasive ductal carcinoma who underwent preoperative breast MRI, oncotype risk scoring, and breast conservation surgery from 2008–2010 were identified. After registration, BPE from the pre and three post-contrast phases was automatically extracted using a k-means clustering algorithm. Four metrics were calculated: initial enhancement (IE) relative to the pre-contrast signal, late enhancement, overall enhancement (OE), and area under the enhancement curve (AUC). Histogram analysis was performed to determine first order metrics which were compared to oncotype risk score groups using Mann–Whitney tests and Spearman rank correlation analysis. Results This study included 80 women (mean age = 51.1 ± 10.3 years); 46 women were categorized as low risk (≤17) and 34 women were categorized as intermediate/high risk (≥18) according to Oncotype Dx. For the mean of the top 10% pixels, significant differences were noted for IE (p = 0.032), OE (p = 0.049), and AUC (p = 0.044). Using the risk score as a continuous variable, correlation analysis revealed a weak but significant correlation with the mean of the top 10% pixels for IE (r = 0.26, p = 0.02), OE (r = 0.25, p = 0.02), and AUC (r = 0.27, p = 0.02). Conclusion BPE metrics of enhancement in the non-tumor breast are associated with tumor Oncotype Dx recurrence score, suggesting that the breast microenvironment may relate to likelihood of recurrence and magnitude of chemotherapy benefit.
Collapse
Affiliation(s)
- Michelle Zhang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Radiology, McGill University, Montreal, QC, Canada
| | - Meredith Sadinski
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Dana Haddad
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Radiology, Montefiore, New York, NY, United States.,Department of Radiology, Mediclinic Middle East, Dubai, United Arab Emirates.,College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Min Sun Bae
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Danny Martinez
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Elizabeth A Morris
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Peter Gibbs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Elizabeth J Sutton
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
31
|
Shui L, Ren H, Yang X, Li J, Chen Z, Yi C, Zhu H, Shui P. The Era of Radiogenomics in Precision Medicine: An Emerging Approach to Support Diagnosis, Treatment Decisions, and Prognostication in Oncology. Front Oncol 2021; 10:570465. [PMID: 33575207 PMCID: PMC7870863 DOI: 10.3389/fonc.2020.570465] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/08/2020] [Indexed: 02/05/2023] Open
Abstract
With the rapid development of new technologies, including artificial intelligence and genome sequencing, radiogenomics has emerged as a state-of-the-art science in the field of individualized medicine. Radiogenomics combines a large volume of quantitative data extracted from medical images with individual genomic phenotypes and constructs a prediction model through deep learning to stratify patients, guide therapeutic strategies, and evaluate clinical outcomes. Recent studies of various types of tumors demonstrate the predictive value of radiogenomics. And some of the issues in the radiogenomic analysis and the solutions from prior works are presented. Although the workflow criteria and international agreed guidelines for statistical methods need to be confirmed, radiogenomics represents a repeatable and cost-effective approach for the detection of continuous changes and is a promising surrogate for invasive interventions. Therefore, radiogenomics could facilitate computer-aided diagnosis, treatment, and prediction of the prognosis in patients with tumors in the routine clinical setting. Here, we summarize the integrated process of radiogenomics and introduce the crucial strategies and statistical algorithms involved in current studies.
Collapse
Affiliation(s)
- Lin Shui
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Haoyu Ren
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Xi Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Li
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Ziwei Chen
- Department of Nephrology, Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, China
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Pixian Shui
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
32
|
Association between Oncotype DX recurrence score and dynamic contrast-enhanced MRI features in patients with estrogen receptor-positive HER2-negative invasive breast cancer. Clin Imaging 2021; 75:131-137. [PMID: 33548871 DOI: 10.1016/j.clinimag.2021.01.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/06/2021] [Accepted: 01/17/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Oncotype DX is a multigene assay used in breast cancer, and the result provided as a 'recurrence score (RS)' corresponds to the risk of a cancer recurrence and the chemotherapeutic benefit in estrogen receptor (ER)-positive human epidermal growth factor receptor (HER)2-negative invasive breast cancer. However, its accessibility is limited. PURPOSE To evaluate whether magnetic resonance imaging (MRI) could be used to predict Oncotype DX RS in patients with ER-positive HER2-negative invasive breast cancer. MATERIAL AND METHODS We enrolled 473 patients with ER-positive HER2-negative invasive breast cancer who underwent a preoperative MRI and Oncotype DX assay between January 2015 and December 2018. The MRI was reviewed and associations between Oncotype DX RS values were evaluated. Logistic regression analysis was used to identify independent predictors of high and low RS. RESULTS Of the 485 cancers, 288 (59.4%) had low (<18), 155 (31.9%) had intermediate (18-30), and 42 (8.7%) had high (≥31) RS. Multiple logistic regression analysis revealed that a round shape (odds ratio [OR] = 2.554, P = 0.089) and low proportion of washout component (OR = 1.011, P = 0.014) were associated with low RS and that heterogeneously dense (OR = 3.205, P = 0.007) or scattered fibroglandular (OR = 3.776, P = 0.005) breast tissue, a non-spiculated margin (OR = 5.435, P = 0.007), and low proportion of persistent component (OR = 1.012, P = 0.036) were associated with high RS. CONCLUSION MRI features showed the potential for the discrimination of Oncotype DX RS in patients with ER-positive HER2-negative invasive breast cancer.
Collapse
|
33
|
[Multimodal, multiparametric and genetic breast imaging]. Radiologe 2021; 61:183-191. [PMID: 33464404 DOI: 10.1007/s00117-020-00801-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2020] [Indexed: 10/22/2022]
Abstract
CLINICAL/METHODOLOGICAL ISSUE Multiparametric magnetic resonance imaging (MRI) aims to visualize and quantify biological, physiological and pathological processes at the cellular and molecular level and provides valuable information about key processes in cancer development and progression. "Omics" strategies (genomics, transcriptomics, proteomics, metabolomics) have many uses in oncology. STANDARD RADIOLOGICAL METHODS Multiparametric MRI of the breast currently includes T2-weighted, diffusion-weighted and dynamic contrast-enhanced MRI (DCE-MRI) METHODOLOGICAL INNOVATIONS: Additional parameters such as proton magetic resonance spectroscopy (MRS), chemical exchange saturation transfer (CEST), blood oxygen level-dependent (BOLD), hyperpolarized (HP) MRI or lipid MRS are currently being developed and are being evaluated in breast cancer diagnostics. ACHIEVEMENTS Radiogenomics is a new direction in medical science that has been made possible by significant advances in imaging and image analysis methods, as well as the development of techniques to extract and correlate various imaging parameters with "omics" data. The aim of radiogenomics is to correlate imaging characteristics (phenotypes) with gene expression patterns, gene mutations and other genome-associated properties and is the evolution of the correlation between radiology and pathology from the anatomical-histological to the molecular level. Quantitative and qualitative imaging biomarkers provide insights into the complex tumor biology. Initial results suggest that radiogemics will play an important role in the diagnosis, prognosis, and treatment of breast cancer. PRACTICAL RECOMMENDATIONS This article provides an overview of the current state of radiogenomics of the breast and future applications and challenges.
Collapse
|
34
|
Pathak P, Jalal AS, Rai R. Breast Cancer Image Classification: A Review. Curr Med Imaging 2020; 17:720-740. [PMID: 33371857 DOI: 10.2174/0929867328666201228125208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/23/2020] [Accepted: 10/14/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Breast cancer represents uncontrolled breast cell growth. Breast cancer is the most diagnosed cancer in women worldwide. Early detection of breast cancer improves the chances of survival and increases treatment options. There are various methods for screening breast cancer, such as mammogram, ultrasound, computed tomography and Magnetic Resonance Imaging (MRI). MRI is gaining prominence as an alternative screening tool for early detection and breast cancer diagnosis. Nevertheless, MRI can hardly be examined without the use of a Computer-Aided Diagnosis (CAD) framework, due to the vast amount of data. OBJECTIVE This paper aims to cover the approaches used in the CAD system for the detection of breast cancer. METHODS In this paper, the methods used in CAD systems are categories into two classes: the conventional approach and artificial intelligence (AI) approach. RESULTS The conventional approach covers the basic steps of image processing, such as preprocessing, segmentation, feature extraction and classification. The AI approach covers the various convolutional and deep learning networks used for diagnosis. CONCLUSION This review discusses some of the core concepts used in breast cancer and presents a comprehensive review of efforts in the past to address this problem.
Collapse
Affiliation(s)
- Pooja Pathak
- Department of Mathematics, GLA University, Mathura, India
| | - Anand Singh Jalal
- Department of Computer Engineering & Applications, GLA University, Mathura, India
| | - Ritu Rai
- Department of Computer Engineering & Applications, GLA University, Mathura, India
| |
Collapse
|
35
|
Wang Q, Mao N, Liu M, Shi Y, Ma H, Dong J, Zhang X, Duan S, Wang B, Xie H. Radiomic analysis on magnetic resonance diffusion weighted image in distinguishing triple-negative breast cancer from other subtypes: a feasibility study. Clin Imaging 2020; 72:136-141. [PMID: 33242692 DOI: 10.1016/j.clinimag.2020.11.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 09/02/2020] [Accepted: 11/12/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE This work aimed to explore whether radiomic features on magnetic resonance diffusion weighted image (MR DWI) can be used to identify triple-negative breast cancer (TNBC) and other subtypes (non-TNBC). MATERIALS AND METHODS This retrospective study included 221 unilateral patients who underwent breast MR imaging prior to neoadjuvant chemotherapy. The subtypes of breast cancer include luminal A (n = 63), luminal B (n = 103), human epidermal growth factor receptor-2 (HER2) overexpressing (n = 30), and triple negative (n = 25). Radiomic features were extracted using Omini-Kinetic software on DWI. Student's t-test and Mann-Whitney U test were used to compare the features between TNBC and non-TNBC patients. Logistic regression analysis and receiver operating characteristic (ROC) curve were used to evaluate the diagnostic efficiency of radiomic features. The Fisher discriminant model was employed to distinguish TNBC and non-TNBC patients automatically. An additional validation dataset with 169 patients was utilized to validate the model. RESULTS A total of 76 imaging features were extracted from each lesion on DWI images, and 12 radiomic features were statistically significant between TNBC and non-TNBC patients (P < 0.05). The area of receiver operating characteristic curve (AUC) was 0.817 to apply logistic regression analysis. The accuracy of Fisher discriminant model in distinguishing TNBC and non-TNBC patients was 95.4%, and leave-one-out cross validation was achieved with an accuracy of 83.7%. The same classification analysis of the validation dataset showed an accuracy of 83.4% and an AUC of 0.804. CONCLUSION Breast lesions exhibit differences in radiomic features from DWI, enabling good discrimination between TNBC and non-TNBC tumors.
Collapse
Affiliation(s)
- Qinglin Wang
- Department of Radiology, Yantai Yuhuangding Hospital, QingDao University School of Medicine, Yantai, Shandong 264000, PR China
| | - Ning Mao
- Department of Radiology, Yantai Yuhuangding Hospital, QingDao University School of Medicine, Yantai, Shandong 264000, PR China
| | - Meijie Liu
- Institute of medical imaging, Binzhou Medical University, Yantai, Shandong 264000, PR China
| | - Yinghong Shi
- Department of Radiology, Yantai Yuhuangding Hospital, QingDao University School of Medicine, Yantai, Shandong 264000, PR China
| | - Heng Ma
- Department of Radiology, Yantai Yuhuangding Hospital, QingDao University School of Medicine, Yantai, Shandong 264000, PR China
| | - Jianjun Dong
- Department of Radiology, Yantai Yuhuangding Hospital, QingDao University School of Medicine, Yantai, Shandong 264000, PR China
| | | | | | - Bin Wang
- Institute of medical imaging, Binzhou Medical University, Yantai, Shandong 264000, PR China.
| | - Haizhu Xie
- Department of Radiology, Yantai Yuhuangding Hospital, QingDao University School of Medicine, Yantai, Shandong 264000, PR China.
| |
Collapse
|
36
|
Cho N. Imaging features of breast cancer molecular subtypes: state of the art. J Pathol Transl Med 2020; 55:16-25. [PMID: 33153242 PMCID: PMC7829574 DOI: 10.4132/jptm.2020.09.03] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 09/06/2020] [Indexed: 12/25/2022] Open
Abstract
Characterization of breast cancer molecular subtypes has been the standard of care for breast cancer management. We aimed to provide a review of imaging features of breast cancer molecular subtypes for the field of precision medicine. We also provide an update on the recent progress in precision medicine for breast cancer, implications for imaging, and recent observations in longitudinal functional imaging with radiomics.
Collapse
Affiliation(s)
- Nariya Cho
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.,Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
37
|
Min H, McClymont D, Chandra SS, Crozier S, Bradley AP. Automatic lesion detection, segmentation and characterization via 3D multiscale morphological sifting in breast MRI. Biomed Phys Eng Express 2020; 6. [PMID: 35045404 DOI: 10.1088/2057-1976/abc45c] [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: 07/06/2020] [Accepted: 10/23/2020] [Indexed: 11/11/2022]
Abstract
Previous studies on computer aided detection/diagnosis (CAD) in 4D breast magnetic resonance imaging (MRI) usually regard lesion detection, segmentation and characterization as separate tasks, and typically require users to manually select 2D MRI slices or regions of interest as the input. In this work, we present a breast MRI CAD system that can handle 4D multimodal breast MRI data, and integrate lesion detection, segmentation and characterization with no user intervention. The proposed CAD system consists of three major stages: region candidate generation, feature extraction and region candidate classification. Breast lesions are firstly extracted as region candidates using the novel 3D multiscale morphological sifting (MMS). The 3D MMS, which uses linear structuring elements to extract lesion-like patterns, can segment lesions from breast images accurately and efficiently. Analytical features are then extracted from all available 4D multimodal breast MRI sequences, including T1-, T2-weighted and DCE sequences, to represent the signal intensity, texture, morphological and enhancement kinetic characteristics of the region candidates. The region candidates are lastly classified as lesion or normal tissue by the random under-sampling boost (RUSboost), and as malignant or benign lesion by the random forest. Evaluated on a breast MRI dataset which contains a total of 117 cases with 141 biopsy-proven lesions (95 malignant and 46 benign lesions), the proposed system achieves a true positive rate (TPR) of 0.90 at 3.19 false positives per patient (FPP) for lesion detection and a TPR of 0.91 at a FPP of 2.95 for identifying malignant lesions without any user intervention. The average dice similarity index (DSI) is0.72±0.15for lesion segmentation. Compared with previously proposed lesion detection, detection-segmentation and detection-characterization systems evaluated on the same breast MRI dataset, the proposed CAD system achieves a favourable performance in breast lesion detection and characterization.
Collapse
Affiliation(s)
- Hang Min
- School of Information Technology and Electrical Engineering, University of Queensland, Australia
| | - Darryl McClymont
- School of Information Technology and Electrical Engineering, University of Queensland, Australia
| | - Shekhar S Chandra
- School of Information Technology and Electrical Engineering, University of Queensland, Australia
| | - Stuart Crozier
- School of Information Technology and Electrical Engineering, University of Queensland, Australia
| | - Andrew P Bradley
- Science and Engineering Faculty, Queensland University of Technology, Australia
| |
Collapse
|
38
|
Pattern Classification Approaches for Breast Cancer Identification via MRI: State-Of-The-Art and Vision for the Future. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10207201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mining algorithms for Dynamic Contrast Enhanced Magnetic Resonance Imaging (DCE-MRI) of breast tissue are discussed. The algorithms are based on recent advances in multi-dimensional signal processing and aim to advance current state-of-the-art computer-aided detection and analysis of breast tumours when these are observed at various states of development. The topics discussed include image feature extraction, information fusion using radiomics, multi-parametric computer-aided classification and diagnosis using information fusion of tensorial datasets as well as Clifford algebra based classification approaches and convolutional neural network deep learning methodologies. The discussion also extends to semi-supervised deep learning and self-supervised strategies as well as generative adversarial networks and algorithms using generated confrontational learning approaches. In order to address the problem of weakly labelled tumour images, generative adversarial deep learning strategies are considered for the classification of different tumour types. The proposed data fusion approaches provide a novel Artificial Intelligence (AI) based framework for more robust image registration that can potentially advance the early identification of heterogeneous tumour types, even when the associated imaged organs are registered as separate entities embedded in more complex geometric spaces. Finally, the general structure of a high-dimensional medical imaging analysis platform that is based on multi-task detection and learning is proposed as a way forward. The proposed algorithm makes use of novel loss functions that form the building blocks for a generated confrontation learning methodology that can be used for tensorial DCE-MRI. Since some of the approaches discussed are also based on time-lapse imaging, conclusions on the rate of proliferation of the disease can be made possible. The proposed framework can potentially reduce the costs associated with the interpretation of medical images by providing automated, faster and more consistent diagnosis.
Collapse
|
39
|
Nichols BS, Chelales E, Wang R, Schulman A, Gallagher J, Greenup RA, Geradts J, Harter J, Marcom PK, Wilke LG, Ramanujam N. Quantitative assessment of distant recurrence risk in early stage breast cancer using a nonlinear combination of pathological, clinical and imaging variables. JOURNAL OF BIOPHOTONICS 2020; 13:e201960235. [PMID: 32573935 PMCID: PMC8521784 DOI: 10.1002/jbio.201960235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/29/2020] [Accepted: 05/30/2020] [Indexed: 06/11/2023]
Abstract
Use of genomic assays to determine distant recurrence risk in patients with early stage breast cancer has expanded and is now included in the American Joint Committee on Cancer staging manual. Algorithmic alternatives using standard clinical and pathology information may provide equivalent benefit in settings where genomic tests, such as OncotypeDx, are unavailable. We developed an artificial neural network (ANN) model to nonlinearly estimate risk of distant cancer recurrence. In addition to clinical and pathological variables, we enhanced our model using intraoperatively determined global mammographic breast density (MBD) and local breast density (LBD). LBD was measured with optical spectral imaging capable of sensing regional concentrations of tissue constituents. A cohort of 56 ER+ patients with an OncotypeDx score was evaluated. We demonstrated that combining MBD/LBD measurements with clinical and pathological variables improves distant recurrence risk prediction accuracy, with high correlation (r = 0.98) to the OncotypeDx recurrence score.
Collapse
Affiliation(s)
- Brandon S. Nichols
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Erika Chelales
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Roujia Wang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Amanda Schulman
- Department of Surgery, The University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jennifer Gallagher
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Rachel A. Greenup
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Joseph Geradts
- Department of Population Sciences, City of Hope, Duarte, California
| | - Josephine Harter
- Department of Pathology, The University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Paul K. Marcom
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Lee G. Wilke
- Department of Surgery, The University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| |
Collapse
|
40
|
Jacobs MA, Umbricht CB, Parekh VS, El Khouli RH, Cope L, Macura KJ, Harvey S, Wolff AC. Integrated Multiparametric Radiomics and Informatics System for Characterizing Breast Tumor Characteristics with the OncotypeDX Gene Assay. Cancers (Basel) 2020; 12:E2772. [PMID: 32992569 PMCID: PMC7601838 DOI: 10.3390/cancers12102772] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 11/16/2022] Open
Abstract
Optimal use of multiparametric magnetic resonance imaging (mpMRI) can identify key MRI parameters and provide unique tissue signatures defining phenotypes of breast cancer. We have developed and implemented a new machine-learning informatic system, termed Informatics Radiomics Integration System (IRIS) that integrates clinical variables, derived from imaging and electronic medical health records (EHR) with multiparametric radiomics (mpRad) for identifying potential risk of local or systemic recurrence in breast cancer patients. We tested the model in patients (n = 80) who had Estrogen Receptor positive disease and underwent OncotypeDX gene testing, radiomic analysis, and breast mpMRI. The IRIS method was trained using the mpMRI, clinical, pathologic, and radiomic descriptors for prediction of the OncotypeDX risk score. The trained mpRad IRIS model had a 95% and specificity was 83% with an Area Under the Curve (AUC) of 0.89 for classifying low risk patients from the intermediate and high-risk groups. The lesion size was larger for the high-risk group (2.9 ± 1.7 mm) and lower for both low risk (1.9 ± 1.3 mm) and intermediate risk (1.7 ± 1.4 mm) groups. The lesion apparent diffusion coefficient (ADC) map values for high- and intermediate-risk groups were significantly (p < 0.05) lower than the low-risk group (1.14 vs. 1.49 × 10-3 mm2/s). These initial studies provide deeper insight into the clinical, pathological, quantitative imaging, and radiomic features, and provide the foundation to relate these features to the assessment of treatment response for improved personalized medicine.
Collapse
Affiliation(s)
- Michael A. Jacobs
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (V.S.P.); (K.J.M.); (S.H.)
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (C.B.U.); (A.C.W.)
| | - Christopher B. Umbricht
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (C.B.U.); (A.C.W.)
| | - Vishwa S. Parekh
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (V.S.P.); (K.J.M.); (S.H.)
- Department of Computer Science, The Johns Hopkins University, Baltimore, MD 21210, USA
| | - Riham H. El Khouli
- Department of Radiology and Radiological Sciences, University of Kentucky, Lexington, KY 40536, USA;
| | - Leslie Cope
- Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
| | - Katarzyna J. Macura
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (V.S.P.); (K.J.M.); (S.H.)
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (C.B.U.); (A.C.W.)
| | - Susan Harvey
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (V.S.P.); (K.J.M.); (S.H.)
- Hologic Inc., 36 Apple Ridge Rd. Danbury, CT 06810, USA
| | - Antonio C. Wolff
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (C.B.U.); (A.C.W.)
| |
Collapse
|
41
|
Wang Y, Wang Y, Guo C, Xie X, Liang S, Zhang R, Pang W, Huang L. Cancer genotypes prediction and associations analysis from imaging phenotypes: a survey on radiogenomics. Biomark Med 2020; 14:1151-1164. [PMID: 32969248 DOI: 10.2217/bmm-2020-0248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this paper, we present a survey on the progress of radiogenomics research, which predicts cancer genotypes from imaging phenotypes and investigates the associations between them. First, we present an overview of the popular technology modalities for obtaining diagnostic medical images. Second, we summarize recently used methodologies for radiogenomics analysis, including statistical analysis, radiomics and deep learning. And then, we give a survey on the recent research based on several types of cancers. Finally, we discuss these studies and propose possible future research directions. In conclusion, we have identified strong correlations between cancer genotypes and imaging phenotypes. In addition, with the rapid growth of medical data, deep learning models show great application potential for radiogenomics.
Collapse
Affiliation(s)
- Yao Wang
- Key Laboratory of Symbol Computation & Knowledge Engineering, Ministry of Education, College of Computer Science & Technology, Jilin University, Changchun, 130012, PR China
| | - Yan Wang
- Key Laboratory of Symbol Computation & Knowledge Engineering, Ministry of Education, College of Computer Science & Technology, Jilin University, Changchun, 130012, PR China.,School of Artificial Intelligence, Jilin University, Changchun 130012, PR China
| | - Chunjie Guo
- Department of Radiology, The First Hospital of Jilin University, Changchun 130012, PR China
| | - Xuping Xie
- Key Laboratory of Symbol Computation & Knowledge Engineering, Ministry of Education, College of Computer Science & Technology, Jilin University, Changchun, 130012, PR China
| | - Sen Liang
- State Key Lab of CAD & CG, Zhejiang University, Hangzhou 310058, PR China
| | - Ruochi Zhang
- School of Artificial Intelligence, Jilin University, Changchun 130012, PR China
| | - Wei Pang
- School of Mathematical & Computer Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Lan Huang
- Key Laboratory of Symbol Computation & Knowledge Engineering, Ministry of Education, College of Computer Science & Technology, Jilin University, Changchun, 130012, PR China.,Zhuhai Laboratory of Key Laboratory of Symbolic Computation & Knowledge Engineering of Ministry of Education, Department of Computer Science & Technology, Zhuhai College of Jilin University, Zhuhai 519041, China
| |
Collapse
|
42
|
Bhatia A, Birger M, Veeraraghavan H, Um H, Tixier F, McKenney AS, Cugliari M, Caviasco A, Bialczak A, Malani R, Flynn J, Zhang Z, Yang TJ, Santomasso BD, Shoushtari AN, Young RJ. MRI radiomic features are associated with survival in melanoma brain metastases treated with immune checkpoint inhibitors. Neuro Oncol 2020; 21:1578-1586. [PMID: 31621883 DOI: 10.1093/neuonc/noz141] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Melanoma brain metastases historically portend a dismal prognosis, but recent advances in immune checkpoint inhibitors (ICIs) have been associated with durable responses in some patients. There are no validated imaging biomarkers associated with outcomes in patients with melanoma brain metastases receiving ICIs. We hypothesized that radiomic analysis of magnetic resonance images (MRIs) could identify higher-order features associated with survival. METHODS Between 2010 and 2019, we retrospectively reviewed patients with melanoma brain metastases who received ICI. After volumes of interest were drawn, several texture and edge descriptors, including first-order, Haralick, Gabor, Sobel, and Laplacian of Gaussian (LoG) features were extracted. Progression was determined using Response Assessment in Neuro-Oncology Brain Metastases. Univariate Cox regression was performed for each radiomic feature with adjustment for multiple comparisons followed by Lasso regression and multivariate analysis. RESULTS Eighty-eight patients with 196 total brain metastases were identified. Median age was 63.5 years (range, 19-91 y). Ninety percent of patients had Eastern Cooperative Oncology Group performance status of 0 or 1 and 35% had elevated lactate dehydrogenase. Sixty-three patients (72%) received ipilimumab, 11 patients (13%) received programmed cell death protein 1 blockade, and 14 patients (16%) received nivolumab plus ipilimumab. Multiple features were associated with increased overall survival (OS), and LoG edge features best explained the variation in outcome (hazard ratio: 0.68, P = 0.001). In multivariate analysis, a similar trend with LoG was seen, but no longer significant with OS. Findings were confirmed in an independent cohort. CONCLUSION Higher-order MRI radiomic features in patients with melanoma brain metastases receiving ICI were associated with a trend toward improved OS.
Collapse
Affiliation(s)
- Ankush Bhatia
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maxwell Birger
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Harini Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hyemin Um
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Florent Tixier
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anna Sophia McKenney
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marina Cugliari
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Annalise Caviasco
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Angelica Bialczak
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rachna Malani
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zhigang Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - T Jonathan Yang
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bianca D Santomasso
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Robert J Young
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
43
|
Preliminary study on discriminating HER2 2+ amplification status of breast cancers based on texture features semi-automatically derived from pre-, post-contrast, and subtraction images of DCE-MRI. PLoS One 2020; 15:e0234800. [PMID: 32555662 PMCID: PMC7299320 DOI: 10.1371/journal.pone.0234800] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 06/02/2020] [Indexed: 01/10/2023] Open
Abstract
Objective To investigate whether texture features extracted from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) are associated with human epidermal growth factor receptor type 2 (HER2) 2+ status of breast cancer. Materials and methods 92 MRI cases including 52 HER2 2+ positive and 40 negative patients confirmed by fluorescence in situ hybridization were retrospectively selected. The lesion area was semi-automatically delineated, and a total of 488 texture features were respectively extracted from precontrast, postcontrast, and subtraction images. The Student’s t-test or Mann-Whitney U test was performed to identify statistically significant features between different HER2 2+ amplification groups. Least absolute shrinkage and selection operator (LASSO) was used to search for the optimal feature subsets. Three machine learning classifiers, logistic regression analysis (LRA), quadratic discriminant analysis (QDA), and support vector machine (SVM), were used with a leave-one-out cross validation method to establish the classification models of HER2 2+ status. Classification performance was evaluated by receiver operating characteristic (ROC) analysis. Results Based on the texture analysis with SVM model, the areas under the ROC curve (AUCs) were 0.890 for subtraction images, 0.736 for postcontrast images, and 0.672 for precontrast images, respectively. For LRA model, the AUCs were 0.884, 0.733, and 0.623, respectively. For QDA model, the AUCs were 0.831, 0.726, and 0.568, respectively. LRA and the SVM model with subtraction images reached significantly better performance than the QDA model (P = 0.0227 and P = 0.0088, respectively). Conclusion Texture features of breast cancer extracted from DCE-MRI are associated with HER2 2+ status. Additional studies are necessary to confirm the present preliminary findings.
Collapse
|
44
|
El Khouli RH, Jacobs MA. Use of MRI for Personalized Treatment of More Aggressive Tumors. Radiology 2020; 295:527-528. [PMID: 32233918 PMCID: PMC7263283 DOI: 10.1148/radiol.2020200678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/10/2020] [Accepted: 03/20/2020] [Indexed: 03/16/2025]
Affiliation(s)
- Riham H. El Khouli
- From the Department of Radiology, Nuclear Medicine and Molecular Imaging, University of Kentucky College of Medicine, 800 Rose St, Lexington, KY 40506-9983 (R.H.E.K.); and Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Md (M.A.J.)
| | - Michael A. Jacobs
- From the Department of Radiology, Nuclear Medicine and Molecular Imaging, University of Kentucky College of Medicine, 800 Rose St, Lexington, KY 40506-9983 (R.H.E.K.); and Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Md (M.A.J.)
| |
Collapse
|
45
|
Ahmed AA, Elmohr MM, Fuentes D, Habra MA, Fisher SB, Perrier ND, Zhang M, Elsayes KM. Radiomic mapping model for prediction of Ki-67 expression in adrenocortical carcinoma. Clin Radiol 2020; 75:479.e17-479.e22. [PMID: 32089260 DOI: 10.1016/j.crad.2020.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/23/2020] [Indexed: 11/18/2022]
Abstract
AIM To determine the value of contrast-enhanced computed tomography (CT)-derived radiomic features in the preoperative prediction of Ki-67 expression in adrenocortical carcinoma (ACC) and to detect significant associations between radiomic features and Ki-67 expression in ACC. MATERIALS AND METHODS For this retrospective analysis, patients with histopathologically proven ACC were reviewed. Radiomic features were extracted for all patients from the preoperative contrast-enhanced abdominal CT images. Statistical analysis identified the radiomic features predicting the Ki-67 index in ACC and analysed the correlation with the Ki-67 index. RESULTS Fifty-three cases of ACC that met eligibility criteria were identified and analysed. Of the radiomic features analysed, 10 showed statistically significant differences between the high and low Ki-67 expression subgroups. Multivariate linear regression analysis yielded a predictive model showing a significant association between radiomic signature and Ki-67 expression status in ACC (R2=0.67, adjusted R2=0.462, p=0.002). Further analysis of the independent predictors showed statistically significant correlation between Ki-67 expression and shape flatness, elongation, and grey-level long run emphasis (p=0.002, 0.01, and 0.04, respectively). The area under the curve for identification of high Ki-67 expression status was 0.78 for shape flatness and 0.7 for shape elongation. CONCLUSION Radiomic features derived from preoperative contrast-enhanced CT images show encouraging results in the prediction of the Ki-67 index in patients with ACC. Morphological features, such as shape flatness and elongation, were superior to other radiomic features in the detection of high Ki-67 expression.
Collapse
Affiliation(s)
- A A Ahmed
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - M M Elmohr
- Department Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - D Fuentes
- Department Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - M A Habra
- Department Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - S B Fisher
- Department Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - N D Perrier
- Department Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - M Zhang
- Department Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - K M Elsayes
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
46
|
Yuen S, Monzawa S, Yanai S, Matsumoto H, Yata Y, Ichinose Y, Deai T, Hashimoto T, Tashiro T, Yamagami K. The association between MRI findings and breast cancer subtypes: focused on the combination patterns on diffusion-weighted and T2-weighted images. Breast Cancer 2020; 27:1029-1037. [PMID: 32377938 DOI: 10.1007/s12282-020-01105-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/28/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE To assess morphology on diffusion-weighted imaging (DWI) and intratumoral signal intensity (SI) on T2-weighted images (T2WI) of breast carcinomas, and to evaluate the association between the combined DWI and T2WI findings and breast cancer subtypes. METHODS Two hundred and eighty breast cancer patients who underwent breast MRI prior to therapy were included in this retrospective study. All had invasive carcinomas, which were classified into five subtypes: Luminal A-like (n = 149), Luminal B-like (n = 63), Hormone receptor-positive HER2 (n = 31), Hormone receptor-negative HER2 (n = 13), or Triple-negative (TN) (n = 24). Based on the morphology on DWI, the tumors were classified into two patterns: DWI-homogeneous or DWI-heterogeneous. If DWI-heterogeneous, an assessment of intratumoral SI on T2WI was performed: tumors with intratumoral high/low SI on T2WI were classified as Hete-H/Hete-L, respectively. The associations between (1) the morphological patterns on DWI and the five subtypes, and (2) the intratumoral SI patterns on T2WI and the five subtypes in DWI-heterogeneous were evaluated. RESULTS There was a significant association between (1) the morphological patterns on DWI and the five subtypes (p < 0.0001), and (2) the intratumoral SI patterns on T2WI and the five subtypes in DWI-heterogeneous (p < 0.0001). DWI-homogeneous was dominant in Luminal A-like (67.1%), and Hete-H was dominant in TN type (75%). Hete-H, suggesting the presence of intratumoral necrosis, included high proliferative and/or aggressive subtypes more frequently (80%) than Hete-L, suggesting the presence of fibrotic focus. Fibrotic focus was seen more commonly in the luminal subtypes. CONCLUSION The combined findings on DWI and T2WI revealed breast carcinomas that were associated with particular subtypes.
Collapse
Affiliation(s)
- Sachiko Yuen
- Breast Research Center, The Shinko Institution for Medical Research, Shinko Hospital, 1-4-47, Wakinohama Chuo, Kobe, 651-0072, Japan. .,Department of Breast Surgery and Oncology, Shinko Hospital, Kobe, Japan.
| | - Shuichi Monzawa
- Department of Diagnostic Radiology, Shinko Hospital, Kobe, Japan
| | - Seiji Yanai
- Breast Research Center, The Shinko Institution for Medical Research, Shinko Hospital, 1-4-47, Wakinohama Chuo, Kobe, 651-0072, Japan.,Department of Breast Surgery and Oncology, Shinko Hospital, Kobe, Japan
| | - Hajime Matsumoto
- Breast Research Center, The Shinko Institution for Medical Research, Shinko Hospital, 1-4-47, Wakinohama Chuo, Kobe, 651-0072, Japan.,Department of Breast Surgery and Oncology, Shinko Hospital, Kobe, Japan
| | - Yoshihiro Yata
- Breast Research Center, The Shinko Institution for Medical Research, Shinko Hospital, 1-4-47, Wakinohama Chuo, Kobe, 651-0072, Japan.,Department of Breast Surgery and Oncology, Shinko Hospital, Kobe, Japan
| | - You Ichinose
- Department of Breast Surgery and Oncology, Shinko Hospital, Kobe, Japan
| | - Teruyuki Deai
- Department of Breast Surgery and Oncology, Shinko Hospital, Kobe, Japan
| | - Takashi Hashimoto
- Department of Breast Surgery and Oncology, Shinko Hospital, Kobe, Japan
| | | | - Kazuhiko Yamagami
- Breast Research Center, The Shinko Institution for Medical Research, Shinko Hospital, 1-4-47, Wakinohama Chuo, Kobe, 651-0072, Japan.,Department of Breast Surgery and Oncology, Shinko Hospital, Kobe, Japan
| |
Collapse
|
47
|
Lu H, Yin J. Texture Analysis of Breast DCE-MRI Based on Intratumoral Subregions for Predicting HER2 2+ Status. Front Oncol 2020; 10:543. [PMID: 32373531 PMCID: PMC7186477 DOI: 10.3389/fonc.2020.00543] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/26/2020] [Indexed: 01/04/2023] Open
Abstract
Background: Breast tumor heterogeneity is related to risk factors that lead to aggressive tumor growth; however, such heterogeneity has not been thoroughly investigated. Purpose: To evaluate the performance of texture features extracted from heterogeneity subregions on subtraction MRI images for identifying human epidermal growth factor receptor 2 (HER2) 2+ status of breast cancers. Materials and Methods: Seventy-six patients with HER2 2+ breast cancer who underwent dynamic contrast-enhanced magnetic resonance imaging were enrolled, including 42 HER2 positive and 34 negative cases confirmed by fluorescence in situ hybridization. The lesion area was delineated semi-automatically on the subtraction MRI images at the second, fourth, and sixth phases (P-1, P-2, and P-3). A regionalization method was used to segment the lesion area into three subregions (rapid, medium, and slow) according to peak arrival time of the contrast agent. We extracted 488 texture features from the whole lesion area and three subregions independently. Wrapper, least absolute shrinkage and selection operator (LASSO), and stepwise methods were used to identify the optimal feature subsets. Univariate analysis was performed as well as support vector machine (SVM) with a leave-one-out-based cross-validation method. The area under the receiver operating characteristic curve (AUC) was calculated to evaluate the performance of the classifiers. Results: In univariate analysis, the variance from medium subregion at P-2 was the best-performing feature for distinguishing HER2 2+ status (AUC = 0.836); for the whole lesion region, the variance at P-2 achieved the best performance (AUC = 0.798). There was no significant difference between the two methods (P = 0.271). In the machine learning with SVM, the best performance (AUC = 0.929) was achieved with LASSO from rapid subregion at P-2; for the whole region, the highest AUC value was 0.847 obtained at P-2 with LASSO. The difference was significant between the two methods (P = 0.021). Conclusion: The texture analysis of heterogeneity subregions based on intratumoral regionalization method showed potential value for recognizing HER2 2+ status in breast cancer.
Collapse
Affiliation(s)
- Hecheng Lu
- School of Medicine and Bioinformatics Engineering, Northeastern University, Shenyang, China.,Department of Radiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - Jiandong Yin
- Department of Radiology, ShengJing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
48
|
Tokuda Y, Yanagawa M, Minamitani K, Naoi Y, Noguchi S, Tomiyama N. Radiogenomics of magnetic resonance imaging and a new multi-gene classifier for predicting recurrence prognosis in estrogen receptor-positive breast cancer: A preliminary study. Medicine (Baltimore) 2020; 99:e19664. [PMID: 32311939 PMCID: PMC7220792 DOI: 10.1097/md.0000000000019664] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
To examine the correlation of qualitative and quantitative dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) results with 95-gene classifier or Curebest 95-gene classifier Breast (95GC) results for recurrence prediction in estrogen receptor-positive breast cancer (ERPBC).This retrospective study included 78 ERPBC patients (age range, 24-74 years) classified into high- (n = 33) and low- (n = 45) risk groups for recurrence based on 95GC and who underwent DCE-MRI between July 2006 and November 2012. For qualitative evaluation, mass shape, margin, and internal enhancement based on BI-RADS MRI lexicon and multiplicity were determined by consensus interpretation by 2 breast radiologists. For quantitative evaluation, mass size, volume ratios of the DCE-MRI kinetics, and both the kurtosis and the skewness of the intensity histogram for the whole mass in the initial and delayed phases were determined. Differences between the 2 risk-groups were analyzed using univariate logistic regression analyses and multiple logistic regression analyses. Receiver-operating characteristic curve cut-off values were used to define the groups.As for the qualitative findings, the difference between the 2 groups was not significant. For the quantitative data, the volume ratio of "medium" in the initial phase differed significantly between the 2 groups (P = .049). The volume ratio of "medium" (P = .006) and of "slow-persistent" (P = .005), and the delayed phase kurtosis (P = .012) in the univariate logistic regression analyses, and in the multiple logistic regression, volume ratio of "medium" >38.9% and delayed phase kurtosis >3.31 were identified as significant high-risk indicators (odds ratio, 5.83 and 3.55; 95% confidence interval, 1.58 to 21.42 and 1.24 to 10.15; P = .008 and P = .018, respectively).A high volume ratio of "medium" in the initial phase and/or high kurtosis in the delayed phase for quantitative evaluation could predict high ERPBC recurrence risk based on 95GC.
Collapse
Affiliation(s)
- Yukiko Tokuda
- Department of Radiology, Osaka University Graduate School of Medicine
| | - Masahiro Yanagawa
- Department of Radiology, Osaka University Graduate School of Medicine
| | | | - Yasuto Naoi
- Breast oncology and surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinzaburo Noguchi
- Breast oncology and surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Noriyuki Tomiyama
- Department of Radiology, Osaka University Graduate School of Medicine
| |
Collapse
|
49
|
Lo Gullo R, Daimiel I, Morris EA, Pinker K. Combining molecular and imaging metrics in cancer: radiogenomics. Insights Imaging 2020; 11:1. [PMID: 31901171 PMCID: PMC6942081 DOI: 10.1186/s13244-019-0795-6] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background Radiogenomics is the extension of radiomics through the combination of genetic and radiomic data. Because genetic testing remains expensive, invasive, and time-consuming, and thus unavailable for all patients, radiogenomics may play an important role in providing accurate imaging surrogates which are correlated with genetic expression, thereby serving as a substitute for genetic testing. Main body In this article, we define the meaning of radiogenomics and the difference between radiomics and radiogenomics. We provide an up-to-date review of the radiomics and radiogenomics literature in oncology, focusing on breast, brain, gynecological, liver, kidney, prostate and lung malignancies. We also discuss the current challenges to radiogenomics analysis. Conclusion Radiomics and radiogenomics are promising to increase precision in diagnosis, assessment of prognosis, and prediction of treatment response, providing valuable information for patient care throughout the course of the disease, given that this information is easily obtainable with imaging. Larger prospective studies and standardization will be needed to define relevant imaging biomarkers before they can be implemented into the clinical workflow.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA.
| | - Isaac Daimiel
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA
| | - Elizabeth A Morris
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA
| | - Katja Pinker
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA.,Department of Biomedical Imaging and Image-guided Therapy, Molecular and Gender Imaging Service, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Wien, Austria
| |
Collapse
|
50
|
Grimm LJ, Mazurowski MA. Breast Cancer Radiogenomics: Current Status and Future Directions. Acad Radiol 2020; 27:39-46. [PMID: 31818385 DOI: 10.1016/j.acra.2019.09.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/17/2019] [Accepted: 09/08/2019] [Indexed: 12/13/2022]
Abstract
Radiogenomics is an area of research that aims to identify associations between imaging phenotypes ("radio-") and tumor genome ("-genomics"). Breast cancer radiogenomics research in particular has been an especially prolific area of investigation in recent years as evidenced by the wide number and variety of publications and conferences presentations. To date, research has primarily been focused on dynamic contrast enhanced pre-operative breast MRI and breast cancer molecular subtypes, but investigations have extended to all breast imaging modalities as well as multiple additional genetic markers including those that are commercially available. Furthermore, both human and computer-extracted features as well as deep learning techniques have been explored. This review will summarize the specific imaging modalities used in radiogenomics analysis, describe the methods of extracting imaging features, and present the types of genomics, molecular, and related information used for analysis. Finally, the limitations and future directions of breast cancer radiogenomics research will be discussed.
Collapse
|