1
|
Luo HJ, Ren JL, Mei Guo L, Liang Niu J, Song XL. MRI-based machine learning radiomics for prediction of HER2 expression status in breast invasive ductal carcinoma. Eur J Radiol Open 2024; 13:100592. [PMID: 39149534 PMCID: PMC11324846 DOI: 10.1016/j.ejro.2024.100592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/08/2024] [Accepted: 07/14/2024] [Indexed: 08/17/2024] Open
Abstract
Background Human epidermal growth factor receptor 2 (HER2) is a tumor biomarker with significant prognostic and therapeutic implications for invasive ductal breast carcinoma (IDC). Objective This study aimed to explore the effectiveness of a multisequence magnetic resonance imaging (MRI)-based machine learning radiomics model in classifying the expression status of HER2, including HER2-positive, HER2-low, and HER2 completely negative (HER2-zero), among patients with IDC. Methods A total of 402 female patients with IDC confirmed through surgical pathology were enrolled and subsequently divided into a training group (n = 250, center I) and a validation group (n = 152, center II). Radiomics features were extracted from the preoperative MRI. A simulated annealing algorithm was used for key feature selection. Two classification tasks were performed: task 1, the classification of HER2-positive vs. HER2-negative (HER2-low and HER2-zero), and task 2, the classification of HER2-low vs. HER2-zero. Logistic regression, random forest (RF), and support vector machine were conducted to establish radiomics models. The performance of the models was evaluated using the area under the curve (AUC) of the operating characteristics (ROC). Results In total, 4506 radiomics features were extracted from multisequence MRI. A radiomics model for prediction of expression state of HER2 was successfully developed. Among the three classification algorithms, RF achieved the highest performance in classifying HER2-positive from HER2-negative and HER2-low from HER2-zero, with AUC values of 0.777 and 0.731, respectively. Conclusions Machine learning-based MRI radiomics may aid in the non-invasive prediction of the different expression status of HER2 in IDC.
Collapse
Affiliation(s)
- Hong-Jian Luo
- Department of Radiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zuiyi, Guizhou province, China
| | | | - Li Mei Guo
- Department of Radiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi province, China
| | - Jin Liang Niu
- Department of Radiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi province, China
| | - Xiao-Li Song
- Department of Radiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi province, China
| |
Collapse
|
2
|
Ma X, Zhang L, Xiao Q, Huang Y, Lin L, Peng W, Gong J, Gu Y. Predicting Prognosis of Phyllodes Tumors Using a Mammography- and Magnetic Resonance Imaging-Based Radiomics Model: A Preliminary Study. Clin Breast Cancer 2024; 24:e571-e582.e1. [PMID: 38839461 DOI: 10.1016/j.clbc.2024.05.006] [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: 11/30/2023] [Revised: 04/25/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
PURPOSE To investigate whether a radiomics model based on mammography (MG) and magnetic resonance imaging (MRI) can be used to predict disease-free survival (DFS) after phyllodes tumor (PT) surgery. METHOD About 131 PT patients who underwent MG and MRI before surgery between January 2010 and December 2020 were retrospectively enrolled, including 15 patients with recurrence and metastasis and 116 without recurrence. 884 and 3138 radiomic features were extracted from MG and MR images, respectively. Then, multiple radiomics models were established to predict the recurrence risk of the patients by applying a support vector machine classifier. The area under the ROC curve (AUC) was calculated to evaluate model performance. After dividing the patients into high- and low-risk groups based on the predicted radiomics scores, survival analysis was conducted to compare differences between the groups. RESULTS In total, 3 MG-related and 5 MRI-related radiomic models were established; the prediction performance of the T1WI feature fusion model was the best, with an AUC value of 0.93. After combining the features of MG and MRI, the AUC increased to 0.95. Furthermore, the MG, MRI and all-image radiomic models had statistically significant differences in survival between the high- and low-risk groups (P < .001). All-image radiomics model showed higher survival performance than the MG and MRI radiomics models alone. CONCLUSIONS Radiomics features based on preoperative MG and MR images can predict DFS after PT surgery, and the prediction score of the image radiomics model can be used as a potential indicator of recurrence risk.
Collapse
Affiliation(s)
- Xiaowen Ma
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qin Xiao
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Huang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Luyi Lin
- 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
| | - 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.
| |
Collapse
|
3
|
Jiang C, Zhang X, Qu T, Yang X, Xiu Y, Yu X, Zhang S, Qiao K, Meng H, Li X, Huang Y. The prediction of pCR and chemosensitivity for breast cancer patients using DLG3, RADL and Pathomics signatures based on machine learning and deep learning. Transl Oncol 2024; 46:101985. [PMID: 38805774 PMCID: PMC11154003 DOI: 10.1016/j.tranon.2024.101985] [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: 03/14/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Limited studies have investigated the predictive value of multiomics signatures (radiomics, deep learning features, pathological features and DLG3) in breast cancer patients who underwent neoadjuvant chemotherapy (NAC). However, no study has explored the relationships among radiomic, pathomic signatures and chemosensitivity. This study aimed to predict pathological complete response (pCR) using multiomics signatures, and to evaluate the predictive utility of radiomic and pathomic signatures for guiding chemotherapy selection. METHODS The oncogenic function of DLG3 was explored in breast cancer cells via DLG3 knockdown. Immunohistochemistry (IHC) was used to evaluate the relationship between DLG3 expression and docetaxel/epirubin sensitivity. Machine learning (ML) and deep learning (DL) algorithms were used to develop multiomics signatures. Survival analysis was conducted by K-M curves and log-rank. Multivariate logistic regression analysis was used to develop nomograms. RESULTS A total of 311 patients with malignant breast tumours who underwent NAC were retrospectively included in this multicentre study. Multiomics (DLG3, RADL and PATHO) signatures could accurately predict pCR (AUC: training: 0.900; testing: 0.814; external validation: 0.792). Its performance is also superior to that of clinical TNM staging and the single RADL signature in different cohorts. Patients in the low DLG3 group more easily achieved pCR, and those in the high RADL Signature_pCR and PATHO_Signature_pCR (OR = 7.93, 95 % CI: 3.49-18, P < 0.001) groups more easily achieved pCR. In the TEC regimen NAC group, patients who achieved pCR had a lower DLG3 score (4.00 ± 2.33 vs. 6.43 ± 3.01, P < 0.05). Patients in the low RADL_Signature_DLG3 and PATHO_Signature_DLG3 groups had lower DLG3 IHC scores (P < 0.05). Patients in the high RADL signature, PATHO signature and DLG3 signature groups had worse DFS and OS. CONCLUSIONS Multiomics signatures (RADL, PATHO and DLG3) demonstrated great potential in predicting the pCR of breast cancer patients who underwent NAC. The RADL and PATHO signatures are associated with DLG3 status and could help doctors or patients choose proper neoadjuvant chemotherapy regimens (TEC regimens). This simple, structured, convenient and inexpensive multiomics model could help clinicians and patients make treatment decisions.
Collapse
Affiliation(s)
- Cong Jiang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - XueFang Zhang
- Department of Pathology, The first people's hospital of Xiangtan City, Xiangtan 411100, China
| | - Tong Qu
- Department of Oncology, The second cancer hospital of Heilongjiang province, Harbin 150086, China
| | - Xinxin Yang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Yuting Xiu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Xiao Yu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Shiyuan Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Kun Qiao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Xuelian Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yuanxi Huang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China.
| |
Collapse
|
4
|
Wang M, Du S, Gao S, Zhao R, Liu S, Jiang W, Peng C, Chai R, Zhang L. MRI-based tumor shrinkage patterns after early neoadjuvant therapy in breast cancer: correlation with molecular subtypes and pathological response after therapy. Breast Cancer Res 2024; 26:26. [PMID: 38347619 PMCID: PMC10863121 DOI: 10.1186/s13058-024-01781-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/09/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND MRI-based tumor shrinkage patterns (TSP) after neoadjuvant therapy (NAT) have been associated with pathological response. However, the understanding of TSP after early NAT remains limited. We aimed to analyze the relationship between TSP after early NAT and pathological response after therapy in different molecular subtypes. METHODS We prospectively enrolled participants with invasive ductal breast cancers who received NAT and performed pretreatment DCE-MRI from September 2020 to August 2022. Early-stage MRIs were performed after the first (1st-MRI) and/or second (2nd-MRI) cycle of NAT. Tumor shrinkage patterns were categorized into four groups: concentric shrinkage, diffuse decrease (DD), decrease of intensity only (DIO), and stable disease (SD). Logistic regression analysis was performed to identify independent variables associated with pathologic complete response (pCR), and stratified analysis according to tumor hormone receptor (HR)/human epidermal growth factor receptor 2 (HER2) disease subtype. RESULTS 344 participants (mean age: 50 years, 113/345 [33%] pCR) with 345 tumors (1 bilateral) had evaluable 1st-MRI or 2nd-MRI to comprise the primary analysis cohort, of which 244 participants with 245 tumors had evaluable 1st-MRI (82/245 [33%] pCR) and 206 participants with 207 tumors had evaluable 2nd-MRI (69/207 [33%] pCR) to comprise the 1st- and 2nd-timepoint subgroup analysis cohorts, respectively. In the primary analysis, multivariate analysis showed that early DD pattern (OR = 12.08; 95% CI 3.34-43.75; p < 0.001) predicted pCR independently of the change in tumor size (OR = 1.37; 95% CI 0.94-2.01; p = 0.106) in HR+/HER2- subtype, and the change in tumor size was a strong pCR predictor in HER2+ (OR = 1.61; 95% CI 1.22-2.13; p = 0.001) and triple-negative breast cancer (TNBC, OR = 1.61; 95% CI 1.22-2.11; p = 0.001). Compared with the change in tumor size, the SD pattern achieved a higher negative predictive value in HER2+ and TNBC. The statistical significance of complete 1st-timepoint subgroup analysis was consistent with the primary analysis. CONCLUSION The diffuse decrease pattern in HR+/HER2- subtype and stable disease in HER2+ and TNBC after early NAT could serve as additional straightforward and comprehensible indicators of treatment response. TRIAL REGISTRATION Trial registration at https://www.chictr.org.cn/ . REGISTRATION NUMBER ChiCTR2000038578, registered September 24, 2020.
Collapse
Affiliation(s)
- Mengfan Wang
- Department of Radiology, The First Hospital of China Medical University, Nanjing North Street 155, Shenyang, 110001, Liaoning Province, China
| | - Siyao Du
- Department of Radiology, The First Hospital of China Medical University, Nanjing North Street 155, Shenyang, 110001, Liaoning Province, China
| | - Si Gao
- Department of Radiology, The First Hospital of China Medical University, Nanjing North Street 155, Shenyang, 110001, Liaoning Province, China
| | - Ruimeng Zhao
- Department of Radiology, The First Hospital of China Medical University, Nanjing North Street 155, Shenyang, 110001, Liaoning Province, China
| | - Shasha Liu
- Department of Radiology, The First Hospital of China Medical University, Nanjing North Street 155, Shenyang, 110001, Liaoning Province, China
| | - Wenhong Jiang
- Department of Radiology, The First Hospital of China Medical University, Nanjing North Street 155, Shenyang, 110001, Liaoning Province, China
| | - Can Peng
- Department of Radiology, The First Hospital of China Medical University, Nanjing North Street 155, Shenyang, 110001, Liaoning Province, China
| | - Ruimei Chai
- Department of Radiology, The First Hospital of China Medical University, Nanjing North Street 155, Shenyang, 110001, Liaoning Province, China
| | - Lina Zhang
- Department of Radiology, The First Hospital of China Medical University, Nanjing North Street 155, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
5
|
Yu Y, Wang Z, Wang Q, Su X, Li Z, Wang R, Guo T, Gao W, Wang H, Zhang B. Radiomic model based on magnetic resonance imaging for predicting pathological complete response after neoadjuvant chemotherapy in breast cancer patients. Front Oncol 2024; 13:1249339. [PMID: 38357424 PMCID: PMC10865896 DOI: 10.3389/fonc.2023.1249339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/02/2023] [Indexed: 02/16/2024] Open
Abstract
Purpose To establish a model combining radiomic and clinicopathological factors based on magnetic resonance imaging to predict pathological complete response (pCR) after neoadjuvant chemotherapy in breast cancer patients. Method MRI images and clinicopathologic data of 329 eligible breast cancer patients from the Affiliated Hospital of Qingdao University from August 2018 to August 2022 were included in this study. All patients received neoadjuvant chemotherapy (NAC), and imaging examinations were performed before and after NAC. A total of 329 patients were randomly allocated to a training set and a test set at a ratio of 7:3. We mainly studied the following three types of prediction models: radiomic models, clinical models, and clinical-radiomic models. All models were evaluated using subject operating characteristic curve analysis and area under the curve (AUC), decision curve analysis (DCA) and calibration curves. Results The AUCs of the clinical prediction model, independent imaging model and clinical combined imaging model in the training set were 0.864 0.968 and 0.984, and those in the test set were 0.724, 0.754 and 0.877, respectively. According to DCA and calibration curves, the clinical-radiomic model showed good predictive performance in both the training set and the test set, and we found that we had developed a more concise clinical-radiomic nomogram. Conclusion We have developed a clinical-radiomic model by integrating radiomic features and clinical factors to predict pCR after NAC in breast cancer patients, thereby contributing to the personalized treatment of patients.
Collapse
Affiliation(s)
- Yimiao Yu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhibo Wang
- Department of Gastroenterological Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qi Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaohui Su
- Department of Galactophore, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhenghao Li
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Galactophore, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruifeng Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tianhui Guo
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wen Gao
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haiji Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Biyuan Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Sun R, Wei L, Hou X, Chen Y, Han B, Xie Y, Nie S. Molecular-subtype guided automatic invasive breast cancer grading using dynamic contrast-enhanced MRI. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 242:107804. [PMID: 37716219 DOI: 10.1016/j.cmpb.2023.107804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 04/05/2023] [Accepted: 09/05/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND AND OBJECTIVES Histological grade and molecular subtype have presented valuable references in assigning personalized or precision medicine as the significant prognostic indicators representing biological behaviors of invasive breast cancer (IBC). To evaluate a two-stage deep learning framework for IBC grading that incorporates with molecular-subtype (MS) information using DCE-MRI. METHODS In Stage I, an innovative neural network called IOS2-DA is developed, which includes a dense atrous-spatial pyramid pooling block with a pooling layer (DA) and inception-octconved blocks with double kernel squeeze-and-excitations (IOS2). This method focuses on the imaging manifestation of IBC grades and performs preliminary prediction using a novel class F1-score loss function. In Stage II, a MS attention branch is introduced to fine-tune the integrated deep vectors from IOS2-DA via Kullback-Leibler divergence. The MS-guided information is weighted with preliminary results to obtain classification values, which are analyzed by ensemble learning for tumor grade prediction on three MRI post-contrast series. Objective assessment is quantitatively evaluated by receiver operating characteristic curve analysis. DeLong test is applied to measure statistical significance (P < 0.05). RESULTS The molecular-subtype guided IOS2-DA performs significantly better than the single IOS2-DA in terms of accuracy (0.927), precision (0.942), AUC (0.927, 95% CI: [0.908, 0.946]), and F1-score (0.930). The gradient-weighted class activation maps show that the feature representations extracted from IOS2-DA are consistent with tumor areas. CONCLUSIONS IOS2-DA elucidates its potential in non-invasive tumor grade prediction. With respect to the correlation between MS and histological grade, it exhibits remarkable clinical prospects in the application of relevant clinical biomarkers to enhance the diagnostic effectiveness of IBC grading. Therefore, DCE-MRI tends to be a feasible imaging modality for the thorough preoperative assessment of breast biological behavior and carcinoma prognosis.
Collapse
Affiliation(s)
- Rong Sun
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jun-Gong Road, Shanghai 200093, China
| | - Long Wei
- School of Computer Science and Technology, Shandong Jianzhu University, Shandong, China
| | - Xuewen Hou
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jun-Gong Road, Shanghai 200093, China
| | - Yang Chen
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jun-Gong Road, Shanghai 200093, China
| | - Baosan Han
- Department of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University School of Medicine, China.
| | - Yuanzhong Xie
- Medical Imaging Center, Tai'an Central Hospital, No. 29 Long-Tan Road, Shandong 271099, China.
| | - Shengdong Nie
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jun-Gong Road, Shanghai 200093, China.
| |
Collapse
|
7
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
8
|
Zhao MR, Ma WJ, Song XC, Li ZJ, Shao ZZ, Lu H, Zhao R, Guo YJ, Ye ZX, Liu PF. Feasibility analysis of magnetic resonance imaging-based radiomics features for preoperative prediction of nuclear grading of ductal carcinoma in situ. Gland Surg 2023; 12:1209-1223. [PMID: 37842532 PMCID: PMC10570967 DOI: 10.21037/gs-23-132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/27/2023] [Indexed: 10/17/2023]
Abstract
Background The nuclear grading of ductal carcinoma in situ (DCIS) affects its clinical risk. The aim of this study was to investigate the possibility of predicting the nuclear grading of DCIS, by magnetic resonance imaging (MRI)-based radiomics features. And to develop a nomogram combining radiomics features and MRI semantic features to explore the potential role of MRI radiomic features in the assessment of DCIS nuclear grading. Methods A total of 156 patients (159 lesions) with DCIS and DCIS with microinvasive (DCIS-MI) were enrolled in this retrospective study, with 112 lesions included in the training cohort and 47 lesions included in the validation cohort. Radiomics features were extracted from Dynamic contrast-enhanced MRI (DCE-MRI) phases 1st and 5th. After feature selection, radiomics signature was constructed and radiomics score (Rad-score) was calculated. Multivariate analysis was used to identify MRI semantic features that were significantly associated with DCIS nuclear grading and combined with Rad-score to construct a Nomogram. Receiver operating characteristic curves were used to evaluate the predictive performance of Rad-score and Nomogram, and decision curve analysis (DCA) was used to evaluate the clinical utility. Results In multivariate analyses of MRI semantic features, larger tumor size and heterogeneous enhancement pattern were significantly associated with high-nuclear grade DCIS (HNG DCIS). In the training cohort, Nomogram had an area under curve (AUC) of 0.879 and Rad-score had an AUC of 0.828. Similarly, in the independent validation cohort, Nomogram had an AUC value of 0.828 and Rad-score had an AUC of 0.772. In both the training and validation cohorts, Nomogram had a significantly higher AUC value than Rad-score (P<0.05). DCA confirmed that Nomogram had a higher net clinical benefit. Conclusions MRI-based radiomic features can be used as potential biomarkers for assessing nuclear grading of DCIS. The nomogram constructed by radiomic features combined with semantic features is feasible in discriminating non-HNG and HNG DCIS.
Collapse
Affiliation(s)
- Meng-Ran Zhao
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wen-Juan Ma
- Department of Breast Imaging, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiang-Chao Song
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhi-Jun Li
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhen-Zhen Shao
- Department of Breast Imaging, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hong Lu
- Department of Breast Imaging, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Rui Zhao
- Department of Breast Imaging, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yi-Jun Guo
- Department of Breast Imaging, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhao-Xiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Pei-Fang Liu
- Department of Breast Imaging, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
9
|
Sun MX, Zhao MJ, Zhao LH, Jiang HR, Duan YX, Li G. A nomogram model based on pre-treatment and post-treatment MR imaging radiomics signatures: application to predict progression-free survival for nasopharyngeal carcinoma. Radiat Oncol 2023; 18:67. [PMID: 37041545 PMCID: PMC10088158 DOI: 10.1186/s13014-023-02257-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 04/03/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND To establish a novel model using radiomics analysis of pre-treatment and post-treatment magnetic resonance (MR) images for prediction of progression-free survival in the patients with stage II-IVA nasopharyngeal carcinoma (NPC) in South China. METHODS One hundred and twenty NPC patients who underwent chemoradiotherapy were enrolled (80 in the training cohort and 40 in the validation cohort). Acquiring data and screening features were performed successively. Totally 1133 radiomics features were extracted from the T2-weight images before and after treatment. Least absolute shrinkage and selection operator regression, recursive feature elimination algorithm, random forest, and minimum-redundancy maximum-relevancy (mRMR) method were used for feature selection. Nomogram discrimination and calibration were evaluated. Harrell's concordance index (C-index) and receiver operating characteristic (ROC) analyses were applied to appraise the prognostic performance of nomograms. Survival curves were plotted using Kaplan-Meier method. RESULTS Integrating independent clinical predictors with pre-treatment and post-treatment radiomics signatures which were calculated in conformity with radiomics features, we established a clinical-and-radiomics nomogram by multivariable Cox regression. Nomogram consisting of 14 pre-treatment and 7 post-treatment selected features has been proved to yield a reliable predictive performance in both training and validation groups. The C-index of clinical-and-radiomics nomogram was 0.953 (all P < 0.05), which was higher than that of clinical (0.861) or radiomics nomograms alone (based on pre-treatment statistics: 0.942; based on post-treatment statistics: 0.944). Moreover, we received Rad-score of pre-treatment named RS1 and post-treatment named RS2 and all were used as independent predictors to divide patients into high-risk and low-risk groups. Kaplan-Meier analysis showed that lower RS1 (less than cutoff value, - 1.488) and RS2 (less than cutoff value, - 0.180) were easier to avoid disease progression (all P < 0.01). It showed clinical benefit with decision curve analysis. CONCLUSIONS MR-based radiomics measured the burden on primary tumor before treatment and the tumor regression after chemoradiotherapy, and was used to build a model to predict progression-free survival (PFS) in the stage II-IVA NPC patients. It can also help to distinguish high-risk patients from low-risk patients, thus guiding personalized treatment decisions effectively.
Collapse
Affiliation(s)
- Mi-Xue Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Meng-Jing Zhao
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Li-Hao Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Hao-Ran Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yu-Xia Duan
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| | - Gang Li
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
10
|
Chitalia R, Miliotis M, Jahani N, Tastsoglou S, McDonald ES, Belenky V, Cohen EA, Newitt D, Van't Veer LJ, Esserman L, Hylton N, DeMichele A, Hatzigeorgiou A, Kontos D. Radiomic tumor phenotypes augment molecular profiling in predicting recurrence free survival after breast neoadjuvant chemotherapy. COMMUNICATIONS MEDICINE 2023; 3:46. [PMID: 36997615 PMCID: PMC10063641 DOI: 10.1038/s43856-023-00273-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 03/10/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Early changes in breast intratumor heterogeneity during neoadjuvant chemotherapy may reflect the tumor's ability to adapt and evade treatment. We investigated the combination of precision medicine predictors of genomic and MRI data towards improved prediction of recurrence free survival (RFS). METHODS A total of 100 women from the ACRIN 6657/I-SPY 1 trial were retrospectively analyzed. We estimated MammaPrint, PAM50 ROR-S, and p53 mutation scores from publicly available gene expression data and generated four, voxel-wise 3-D radiomic kinetic maps from DCE-MR images at both pre- and early-treatment time points. Within the primary lesion from each kinetic map, features of change in radiomic heterogeneity were summarized into 6 principal components. RESULTS We identify two imaging phenotypes of change in intratumor heterogeneity (p < 0.01) demonstrating significant Kaplan-Meier curve separation (p < 0.001). Adding phenotypes to established prognostic factors, functional tumor volume (FTV), MammaPrint, PAM50, and p53 scores in a Cox regression model improves the concordance statistic for predicting RFS from 0.73 to 0.79 (p = 0.002). CONCLUSIONS These results demonstrate an important step in combining personalized molecular signatures and longitudinal imaging data towards improved prognosis.
Collapse
Affiliation(s)
- Rhea Chitalia
- Department of Bioengineering, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Marios Miliotis
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- DIANA-Lab, Hellenic Pasteur Institute, Athens, Greece
| | - Nariman Jahani
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Spyros Tastsoglou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- DIANA-Lab, Hellenic Pasteur Institute, Athens, Greece
| | - Elizabeth S McDonald
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Vivian Belenky
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Eric A Cohen
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - David Newitt
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA
| | - Laura J Van't Veer
- Department of Surgery and Oncology, University of California, San Francisco, USA
| | - Laura Esserman
- Department of Surgery and Oncology, University of California, San Francisco, USA
| | - Nola Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA
| | - Angela DeMichele
- Department of Medicine, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Artemis Hatzigeorgiou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- DIANA-Lab, Hellenic Pasteur Institute, Athens, Greece
| | - Despina Kontos
- Department of Radiology, Division of Hematology/Oncology, University of Pennsylvania, Perelman School of Medicine 3400 Spruce Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
11
|
Pesapane F, De Marco P, Rapino A, Lombardo E, Nicosia L, Tantrige P, Rotili A, Bozzini AC, Penco S, Dominelli V, Trentin C, Ferrari F, Farina M, Meneghetti L, Latronico A, Abbate F, Origgi D, Carrafiello G, Cassano E. How Radiomics Can Improve Breast Cancer Diagnosis and Treatment. J Clin Med 2023; 12:jcm12041372. [PMID: 36835908 PMCID: PMC9963325 DOI: 10.3390/jcm12041372] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Recent technological advances in the field of artificial intelligence hold promise in addressing medical challenges in breast cancer care, such as early diagnosis, cancer subtype determination and molecular profiling, prediction of lymph node metastases, and prognostication of treatment response and probability of recurrence. Radiomics is a quantitative approach to medical imaging, which aims to enhance the existing data available to clinicians by means of advanced mathematical analysis using artificial intelligence. Various published studies from different fields in imaging have highlighted the potential of radiomics to enhance clinical decision making. In this review, we describe the evolution of AI in breast imaging and its frontiers, focusing on handcrafted and deep learning radiomics. We present a typical workflow of a radiomics analysis and a practical "how-to" guide. Finally, we summarize the methodology and implementation of radiomics in breast cancer, based on the most recent scientific literature to help researchers and clinicians gain fundamental knowledge of this emerging technology. Alongside this, we discuss the current limitations of radiomics and challenges of integration into clinical practice with conceptual consistency, data curation, technical reproducibility, adequate accuracy, and clinical translation. The incorporation of radiomics with clinical, histopathological, and genomic information will enable physicians to move forward to a higher level of personalized management of patients with breast cancer.
Collapse
Affiliation(s)
- Filippo Pesapane
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
- Correspondence: ; Tel.: +39-02-574891
| | - Paolo De Marco
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Anna Rapino
- Postgraduation School in Radiodiagnostics, University of Milan, 20122 Milan, Italy
| | - Eleonora Lombardo
- UOC of Diagnostic Imaging, Policlinico Tor Vergata University, 00133 Rome, Italy
| | - Luca Nicosia
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Priyan Tantrige
- Department of Radiology, King’s College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Anna Rotili
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Anna Carla Bozzini
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Silvia Penco
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Valeria Dominelli
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Chiara Trentin
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Federica Ferrari
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Mariagiorgia Farina
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lorenza Meneghetti
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Antuono Latronico
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Francesca Abbate
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Daniela Origgi
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Gianpaolo Carrafiello
- Department of Radiology, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Health Sciences, University of Milan, 20122 Milan, Italy
| | - Enrico Cassano
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| |
Collapse
|
12
|
Huang EP, O'Connor JPB, McShane LM, Giger ML, Lambin P, Kinahan PE, Siegel EL, Shankar LK. Criteria for the translation of radiomics into clinically useful tests. Nat Rev Clin Oncol 2023; 20:69-82. [PMID: 36443594 PMCID: PMC9707172 DOI: 10.1038/s41571-022-00707-0] [Citation(s) in RCA: 109] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2022] [Indexed: 11/29/2022]
Abstract
Computer-extracted tumour characteristics have been incorporated into medical imaging computer-aided diagnosis (CAD) algorithms for decades. With the advent of radiomics, an extension of CAD involving high-throughput computer-extracted quantitative characterization of healthy or pathological structures and processes as captured by medical imaging, interest in such computer-extracted measurements has increased substantially. However, despite the thousands of radiomic studies, the number of settings in which radiomics has been successfully translated into a clinically useful tool or has obtained FDA clearance is comparatively small. This relative dearth might be attributable to factors such as the varying imaging and radiomic feature extraction protocols used from study to study, the numerous potential pitfalls in the analysis of radiomic data, and the lack of studies showing that acting upon a radiomic-based tool leads to a favourable benefit-risk balance for the patient. Several guidelines on specific aspects of radiomic data acquisition and analysis are already available, although a similar roadmap for the overall process of translating radiomics into tools that can be used in clinical care is needed. Herein, we provide 16 criteria for the effective execution of this process in the hopes that they will guide the development of more clinically useful radiomic tests in the future.
Collapse
Affiliation(s)
- Erich P Huang
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, MD, USA.
| | - James P B O'Connor
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Lisa M McShane
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | | | - Philippe Lambin
- Department of Precision Medicine, Maastricht University, Maastricht, Netherlands
| | - Paul E Kinahan
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Eliot L Siegel
- Department of Diagnostic Radiology, University of Maryland, Baltimore, MD, USA
| | - Lalitha K Shankar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
13
|
Caballo M, Sanderink WBG, Han L, Gao Y, Athanasiou A, Mann RM. Four-Dimensional Machine Learning Radiomics for the Pretreatment Assessment of Breast Cancer Pathologic Complete Response to Neoadjuvant Chemotherapy in Dynamic Contrast-Enhanced MRI. J Magn Reson Imaging 2023; 57:97-110. [PMID: 35633290 PMCID: PMC10083908 DOI: 10.1002/jmri.28273] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Breast cancer response to neoadjuvant chemotherapy (NAC) is typically evaluated through the assessment of tumor size reduction after a few cycles of NAC. In case of treatment ineffectiveness, this results in the patient suffering potentially severe secondary effects without achieving any actual benefit. PURPOSE To identify patients achieving pathologic complete response (pCR) after NAC by spatio-temporal radiomic analysis of dynamic contrast-enhanced (DCE) MRI images acquired before treatment. STUDY TYPE Single-center, retrospective. POPULATION A total of 251 DCE-MRI pretreatment images of breast cancer patients. FIELD STRENGTH/SEQUENCE 1.5 T/3 T, T1-weighted DCE-MRI. ASSESSMENT Tumor and peritumoral regions were segmented, and 348 radiomic features that quantify texture temporal variation, enhancement kinetics heterogeneity, and morphology were extracted. Based on subsets of features identified through forward selection, machine learning (ML) logistic regression models were trained separately with all images and stratifying on cancer molecular subtype and validated with leave-one-out cross-validation. STATISTICAL TESTS Feature significance was assessed using the Mann-Whitney U-test. Significance of the area under the receiver operating characteristics (ROC) curve (AUC) of the ML models was assessed using the associated 95% confidence interval (CI). Significance threshold was set to 0.05, adjusted with Bonferroni correction. RESULTS Nine features related to texture temporal variation and enhancement kinetics heterogeneity were significant in the discrimination of cases achieving pCR vs. non-pCR. The ML models achieved significant AUC of 0.707 (all cancers, n = 251, 59 pCR), 0.824 (luminal A, n = 107, 14 pCR), 0.823 (luminal B, n = 47, 15 pCR), 0.844 (HER2 enriched, n = 25, 11 pCR), 0.803 (triple negative, n = 72, 19 pCR). DATA CONCLUSIONS Differences in imaging phenotypes were found between complete and noncomplete responders. Furthermore, ML models trained per cancer subtype achieved high performance in classifying pCR vs. non-pCR cases. They may, therefore, have potential to help stratify patients according to the level of response predicted before treatment, pending further validation with larger prospective cohorts. EVIDENCE LEVEL 4 TECHNICAL EFFICACY: Stage 4.
Collapse
Affiliation(s)
- Marco Caballo
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Luyi Han
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Yuan Gao
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | | | - Ritse M Mann
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Radiomic and Volumetric Measurements as Clinical Trial Endpoints—A Comprehensive Review. Cancers (Basel) 2022; 14:cancers14205076. [PMID: 36291865 PMCID: PMC9599928 DOI: 10.3390/cancers14205076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary The extraction of quantitative data from standard-of-care imaging modalities offers opportunities to improve the relevance and salience of imaging biomarkers used in drug development. This review aims to identify the challenges and opportunities for discovering new imaging-based biomarkers based on radiomic and volumetric assessment in the single-site solid tumor sites: breast cancer, rectal cancer, lung cancer and glioblastoma. Developing approaches to harmonize three essential areas: segmentation, validation and data sharing may expedite regulatory approval and adoption of novel cancer imaging biomarkers. Abstract Clinical trials for oncology drug development have long relied on surrogate outcome biomarkers that assess changes in tumor burden to accelerate drug registration (i.e., Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1) criteria). Drug-induced reduction in tumor size represents an imperfect surrogate marker for drug activity and yet a radiologically determined objective response rate is a widely used endpoint for Phase 2 trials. With the addition of therapies targeting complex biological systems such as immune system and DNA damage repair pathways, incorporation of integrative response and outcome biomarkers may add more predictive value. We performed a review of the relevant literature in four representative tumor types (breast cancer, rectal cancer, lung cancer and glioblastoma) to assess the preparedness of volumetric and radiomics metrics as clinical trial endpoints. We identified three key areas—segmentation, validation and data sharing strategies—where concerted efforts are required to enable progress of volumetric- and radiomics-based clinical trial endpoints for wider clinical implementation.
Collapse
|
15
|
Jones MA, Islam W, Faiz R, Chen X, Zheng B. Applying artificial intelligence technology to assist with breast cancer diagnosis and prognosis prediction. Front Oncol 2022; 12:980793. [PMID: 36119479 PMCID: PMC9471147 DOI: 10.3389/fonc.2022.980793] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/04/2022] [Indexed: 12/27/2022] Open
Abstract
Breast cancer remains the most diagnosed cancer in women. Advances in medical imaging modalities and technologies have greatly aided in the early detection of breast cancer and the decline of patient mortality rates. However, reading and interpreting breast images remains difficult due to the high heterogeneity of breast tumors and fibro-glandular tissue, which results in lower cancer detection sensitivity and specificity and large inter-reader variability. In order to help overcome these clinical challenges, researchers have made great efforts to develop computer-aided detection and/or diagnosis (CAD) schemes of breast images to provide radiologists with decision-making support tools. Recent rapid advances in high throughput data analysis methods and artificial intelligence (AI) technologies, particularly radiomics and deep learning techniques, have led to an exponential increase in the development of new AI-based models of breast images that cover a broad range of application topics. In this review paper, we focus on reviewing recent advances in better understanding the association between radiomics features and tumor microenvironment and the progress in developing new AI-based quantitative image feature analysis models in three realms of breast cancer: predicting breast cancer risk, the likelihood of tumor malignancy, and tumor response to treatment. The outlook and three major challenges of applying new AI-based models of breast images to clinical practice are also discussed. Through this review we conclude that although developing new AI-based models of breast images has achieved significant progress and promising results, several obstacles to applying these new AI-based models to clinical practice remain. Therefore, more research effort is needed in future studies.
Collapse
Affiliation(s)
- Meredith A. Jones
- School of Biomedical Engineering, University of Oklahoma, Norman, OK, United States
| | - Warid Islam
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, United States
| | - Rozwat Faiz
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, United States
| | - Xuxin Chen
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, United States
| | - Bin Zheng
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, United States
| |
Collapse
|
16
|
Pesapane F, Agazzi GM, Rotili A, Ferrari F, Cardillo A, Penco S, Dominelli V, D'Ecclesiis O, Vignati S, Raimondi S, Bozzini A, Pizzamiglio M, Petralia G, Nicosia L, Cassano E. Prediction of the Pathological Response to Neoadjuvant Chemotherapy in Breast Cancer Patients With MRI-Radiomics: A Systematic Review and Meta-analysis. Curr Probl Cancer 2022; 46:100883. [PMID: 35914383 DOI: 10.1016/j.currproblcancer.2022.100883] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 12/30/2022]
Abstract
We performed a systematic review and a meta-analysis of studies using MRI-radiomics for predicting the pathological complete response in breast cancer patients undergoing neoadjuvant therapy , and we evaluated their methodological quality using the radiomics-quality-score (RQS). Random effects meta-analysis was performed pooling area under the receiver operating characteristics curves. Publication-bias was assessed using the Egger's test and visually inspecting the funnel plot. Forty-three studies were included in the qualitative review and 34 in the meta-analysis. Summary area under the receiver operating characteristics curve was 0,78 (95%CI:0,74-0,81). Heterogeneity according to the I2 statistic was substantial (71%) and there was no evidence of publication bias (P-value = 0,2). The average RQS was 12,7 (range:-1-26), with an intra-class correlation coefficient of 0.93 (95%CI:0.61-0.97). Year of publication, field intensity and synthetic RQS score do not appear to be moderators of the effect (P-value = 0.36, P-value = 0.28 and P-value = 0.92, respectively). MRI-radiomics may predict response to neoadjuvant therapy in breast cancer patients but the heterogeneity of the current studies is still substantial.
Collapse
Affiliation(s)
- Filippo Pesapane
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy.
| | | | - Anna Rotili
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Ferrari
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Andrea Cardillo
- Radiology Department, Università degli studi di Torino, Turin, Italy
| | - Silvia Penco
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Valeria Dominelli
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Oriana D'Ecclesiis
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Silvano Vignati
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Raimondi
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Anna Bozzini
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Pizzamiglio
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Petralia
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy; Division of Radiology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Luca Nicosia
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Enrico Cassano
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
17
|
Xu A, Chu X, Zhang S, Zheng J, Shi D, Lv S, Li F, Weng X. Prediction Breast Molecular Typing of Invasive Ductal Carcinoma Based on Dynamic Contrast Enhancement Magnetic Resonance Imaging Radiomics Characteristics: A Feasibility Study. Front Oncol 2022; 12:799232. [PMID: 35664741 PMCID: PMC9160981 DOI: 10.3389/fonc.2022.799232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To investigate the feasibility of radiomics in predicting molecular subtype of breast invasive ductal carcinoma (IDC) based on dynamic contrast enhancement magnetic resonance imaging (DCE-MRI). Methods A total of 303 cases with pathologically confirmed IDC from January 2018 to March 2021 were enrolled in this study, including 223 cases from Fudan University Shanghai Cancer Center (training/test set) and 80 cases from Shaoxing Central Hospital (validation set). All the cases were classified as HR+/Luminal, HER2-enriched, and TNBC according to immunohistochemistry. DCE-MRI original images were treated by semi-automated segmentation to initially extract original and wavelet-transformed radiomic features. The extended logistic regression with least absolute shrinkage and selection operator (LASSO) penalty was applied to identify the optimal radiomic features, which were then used to establish predictive models combined with significant clinical risk factors. Receiver operating characteristic curve (ROC), calibration curve, and decision curve analysis were adopted to evaluate the effectiveness and clinical benefit of the models established. Results Of the 223 cases from Fudan University Shanghai Cancer Center, HR+/Luminal cancers were diagnosed in 116 cases (52.02%), HER2-enriched in 71 cases (31.84%), and TNBC in 36 cases (16.14%). Based on the training set, 788 radiomic features were extracted in total and 8 optimal features were further identified, including 2 first-order features, 1 gray-level run length matrix (GLRLM), 4 gray-level co-occurrence matrices (GLCM), and 1 3D shape feature. Three multi-class classification models were constructed by extended logistic regression: clinical model (age, menopause, tumor location, Ki-67, histological grade, and lymph node metastasis), radiomic model, and combined model. The macro-average areas under the ROC curve (macro-AUC) for the three models were 0.71, 0.81, and 0.84 in the training set, 0.73, 0.81, and 0.84 in the test set, and 0.76, 0.82, and 0.83 in the validation set, respectively. Conclusion The DCE-MRI-based radiomic features are significant biomarkers for distinguishing molecular subtypes of breast cancer noninvasively. Notably, the classification performance could be improved with the fusion analysis of multi-modal features.
Collapse
Affiliation(s)
- Aqiao Xu
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| | - Xiufeng Chu
- Department of Surgical, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| | - Shengjian Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jing Zheng
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| | - Dabao Shi
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| | - Shasha Lv
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| | - Feng Li
- Department of Research Collaboration, Research & Development Center (R&D), Beijing Deepwise & League of Doctor of Philosophy (PHD) Technology Co., Ltd, Beijing, China
| | - Xiaobo Weng
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| |
Collapse
|
18
|
Hirsch L, Huang Y, Luo S, Rossi Saccarelli C, Lo Gullo R, Daimiel Naranjo I, Bitencourt AGV, Onishi N, Ko ES, Leithner D, Avendano D, Eskreis-Winkler S, Hughes M, Martinez DF, Pinker K, Juluru K, El-Rowmeim AE, Elnajjar P, Morris EA, Makse HA, Parra LC, Sutton EJ. Radiologist-Level Performance by Using Deep Learning for Segmentation of Breast Cancers on MRI Scans. Radiol Artif Intell 2022; 4:e200231. [PMID: 35146431 PMCID: PMC8823456 DOI: 10.1148/ryai.200231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/13/2021] [Accepted: 11/04/2021] [Indexed: 11/17/2022]
Abstract
Purpose To develop a deep network architecture that would achieve fully automated
radiologist-level segmentation of cancers at breast MRI. Materials and Methods In this retrospective study, 38 229 examinations (composed of
64 063 individual breast scans from 14 475 patients) were
performed in female patients (age range, 12–94 years; mean age,
52 years ± 10 [standard deviation]) who presented between 2002
and 2014 at a single clinical site. A total of 2555 breast cancers were
selected that had been segmented on two-dimensional (2D) images by
radiologists, as well as 60 108 benign breasts that served as
examples of noncancerous tissue; all these were used for model training.
For testing, an additional 250 breast cancers were segmented
independently on 2D images by four radiologists. Authors selected among
several three-dimensional (3D) deep convolutional neural network
architectures, input modalities, and harmonization methods. The outcome
measure was the Dice score for 2D segmentation, which was compared
between the network and radiologists by using the Wilcoxon signed rank
test and the two one-sided test procedure. Results The highest-performing network on the training set was a 3D U-Net with
dynamic contrast-enhanced MRI as input and with intensity normalized for
each examination. In the test set, the median Dice score of this network
was 0.77 (interquartile range, 0.26). The performance of the network was
equivalent to that of the radiologists (two one-sided test procedures
with radiologist performance of 0.69–0.84 as equivalence bounds,
P < .001 for both; n =
250). Conclusion When trained on a sufficiently large dataset, the developed 3D U-Net
performed as well as fellowship-trained radiologists in detailed 2D
segmentation of breast cancers at routine clinical MRI. Keywords: MRI, Breast, Segmentation, Supervised Learning,
Convolutional Neural Network (CNN), Deep Learning Algorithms, Machine
Learning Algorithms Published under a CC BY 4.0 license. Supplemental material is available for this
article.
Collapse
Affiliation(s)
- Lukas Hirsch
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Yu Huang
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Shaojun Luo
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Carolina Rossi Saccarelli
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Roberto Lo Gullo
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Isaac Daimiel Naranjo
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Almir G V Bitencourt
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Natsuko Onishi
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Eun Sook Ko
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Doris Leithner
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Daly Avendano
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Sarah Eskreis-Winkler
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Mary Hughes
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Danny F Martinez
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Katja Pinker
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Krishna Juluru
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Amin E El-Rowmeim
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Pierre Elnajjar
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Elizabeth A Morris
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Hernan A Makse
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Lucas C Parra
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| | - Elizabeth J Sutton
- Department of Biomedical Engineering (L.H., Y.H., L.C.P.) and the Benjamin Levich Institute and Department of Physics (S.L., H.A.M.), the City College of the City University of New York, 160 Convent Ave, New York, NY 10031; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 (Y.H., C.R.S., R.L.G., I.D.N., A.G.V.B., N.O., E.S.K., D.L., D.A., S.E.W., M.H., D.F.M., K.P., K.J., A.E.E., P.E., E.A.M., E.J.S.); Department of Imaging, A.C. Camargo Cancer Center, São Paulo, Brazil (A.G.V.B.); Department of Radiology, University of California, San Francisco, San Francisco, Calif (N.O.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (E.S.K.); and Department of Breast Imaging, Breast Cancer Center TecSalud, ITESM Monterrey, Monterrey, Mexico (D.A.)
| |
Collapse
|
19
|
Abstract
This article gives a brief overview of the development of artificial intelligence in clinical breast imaging. For multiple decades, artificial intelligence (AI) methods have been developed and translated for breast imaging tasks such as detection, diagnosis, and assessing response to therapy. As imaging modalities arise to support breast cancer screening programs and diagnostic examinations, including full-field digital mammography, breast tomosynthesis, ultrasound, and MRI, AI techniques parallel the efforts with more complex algorithms, faster computers, and larger data sets. AI methods include human-engineered radiomics algorithms and deep learning methods. Examples of these AI-supported clinical tasks are given along with commentary on the future.
Collapse
Affiliation(s)
- Qiyuan Hu
- Committee on Medical Physics, Department of Radiology, The University of Chicago, 5841 S Maryland Avenue, MC2026, Chicago, IL 60637, USA
| | - Maryellen L Giger
- Committee on Medical Physics, Department of Radiology, The University of Chicago, 5841 S Maryland Avenue, MC2026, Chicago, IL 60637, USA.
| |
Collapse
|
20
|
Machine Learning: Applications and Advanced Progresses of Radiomics in Endocrine Neoplasms. JOURNAL OF ONCOLOGY 2021; 2021:8615450. [PMID: 34671399 PMCID: PMC8523238 DOI: 10.1155/2021/8615450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/13/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
Endocrine neoplasms remain a great threat to human health. It is extremely important to make a clear diagnosis and timely treatment of endocrine tumors. Machine learning includes radiomics, which has long been utilized in clinical cancer research. Radiomics refers to the extraction of valuable information by analyzing a large amount of standard data with high-throughput medical images mainly including computed tomography, positron emission tomography, magnetic resonance imaging, and ultrasound. With the quantitative imaging analysis and model building, radiomics can reflect specific underlying characteristics of a disease that otherwise could not be evaluated visually. More and more promising results of radiomics in oncological practice have been seen in recent years. Radiomics may have the potential to supplement traditional imaging analysis and assist in providing precision medicine for patients. Radiomics had developed rapidly in endocrine neoplasms practice in the past decade. In this review, we would introduce the general workflow of radiomics and summarize the applications and developments of radiomics in endocrine neoplasms in recent years. The limitations of current radiomic research studies and future development directions would also be discussed.
Collapse
|
21
|
Musall BC, Abdelhafez AH, Adrada BE, Candelaria RP, Mohamed RM, Boge M, Le-Petross H, Arribas E, Lane DL, Spak DA, Leung JW, Hwang KP, Son JB, Elshafeey NA, Mahmoud HS, Wei P, Sun J, Zhang S, White JB, Ravenberg EE, Litton JK, Damodaran S, Thompson AM, Moulder SL, Yang WT, Pagel MD, Rauch GM, Ma J. Functional Tumor Volume by Fast Dynamic Contrast-Enhanced MRI for Predicting Neoadjuvant Systemic Therapy Response in Triple-Negative Breast Cancer. J Magn Reson Imaging 2021; 54:251-260. [PMID: 33586845 PMCID: PMC11830147 DOI: 10.1002/jmri.27557] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Dynamic contrast-enhanced (DCE) MRI is useful for diagnosis and assessment of treatment response in breast cancer. Fast DCE MRI offers a higher sampling rate of contrast enhancement curves in comparison to conventional DCE MRI, potentially characterizing tumor perfusion kinetics more accurately for measurement of functional tumor volume (FTV) as a predictor of treatment response. PURPOSE To investigate FTV by fast DCE MRI as a predictor of neoadjuvant systemic therapy (NAST) response in triple-negative breast cancer (TNBC). STUDY TYPE Prospective. POPULATION/SUBJECTS Sixty patients with biopsy-confirmed TNBC between December 2016 and September 2020. FIELD STRENGTH/SEQUENCE A 3.0 T/3D fast spoiled gradient echo-based DCE MRI ASSESSMENT: Patients underwent MRI at baseline and after four cycles (C4) of NAST, followed by definitive surgery. DCE subtraction images were analyzed in consensus by two breast radiologists with 5 (A.H.A.) and 2 (H.S.M.) years of experience. Tumor volumes (TV) were measured on early and late subtractions. Tumors were segmented on 1 and 2.5-minute early phases subtractions and FTV was determined using optimized signal enhancement thresholds. Interpolated enhancement curves from segmented voxels were used to determine optimal early phase timing. STATISTICAL TESTS Tumor volumes were compared between patients who had a pathologic complete response (pCR) and those who did not using the area under the receiver operating curve (AUC) and Mann-Whitney U test. RESULTS About 26 of 60 patients (43%) had pCR. FTV at 1 minute after injection at C4 provided the best discrimination between pCR and non-pCR, with AUC (95% confidence interval [CI]) = 0.85 (0.74,0.95) (P < 0.05). The 1-minute timing was optimal for FTV measurements at C4 and for the change between C4 and baseline. TV from the early phase at C4 also yielded a good AUC (95%CI) of 0.82 (0.71,0.93) (P < 0.05). DATA CONCLUSION FTV and TV measured at 1 minute after injection can predict response to NAST in TNBC. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY: 4.
Collapse
Affiliation(s)
- Benjamin C. Musall
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abeer H. Abdelhafez
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Beatriz E. Adrada
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rosalind P. Candelaria
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rania M.M. Mohamed
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Medine Boge
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Huong Le-Petross
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elsa Arribas
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Deanna L. Lane
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David A. Spak
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jessica W.T. Leung
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ken-Pin Hwang
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jong Bum Son
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nabil A. Elshafeey
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hagar S. Mahmoud
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jia Sun
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shu Zhang
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jason B. White
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elizabeth E. Ravenberg
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer K. Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Senthil Damodaran
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Stacy L. Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei T. Yang
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mark D. Pagel
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gaiane M. Rauch
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
22
|
Recent Radiomics Advancements in Breast Cancer: Lessons and Pitfalls for the Next Future. ACTA ACUST UNITED AC 2021; 28:2351-2372. [PMID: 34202321 PMCID: PMC8293249 DOI: 10.3390/curroncol28040217] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022]
Abstract
Radiomics is an emerging translational field of medicine based on the extraction of high-dimensional data from radiological images, with the purpose to reach reliable models to be applied into clinical practice for the purposes of diagnosis, prognosis and evaluation of disease response to treatment. We aim to provide the basic information on radiomics to radiologists and clinicians who are focused on breast cancer care, encouraging cooperation with scientists to mine data for a better application in clinical practice. We investigate the workflow and clinical application of radiomics in breast cancer care, as well as the outlook and challenges based on recent studies. Currently, radiomics has the potential ability to distinguish between benign and malignant breast lesions, to predict breast cancer’s molecular subtypes, the response to neoadjuvant chemotherapy and the lymph node metastases. Even though radiomics has been used in tumor diagnosis and prognosis, it is still in the research phase and some challenges need to be faced to obtain a clinical translation. In this review, we discuss the current limitations and promises of radiomics for improvement in further research.
Collapse
|
23
|
Comes MC, La Forgia D, Didonna V, Fanizzi A, Giotta F, Latorre A, Martinelli E, Mencattini A, Paradiso AV, Tamborra P, Terenzio A, Zito A, Lorusso V, Massafra R. Early Prediction of Breast Cancer Recurrence for Patients Treated with Neoadjuvant Chemotherapy: A Transfer Learning Approach on DCE-MRIs. Cancers (Basel) 2021; 13:2298. [PMID: 34064923 PMCID: PMC8151784 DOI: 10.3390/cancers13102298] [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: 03/01/2021] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer treatment planning benefits from an accurate early prediction of the treatment efficacy. The goal of this study is to give an early prediction of three-year Breast Cancer Recurrence (BCR) for patients who underwent neoadjuvant chemotherapy. We addressed the task from a new perspective based on transfer learning applied to pre-treatment and early-treatment DCE-MRI scans. Firstly, low-level features were automatically extracted from MR images using a pre-trained Convolutional Neural Network (CNN) architecture without human intervention. Subsequently, the prediction model was built with an optimal subset of CNN features and evaluated on two sets of patients from I-SPY1 TRIAL and BREAST-MRI-NACT-Pilot public databases: a fine-tuning dataset (70 not recurrent and 26 recurrent cases), which was primarily used to find the optimal subset of CNN features, and an independent test (45 not recurrent and 17 recurrent cases), whose patients had not been involved in the feature selection process. The best results were achieved when the optimal CNN features were augmented by four clinical variables (age, ER, PgR, HER2+), reaching an accuracy of 91.7% and 85.2%, a sensitivity of 80.8% and 84.6%, a specificity of 95.7% and 85.4%, and an AUC value of 0.93 and 0.83 on the fine-tuning dataset and the independent test, respectively. Finally, the CNN features extracted from pre-treatment and early-treatment exams were revealed to be strong predictors of BCR.
Collapse
Affiliation(s)
- Maria Colomba Comes
- Struttura Semplice Dipartimentale di Fisica Sanitaria, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (M.C.C.); (V.D.); (P.T.); (R.M.)
| | - Daniele La Forgia
- Struttura Semplice Dipartimentale di Radiologia Senologica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Vittorio Didonna
- Struttura Semplice Dipartimentale di Fisica Sanitaria, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (M.C.C.); (V.D.); (P.T.); (R.M.)
| | - Annarita Fanizzi
- Struttura Semplice Dipartimentale di Fisica Sanitaria, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (M.C.C.); (V.D.); (P.T.); (R.M.)
| | - Francesco Giotta
- Unità Operativa Complessa di Oncologia Medica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (F.G.); (A.L.); (V.L.)
| | - Agnese Latorre
- Unità Operativa Complessa di Oncologia Medica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (F.G.); (A.L.); (V.L.)
| | - Eugenio Martinelli
- Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (ICLOC), University of Rome Tor Vergata, 00133 Rome, Italy; (E.M.); (A.M.)
- Dipartimento di Ingegneria Elettronica, Università di Roma Tor Vergata, Via del Politecnico 1, 00133 Roma, Italy
| | - Arianna Mencattini
- Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (ICLOC), University of Rome Tor Vergata, 00133 Rome, Italy; (E.M.); (A.M.)
- Dipartimento di Ingegneria Elettronica, Università di Roma Tor Vergata, Via del Politecnico 1, 00133 Roma, Italy
| | - Angelo Virgilio Paradiso
- Oncologia Medica Sperimentale, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Pasquale Tamborra
- Struttura Semplice Dipartimentale di Fisica Sanitaria, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (M.C.C.); (V.D.); (P.T.); (R.M.)
| | - Antonella Terenzio
- Unità di Oncologia Medica, Università Campus Bio-Medico, 00128 Roma, Italy;
| | - Alfredo Zito
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Vito Lorusso
- Unità Operativa Complessa di Oncologia Medica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (F.G.); (A.L.); (V.L.)
| | - Raffaella Massafra
- Struttura Semplice Dipartimentale di Fisica Sanitaria, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (M.C.C.); (V.D.); (P.T.); (R.M.)
| |
Collapse
|
24
|
Xiong L, Chen H, Tang X, Chen B, Jiang X, Liu L, Feng Y, Liu L, Li L. Ultrasound-Based Radiomics Analysis for Predicting Disease-Free Survival of Invasive Breast Cancer. Front Oncol 2021; 11:621993. [PMID: 33996546 PMCID: PMC8117589 DOI: 10.3389/fonc.2021.621993] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/06/2021] [Indexed: 12/31/2022] Open
Abstract
Background Accurate prediction of recurrence is crucial for personalized treatment in breast cancer, and whether the radiomics features of ultrasound (US) could be used to predict recurrence of breast cancer is still uncertain. Here, we developed a radiomics signature based on preoperative US to predict disease-free survival (DFS) in patients with invasive breast cancer and assess its additional value to the clinicopathological predictors for individualized DFS prediction. Methods We identified 620 patients with invasive breast cancer and randomly divided them into the training (n = 372) and validation (n = 248) cohorts. A radiomics signature was constructed using least absolute shrinkage and selection operator (LASSO) Cox regression in the training cohort and validated in the validation cohort. Univariate and multivariate Cox proportional hazards model and Kaplan–Meier survival analysis were used to determine the association of the radiomics signature and clinicopathological variables with DFS. To evaluate the additional value of the radiomics signature for DFS prediction, a radiomics nomogram combining the radiomics signature and clinicopathological predictors was constructed and assessed in terms of discrimination, calibration, reclassification, and clinical usefulness. Results The radiomics signature was significantly associated with DFS, independent of the clinicopathological predictors. The radiomics nomogram performed better than the clinicopathological nomogram (C-index, 0.796 vs. 0.761) and provided better calibration and positive net reclassification improvement (0.147, P = 0.035) in the validation cohort. Decision curve analysis also demonstrated that the radiomics nomogram was clinically useful. Conclusion US radiomics signature is a potential imaging biomarker for risk stratification of DFS in invasive breast cancer, and US-based radiomics nomogram improved accuracy of DFS prediction.
Collapse
Affiliation(s)
- Lang Xiong
- Department of Medical Imaging, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Haolin Chen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Xiaofeng Tang
- Department of Ultrasound, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Biyun Chen
- Department of Medical Imaging, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xinhua Jiang
- Department of Medical Imaging, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Lizhi Liu
- Department of Medical Imaging, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Longzhong Liu
- Department of Ultrasound, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Li Li
- Department of Medical Imaging, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
25
|
Moyya PD, Asaithambi M. Radiomics- Quantitative Biomarker Analysis for Breast Cancer Diagnosis and Prediction: A Review. Curr Med Imaging 2021; 18:3-17. [PMID: 33655872 DOI: 10.2174/1573405617666210303102526] [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: 10/28/2020] [Revised: 01/06/2021] [Accepted: 01/14/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cancer of the breast has become a global problem for women's health. Though concerns regarding early detection and accurate diagnosis were raised, an effort is required for precision medicine as well as personalized treatment. In the past years, the area of medicinal imaging has seen an unprecedented growth that leads to an advancement of radiomics, which provides countless quantitative biomarkers extracted from modern diagnostic images, including a detailed tumor characterization of breast malignancy. DISCUSSION In this research, we presented the methodology and implementation of radiomics, together with its future trends and challenges by the basis of published papers. Radiomics could distinguish between malignant from benign tumors, predict prognostic factors, molecular subtypes of breast carcinoma, treatment response to neoadjuvant chemotherapy (NAC), and recurrence survival. The incorporation of quantitative knowledge with clinical, histopathological and genomic information will enable physicians to afford customized care of treatment for patients with breast cancer. CONCLUSION Our research was intended to help physicians and radiologists learn fundamental knowledge about radiomics and also to work collaboratively with researchers to explore evidence for further usage in clinical practice.
Collapse
Affiliation(s)
- Priscilla Dinkar Moyya
- School of Electronics Engineering, Vellore Institute of Technology, Vellore, Tamil Nadu-632014. India
| | - Mythili Asaithambi
- School of Electronics Engineering, Vellore Institute of Technology, Vellore, Tamil Nadu-632014. India
| |
Collapse
|
26
|
Liu T, Song S, Wang X, Hao J. Small-molecule inhibitors of breast cancer-related targets: Potential therapeutic agents for breast cancer. Eur J Med Chem 2021; 210:112954. [PMID: 33158576 DOI: 10.1016/j.ejmech.2020.112954] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022]
Abstract
Despite dramatic advances in cancer research and therapy, breast cancer remains a tricky health problem and represents a top biomedical research priority. Nowadays, breast cancer is still the leading cause of malignancy-related deaths in women, and incidence and mortality rates of it are expected to increase significantly the next years. Currently more and more researchers are interested in the study of breast cancer by its arising in young women. The common treatment options of breast cancer are chemotherapy, immunotherapy, hormone therapy, surgery, and radiotherapy. Most of them require chemical agents, such as PARP inhibitors, CDK4/6 inhibitors, and HER2 inhibitors. Recent studies suggest that some targets or pathways, including BRD4, PLK1, PD-L1, HDAC, and PI3K/AKT/mTOR, are tightly related to the occurrence and development of breast cancer. This article reviews the interplay between these targets and breast cancer and summarizes the progress of current research on small molecule inhibitors of these anti-breast cancer targets. The review aims to provide structural and theoretical basis for designing novel anti-breast cancer agents.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China.
| | - Shubin Song
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Xu Wang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, United States
| | - Jifu Hao
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
| |
Collapse
|
27
|
Yixin HMD, Fei LMD, Jianhua ZMD. Current Status and Advances in Imaging Evaluation of Neoadjuvant Chemotherapy of Breast Cancer. ADVANCED ULTRASOUND IN DIAGNOSIS AND THERAPY 2021. [DOI: 10.37015/audt.2021.190036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
28
|
|
29
|
Giger ML. AI/Machine Learning in Medical Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00052-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
30
|
Exploring MRI based radiomics analysis of intratumoral spatial heterogeneity in locally advanced nasopharyngeal carcinoma treated with intensity modulated radiotherapy. PLoS One 2020; 15:e0240043. [PMID: 33017440 PMCID: PMC7535039 DOI: 10.1371/journal.pone.0240043] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 09/18/2020] [Indexed: 01/28/2023] Open
Abstract
Background We hypothesized that spatial heterogeneity exists between recurrent and non-recurrent regions within a tumor. The aim of this study was to determine if there is a difference between radiomics features derived from recurrent versus non recurrent regions within the tumor based on pre-treatment MRI. Methods A total of 14 T4NxM0 NPC patients with histologically proven “in field” recurrence in the post nasal space following curative intent IMRT were included in this study. Pretreatment MRI were co-registered with MRI at the time of recurrence for the delineation of gross tumor volume at diagnosis(GTV) and at recurrence(GTVr). A total of 7 histogram features and 40 texture features were computed from the recurrent(GTVr) and non-recurrent region(GTV-GTVr). Paired t-tests and Wilcoxon signed-rank tests were carried out on the 47 quantified radiomics features. Results A total of 7 features were significantly different between recurrent and non-recurrent regions. Other than the variance from intensity-based histogram, the remaining six significant features were either from the gray-level size zone matrix (GLSZM) or the neighbourhood gray-tone difference matrix (NGTDM). Conclusions The radiomic features extracted from pre-treatment MRI can potentially reflect the difference between recurrent and non-recurrent regions within a tumor and has a potential role in pre-treatment identification of intra-tumoral radio-resistance for selective dose escalation.
Collapse
|
31
|
Qiu X, Jiang Y, Zhao Q, Yan C, Huang M, Jiang T. Could Ultrasound-Based Radiomics Noninvasively Predict Axillary Lymph Node Metastasis in Breast Cancer? JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2020; 39:1897-1905. [PMID: 32329142 PMCID: PMC7540260 DOI: 10.1002/jum.15294] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVES This work aimed to investigate whether quantitative radiomics imaging features extracted from ultrasound (US) can noninvasively predict breast cancer (BC) metastasis to axillary lymph nodes (ALNs). METHODS Presurgical B-mode US data of 196 patients with BC were retrospectively studied. The cases were divided into the training and validation cohorts (n = 141 versus 55). The elastic net regression technique was used for selecting features and building a signature in the training cohort. A linear combination of the selected features weighted by their respective coefficients produced a radiomics signature for each individual. A radiomics nomogram was established based on the radiomics signature and US-reported ALN status. In a receiver operating characteristic curve analysis, areas under the curves (AUCs) were determined for assessing the accuracy of the prediction model in predicting ALN metastasis in both cohorts. The clinical value was assessed by a decision curve analysis. RESULTS In all, 843 radiomics features per case were obtained from expert-delineated lesions on US imaging in this study. Through radiomics feature selection, 21 features were selected to constitute the radiomics signature for predicting ALN metastasis. Area under the curve values of 0.778 and 0.725 were obtained in the training and validation cohorts, respectively, indicating moderate predictive ability. The radiomics nomogram comprising the radiomics signature and US-reported ALN status showed the best performance for ALN detection in the training cohort (AUC, 0.816) but moderate performance in the validation cohort (AUC, 0.759). The decision curve showed that both the radiomics signature and nomogram displayed good clinical utility. CONCLUSIONS This pilot radiomics study provided a noninvasive method for predicting presurgical ALN metastasis status in BC.
Collapse
Affiliation(s)
- Xiaoying Qiu
- Departments of UltrasonographyFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| | - Yongluo Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Qiyu Zhao
- Departments of UltrasonographyFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
- Hepatobiliary and Pancreatic SurgeryFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| | - Chunhong Yan
- Departments of UltrasonographyFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| | - Min Huang
- Departments of UltrasonographyFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| | - Tian'an Jiang
- Departments of UltrasonographyFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
- Hepatobiliary and Pancreatic SurgeryFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| |
Collapse
|
32
|
Sutton EJ, Onishi N, Fehr DA, Dashevsky BZ, Sadinski M, Pinker K, Martinez DF, Brogi E, Braunstein L, Razavi P, El-Tamer M, Sacchini V, Deasy JO, Morris EA, Veeraraghavan H. A machine learning model that classifies breast cancer pathologic complete response on MRI post-neoadjuvant chemotherapy. Breast Cancer Res 2020; 22:57. [PMID: 32466777 PMCID: PMC7254668 DOI: 10.1186/s13058-020-01291-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 05/10/2020] [Indexed: 12/13/2022] Open
Abstract
Background For breast cancer patients undergoing neoadjuvant chemotherapy (NAC), pathologic complete response (pCR; no invasive or in situ) cannot be assessed non-invasively so all patients undergo surgery. The aim of our study was to develop and validate a radiomics classifier that classifies breast cancer pCR post-NAC on MRI prior to surgery. Methods This retrospective study included women treated with NAC for breast cancer from 2014 to 2016 with (1) pre- and post-NAC breast MRI and (2) post-NAC surgical pathology report assessing response. Automated radiomics analysis of pre- and post-NAC breast MRI involved image segmentation, radiomics feature extraction, feature pre-filtering, and classifier building through recursive feature elimination random forest (RFE-RF) machine learning. The RFE-RF classifier was trained with nested five-fold cross-validation using (a) radiomics only (model 1) and (b) radiomics and molecular subtype (model 2). Class imbalance was addressed using the synthetic minority oversampling technique. Results Two hundred seventy-three women with 278 invasive breast cancers were included; the training set consisted of 222 cancers (61 pCR, 161 no-pCR; mean age 51.8 years, SD 11.8), and the independent test set consisted of 56 cancers (13 pCR, 43 no-pCR; mean age 51.3 years, SD 11.8). There was no significant difference in pCR or molecular subtype between the training and test sets. Model 1 achieved a cross-validation AUROC of 0.72 (95% CI 0.64, 0.79) and a similarly accurate (P = 0.1) AUROC of 0.83 (95% CI 0.71, 0.94) in both the training and test sets. Model 2 achieved a cross-validation AUROC of 0.80 (95% CI 0.72, 0.87) and a similar (P = 0.9) AUROC of 0.78 (95% CI 0.62, 0.94) in both the training and test sets. Conclusions This study validated a radiomics classifier combining radiomics with molecular subtypes that accurately classifies pCR on MRI post-NAC.
Collapse
Affiliation(s)
- Elizabeth J Sutton
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Natsuko Onishi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Duc A Fehr
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brittany Z Dashevsky
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meredith Sadinski
- 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
| | - Danny F Martinez
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Edi Brogi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lior Braunstein
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mahmoud El-Tamer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Virgilio Sacchini
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth A Morris
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Harini Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
33
|
Trivizakis E, Papadakis GZ, Souglakos I, Papanikolaou N, Koumakis L, Spandidos DA, Tsatsakis A, Karantanas AH, Marias K. Artificial intelligence radiogenomics for advancing precision and effectiveness in oncologic care (Review). Int J Oncol 2020; 57:43-53. [PMID: 32467997 PMCID: PMC7252460 DOI: 10.3892/ijo.2020.5063] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/05/2020] [Indexed: 12/11/2022] Open
Abstract
The new era of artificial intelligence (AI) has introduced revolutionary data-driven analysis paradigms that have led to significant advancements in information processing techniques in the context of clinical decision-support systems. These advances have created unprecedented momentum in computational medical imaging applications and have given rise to new precision medicine research areas. Radiogenomics is a novel research field focusing on establishing associations between radiological features and genomic or molecular expression in order to shed light on the underlying disease mechanisms and enhance diagnostic procedures towards personalized medicine. The aim of the current review was to elucidate recent advances in radiogenomics research, focusing on deep learning with emphasis on radiology and oncology applications. The main deep learning radiogenomics architectures, together with the clinical questions addressed, and the achieved genetic or molecular correlations are presented, while a performance comparison of the proposed methodologies is conducted. Finally, current limitations, potentially understudied topics and future research directions are discussed.
Collapse
Affiliation(s)
- Eleftherios Trivizakis
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece
| | - Georgios Z Papadakis
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece
| | - Ioannis Souglakos
- Laboratory of Translational Oncology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Nikolaos Papanikolaou
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece
| | - Lefteris Koumakis
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Aristidis Tsatsakis
- Laboratory of Forensic Sciences and Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Apostolos H Karantanas
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece
| | - Kostas Marias
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece
| |
Collapse
|
34
|
Conti A, Duggento A, Indovina I, Guerrisi M, Toschi N. Radiomics in breast cancer classification and prediction. Semin Cancer Biol 2020; 72:238-250. [PMID: 32371013 DOI: 10.1016/j.semcancer.2020.04.002] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022]
Abstract
Breast Cancer (BC) is the common form of cancer in women. Its diagnosis and screening are usually performed through different imaging modalities such as mammography, magnetic resonance imaging and ultrasound. However, mammography and ultrasound-imaging techniques have limited sensitivity and specificity both in identifying lesions and in differentiating malign from benign lesions, especially in presence of dense breast parenchyma. Due to the higher resolution of magnetic resonance images, MRI represents the method with the higher specificity and sensitivity among all the available tools, in both lesions' identification and diagnosis. However, especially for diagnosis, even MRI has limitations that are only partially solved if combined with mammography. Unfortunately, due to the limits of all these imaging tools, in order to have a certain diagnosis, patients often receive painful and costly bioptics procedures. In this context, several computational approaches have been developed to increase sensitivity, while maintaining the same specificity, in BC diagnosis and screening. Amongst these, radiomics has been increasingly gaining ground in oncology to improve cancer diagnosis, prognosis and treatment. Radiomics derives multiple quantitative features from single or multiple medical imaging modalities, highlighting image traits which are not visible to the naked eye and hence significantly augmenting the discriminatory and predictive potential of medical imaging. This review article aims to summarize the state of the art in radiomics-based BC research. The dominating evidence extracted from the literature points towards a high potential of radiomics in disentangling malignant from benign breast lesions, classifying BC types and grades and also in predicting treatment response and recurrence risk. In the era of personalized medicine, radiomics has the potential to improve diagnosis, prognosis, prediction, monitoring, image-based intervention, and assessment of therapeutic response in BC.
Collapse
Affiliation(s)
- Allegra Conti
- Laboratory of Neuromotor Physiology, IRCCS Santa Lucia Foundation, Via Ardeatina, 306, 00179, Rome, Italy; Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| | - Andrea Duggento
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| | - Iole Indovina
- Laboratory of Neuromotor Physiology, IRCCS Santa Lucia Foundation, Via Ardeatina, 306, 00179, Rome, Italy; Department of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Maria Guerrisi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Boston, MA, United States.
| |
Collapse
|
35
|
Pesapane F, Suter MB, Rotili A, Penco S, Nigro O, Cremonesi M, Bellomi M, Jereczek-Fossa BA, Pinotti G, Cassano E. Will traditional biopsy be substituted by radiomics and liquid biopsy for breast cancer diagnosis and characterisation? Med Oncol 2020; 37:29. [PMID: 32180032 DOI: 10.1007/s12032-020-01353-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/26/2020] [Indexed: 02/06/2023]
Abstract
The diagnosis of breast cancer currently relies on radiological and clinical evaluation, confirmed by histopathological examination. However, such approach has some limitations as the suboptimal sensitivity, the long turnaround time for recall tests, the invasiveness of the procedure and the risk that some features of target lesions may remain undetected, making re-biopsy a necessity. Recent technological advances in the field of artificial intelligence hold promise in addressing such medical challenges not only in cancer diagnosis, but also in treatment assessment, and monitoring of disease progression. In the perspective of a truly personalised medicine, based on the early diagnosis and individually tailored treatments, two new technologies, namely radiomics and liquid biopsy, are rising as means to obtain information from diagnosis to molecular profiling and response assessment, without the need of a biopsied tissue sample. Radiomics works through the extraction of quantitative peculiar features of cancer from radiological data, while liquid biopsy gets the whole of the malignancy's biology from something as easy as a blood sample. Both techniques hopefully will identify diagnostic and prognostic information of breast cancer potentially reducing the need for invasive (and often difficult to perform) biopsies and favouring an approach that is as personalised as possible for each patient. Nevertheless, such techniques will not substitute tissue biopsy in the near future, and even in further times they will require the aid of other parameters to be correctly interpreted and acted upon.
Collapse
Affiliation(s)
- Filippo Pesapane
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy.
| | | | - Anna Rotili
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Silvia Penco
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Olga Nigro
- Medical Oncology, ASST Sette Laghi, Viale Borri 57, 21100, Varese, VA, Italy
| | - Marta Cremonesi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Massimo Bellomi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Radiology, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Graziella Pinotti
- Medical Oncology, ASST Sette Laghi, Viale Borri 57, 21100, Varese, VA, Italy
| | - Enrico Cassano
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| |
Collapse
|
36
|
El Naqa I, Haider MA, Giger ML, Ten Haken RK. Artificial Intelligence: reshaping the practice of radiological sciences in the 21st century. Br J Radiol 2020; 93:20190855. [PMID: 31965813 PMCID: PMC7055429 DOI: 10.1259/bjr.20190855] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Advances in computing hardware and software platforms have led to the recent resurgence in artificial intelligence (AI) touching almost every aspect of our daily lives by its capability for automating complex tasks or providing superior predictive analytics. AI applications are currently spanning many diverse fields from economics to entertainment, to manufacturing, as well as medicine. Since modern AI's inception decades ago, practitioners in radiological sciences have been pioneering its development and implementation in medicine, particularly in areas related to diagnostic imaging and therapy. In this anniversary article, we embark on a journey to reflect on the learned lessons from past AI's chequered history. We further summarize the current status of AI in radiological sciences, highlighting, with examples, its impressive achievements and effect on re-shaping the practice of medical imaging and radiotherapy in the areas of computer-aided detection, diagnosis, prognosis, and decision support. Moving beyond the commercial hype of AI into reality, we discuss the current challenges to overcome, for AI to achieve its promised hope of providing better precision healthcare for each patient while reducing cost burden on their families and the society at large.
Collapse
Affiliation(s)
- Issam El Naqa
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Masoom A Haider
- Department of Medical Imaging and Lunenfeld-Tanenbaum Research Institute, University of Toronto, Toronto, ON, Canada
| | | | - Randall K Ten Haken
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
37
|
Ye DM, Wang HT, Yu T. The Application of Radiomics in Breast MRI: A Review. Technol Cancer Res Treat 2020; 19:1533033820916191. [PMID: 32347167 PMCID: PMC7225803 DOI: 10.1177/1533033820916191] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/21/2020] [Accepted: 02/27/2020] [Indexed: 12/13/2022] Open
Abstract
Breast cancer has been a worldwide burden of women's health. Although concerns have been raised for early diagnosis and timely treatment, the efforts are still needed for precision medicine and individualized treatment. Radiomics is a new technology with immense potential to obtain mineable data to provide rich information about the diagnosis and prognosis of breast cancer. In our study, we introduced the workflow and application of radiomics as well as its outlook and challenges based on published studies. Radiomics has the potential ability to differentiate between malignant and benign breast lesions, predict axillary lymph node status, molecular subtypes of breast cancer, tumor response to chemotherapy, and survival outcomes. Our study aimed to help clinicians and radiologists to know the basic information of radiomics and encourage cooperation with scientists to mine data for better application in clinical practice.
Collapse
Affiliation(s)
- Dong-Man Ye
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People’s Republic of China
| | - Hao-Tian Wang
- Dalian Medical University, The First Clinical College, Dalian, Liaoning Province, People’s Republic of China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People’s Republic of China
| |
Collapse
|
38
|
Greenwalt I, Zaza N, Das S, Li BD. Precision Medicine and Targeted Therapies in Breast Cancer. Surg Oncol Clin N Am 2020; 29:51-62. [DOI: 10.1016/j.soc.2019.08.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
39
|
Davatzikos C, Sotiras A, Fan Y, Habes M, Erus G, Rathore S, Bakas S, Chitalia R, Gastounioti A, Kontos D. Precision diagnostics based on machine learning-derived imaging signatures. Magn Reson Imaging 2019; 64:49-61. [PMID: 31071473 PMCID: PMC6832825 DOI: 10.1016/j.mri.2019.04.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/24/2019] [Accepted: 04/29/2019] [Indexed: 01/08/2023]
Abstract
The complexity of modern multi-parametric MRI has increasingly challenged conventional interpretations of such images. Machine learning has emerged as a powerful approach to integrating diverse and complex imaging data into signatures of diagnostic and predictive value. It has also allowed us to progress from group comparisons to imaging biomarkers that offer value on an individual basis. We review several directions of research around this topic, emphasizing the use of machine learning in personalized predictions of clinical outcome, in breaking down broad umbrella diagnostic categories into more detailed and precise subtypes, and in non-invasively estimating cancer molecular characteristics. These methods and studies contribute to the field of precision medicine, by introducing more specific diagnostic and predictive biomarkers of clinical outcome, therefore pointing to better matching of treatments to patients.
Collapse
Affiliation(s)
- Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, United States of America.
| | - Aristeidis Sotiras
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, United States of America
| | - Yong Fan
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, United States of America
| | - Mohamad Habes
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, United States of America
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, United States of America
| | - Saima Rathore
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, United States of America
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, United States of America
| | - Rhea Chitalia
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, United States of America
| | - Aimilia Gastounioti
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, United States of America
| | - Despina Kontos
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, United States of America
| |
Collapse
|
40
|
Greenwood HI, Wilmes LJ, Kelil T, Joe BN. Role of Breast MRI in the Evaluation and Detection of DCIS: Opportunities and Challenges. J Magn Reson Imaging 2019; 52:697-709. [PMID: 31746088 DOI: 10.1002/jmri.26985] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/29/2022] Open
Abstract
Historically, breast magnetic resonance imaging (MRI) was not considered an effective modality in the evaluation of ductal carcinoma in situ (DCIS). Over the past decade this has changed, with studies demonstrating that MRI is the most sensitive imaging tool for detection of all grades of DCIS. It has been suggested that not only is breast MRI the most sensitive imaging tool for detection but it may also detect the most clinically relevant DCIS lesions. The role and outcomes of MRI in the preoperative setting for patients with DCIS remains controversial; however, several studies have shown benefit in the preoperative evaluation of extent of disease as well as predicting an underlying invasive component. The most common presentation of DCIS on MRI is nonmass enhancement (NME) in a linear or segmental distribution pattern. Maximizing breast MRI spatial resolution is therefore beneficial, given the frequent presentation of DCIS as NME on MRI. Emerging MRI techniques, such as diffusion-weighted imaging (DWI), have shown promising potential to discriminate DCIS from benign and invasive lesions. Future opportunities including advanced imaging visual techniques, radiomics/radiogenomics, and machine learning / artificial intelligence may also be applicable to the detection and treatment of DCIS. Level of Evidence: 3 Technical Efficacy Stage: 3 J. Magn. Reson. Imaging 2019. J. Magn. Reson. Imaging 2020;52:697-709.
Collapse
Affiliation(s)
- Heather I Greenwood
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Lisa J Wilmes
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Tatiana Kelil
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Bonnie N Joe
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| |
Collapse
|
41
|
Impact of Machine Learning With Multiparametric Magnetic Resonance Imaging of the Breast for Early Prediction of Response to Neoadjuvant Chemotherapy and Survival Outcomes in Breast Cancer Patients. Invest Radiol 2019; 54:110-117. [PMID: 30358693 DOI: 10.1097/rli.0000000000000518] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE The aim of this study was to assess the potential of machine learning with multiparametric magnetic resonance imaging (mpMRI) for the early prediction of pathological complete response (pCR) to neoadjuvant chemotherapy (NAC) and of survival outcomes in breast cancer patients. MATERIALS AND METHODS This institutional review board-approved prospective study included 38 women (median age, 46.5 years; range, 25-70 years) with breast cancer who were scheduled for NAC and underwent mpMRI of the breast at 3 T with dynamic contrast-enhanced (DCE), diffusion-weighted imaging (DWI), and T2-weighted imaging before and after 2 cycles of NAC. For each lesion, 23 features were extracted: qualitative T2-weighted and DCE-MRI features according to BI-RADS (Breast Imaging Reporting and Data System), quantitative pharmacokinetic DCE features (mean plasma flow, volume distribution, mean transit time), and DWI apparent diffusion coefficient (ADC) values. To apply machine learning to mpMRI, 8 classifiers including linear support vector machine, linear discriminant analysis, logistic regression, random forests, stochastic gradient descent, decision tree, adaptive boosting, and extreme gradient boosting (XGBoost) were used to rank the features. Histopathologic residual cancer burden (RCB) class (with RCB 0 being a pCR), recurrence-free survival (RFS), and disease-specific survival (DSS) were used as the standards of reference. Classification accuracy with area under the receiving operating characteristic curve (AUC) was assessed using all the extracted qualitative and quantitative features for pCR as defined by RCB class, RFS, and DSS using recursive feature elimination. To overcome overfitting, 4-fold cross-validation was used. RESULTS Machine learning with mpMRI achieved stable performance as shown by mean classification accuracies for the prediction of RCB class (AUC, 0.86) and DSS (AUC, 0.92) based on XGBoost and the prediction of RFS (AUC, 0.83) with logistic regression. The XGBoost classifier achieved the most stable performance with high accuracies compared with other classifiers. The most relevant features for the prediction of RCB class were as follows: changes in lesion size, complete pattern of shrinkage, and mean transit time on DCE-MRI; minimum ADC on DWI; and peritumoral edema on T2-weighted imaging. The most relevant features for prediction of RFS were as follows: volume distribution, mean plasma flow, and mean transit time; DCE-MRI lesion size; minimum, maximum, and mean ADC with DWI. The most relevant features for prediction of DSS were as follows: lesion size, volume distribution, and mean plasma flow on DCE-MRI, and maximum ADC with DWI. CONCLUSIONS Machine learning with mpMRI of the breast enables early prediction of pCR to NAC as well as survival outcomes in breast cancer patients with high accuracy and thus may provide valuable predictive information to guide treatment decisions.
Collapse
|
42
|
Sheth D, Giger ML. Artificial intelligence in the interpretation of breast cancer on MRI. J Magn Reson Imaging 2019; 51:1310-1324. [PMID: 31343790 DOI: 10.1002/jmri.26878] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022] Open
Abstract
Advances in both imaging and computers have led to the rise in the potential use of artificial intelligence (AI) in various tasks in breast imaging, going beyond the current use in computer-aided detection to include diagnosis, prognosis, response to therapy, and risk assessment. The automated capabilities of AI offer the potential to enhance the diagnostic expertise of clinicians, including accurate demarcation of tumor volume, extraction of characteristic cancer phenotypes, translation of tumoral phenotype features to clinical genotype implications, and risk prediction. The combination of image-specific findings with the underlying genomic, pathologic, and clinical features is becoming of increasing value in breast cancer. The concurrent emergence of newer imaging techniques has provided radiologists with greater diagnostic tools and image datasets to analyze and interpret. Integrating an AI-based workflow within breast imaging enables the integration of multiple data streams into powerful multidisciplinary applications that may lead the path to personalized patient-specific medicine. In this article we describe the goals of AI in breast cancer imaging, in particular MRI, and review the literature as it relates to the current application, potential, and limitations in breast cancer. Level of Evidence: 3 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2020;51:1310-1324.
Collapse
Affiliation(s)
- Deepa Sheth
- Department of Radiology, University of Chicago, Chicago, Illinois, USA
| | - Maryellen L Giger
- Department of Radiology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
43
|
Reig B, Heacock L, Geras KJ, Moy L. Machine learning in breast MRI. J Magn Reson Imaging 2019; 52:998-1018. [PMID: 31276247 DOI: 10.1002/jmri.26852] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 12/13/2022] Open
Abstract
Machine-learning techniques have led to remarkable advances in data extraction and analysis of medical imaging. Applications of machine learning to breast MRI continue to expand rapidly as increasingly accurate 3D breast and lesion segmentation allows the combination of radiologist-level interpretation (eg, BI-RADS lexicon), data from advanced multiparametric imaging techniques, and patient-level data such as genetic risk markers. Advances in breast MRI feature extraction have led to rapid dataset analysis, which offers promise in large pooled multiinstitutional data analysis. The object of this review is to provide an overview of machine-learning and deep-learning techniques for breast MRI, including supervised and unsupervised methods, anatomic breast segmentation, and lesion segmentation. Finally, it explores the role of machine learning, current limitations, and future applications to texture analysis, radiomics, and radiogenomics. Level of Evidence: 3 Technical Efficacy Stage: 2 J. Magn. Reson. Imaging 2019. J. Magn. Reson. Imaging 2020;52:998-1018.
Collapse
Affiliation(s)
- Beatriu Reig
- The Department of Radiology, New York University School of Medicine, New York, New York, USA
| | - Laura Heacock
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York, USA
| | - Krzysztof J Geras
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York, USA
| | - Linda Moy
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York, USA.,Center for Advanced Imaging Innovation and Research (CAI2 R), New York University School of Medicine, New York, New York, USA
| |
Collapse
|
44
|
Drukker K, Edwards A, Doyle C, Papaioannou J, Kulkarni K, Giger ML. Breast MRI radiomics for the pretreatment prediction of response to neoadjuvant chemotherapy in node-positive breast cancer patients. J Med Imaging (Bellingham) 2019; 6:034502. [PMID: 31592438 PMCID: PMC6768440 DOI: 10.1117/1.jmi.6.3.034502] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/09/2019] [Indexed: 12/16/2022] Open
Abstract
The purpose of this study was to evaluate breast MRI radiomics in predicting, prior to any treatment, the response to neoadjuvant chemotherapy (NAC) in patients with invasive lymph node (LN)-positive breast cancer for two tasks: (1) prediction of pathologic complete response and (2) prediction of post-NAC LN status. Our study included 158 patients, with 19 showing post-NAC complete pathologic response (pathologic TNM stage T0,N0,MX) and 139 showing incomplete response. Forty-two patients were post-NAC LN-negative, and 116 were post-NAC LN-positive. We further analyzed prediction of response by hormone receptor subtype of the primary cancer (77 hormone receptor-positive, 39 HER2-enriched, 38 triple negative, and 4 cancers with unknown receptor status). Only pre-NAC MRIs underwent computer analysis, initialized by an expert breast radiologist indicating index cancers and metastatic axillary sentinel LNs on DCE-MRI images. Forty-nine computer-extracted radiomics features were obtained, both for the primary cancers and for the metastatic sentinel LNs. Since the dataset contained MRIs acquired at 1.5 T and at 3.0 T, we eliminated features affected by magnet strength using the Mann-Whitney U-test with the null-hypothesis that 1.5 T and 3.0 T samples were selected from populations having the same distribution. Bootstrapping and ROC analysis were used to assess performance of individual features in the two classification tasks. Eighteen features appeared unaffected by magnet strength. Pre-NAC tumor features generally appeared uninformative in predicting response to therapy. In contrast, some pre-NAC LN features were able to predict response: two pre-NAC LN features were able to predict pathologic complete response (area under the ROC curve (AUC) up to 0.82 [0.70; 0.88]), and another two were able to predict post-NAC LN-status (AUC up to 0.72 [0.62; 0.77]), respectively. In the analysis by a hormone receptor subtype, several potentially useful features were identified for predicting response to therapy in the hormone receptor-positive and HER2-enriched cancers.
Collapse
Affiliation(s)
- Karen Drukker
- University of Chicago, Department of Radiology, Chicago, Illinois, United States
| | - Alexandra Edwards
- University of Chicago, Department of Radiology, Chicago, Illinois, United States
| | - Christopher Doyle
- University of Chicago, Department of Radiology, Chicago, Illinois, United States
| | - John Papaioannou
- University of Chicago, Department of Radiology, Chicago, Illinois, United States
| | - Kirti Kulkarni
- University of Chicago, Department of Radiology, Chicago, Illinois, United States
| | - Maryellen L. Giger
- University of Chicago, Department of Radiology, Chicago, Illinois, United States
| |
Collapse
|
45
|
Bi WL, Hosny A, Schabath MB, Giger ML, Birkbak NJ, Mehrtash A, Allison T, Arnaout O, Abbosh C, Dunn IF, Mak RH, Tamimi RM, Tempany CM, Swanton C, Hoffmann U, Schwartz LH, Gillies RJ, Huang RY, Aerts HJWL. Artificial intelligence in cancer imaging: Clinical challenges and applications. CA Cancer J Clin 2019; 69:127-157. [PMID: 30720861 PMCID: PMC6403009 DOI: 10.3322/caac.21552] [Citation(s) in RCA: 770] [Impact Index Per Article: 128.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Judgement, as one of the core tenets of medicine, relies upon the integration of multilayered data with nuanced decision making. Cancer offers a unique context for medical decisions given not only its variegated forms with evolution of disease but also the need to take into account the individual condition of patients, their ability to receive treatment, and their responses to treatment. Challenges remain in the accurate detection, characterization, and monitoring of cancers despite improved technologies. Radiographic assessment of disease most commonly relies upon visual evaluations, the interpretations of which may be augmented by advanced computational analyses. In particular, artificial intelligence (AI) promises to make great strides in the qualitative interpretation of cancer imaging by expert clinicians, including volumetric delineation of tumors over time, extrapolation of the tumor genotype and biological course from its radiographic phenotype, prediction of clinical outcome, and assessment of the impact of disease and treatment on adjacent organs. AI may automate processes in the initial interpretation of images and shift the clinical workflow of radiographic detection, management decisions on whether or not to administer an intervention, and subsequent observation to a yet to be envisioned paradigm. Here, the authors review the current state of AI as applied to medical imaging of cancer and describe advances in 4 tumor types (lung, brain, breast, and prostate) to illustrate how common clinical problems are being addressed. Although most studies evaluating AI applications in oncology to date have not been vigorously validated for reproducibility and generalizability, the results do highlight increasingly concerted efforts in pushing AI technology to clinical use and to impact future directions in cancer care.
Collapse
Affiliation(s)
- Wenya Linda Bi
- Assistant Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Ahmed Hosny
- Research Scientist, Department of Radiation Oncology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Matthew B. Schabath
- Associate Member, Department of Cancer EpidemiologyH. Lee Moffitt Cancer Center and Research InstituteTampaFL
| | - Maryellen L. Giger
- Professor of Radiology, Department of RadiologyUniversity of ChicagoChicagoIL
| | - Nicolai J. Birkbak
- Research Associate, The Francis Crick InstituteLondonUnited Kingdom
- Research Associate, University College London Cancer InstituteLondonUnited Kingdom
| | - Alireza Mehrtash
- Research Assistant, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
- Research Assistant, Department of Electrical and Computer EngineeringUniversity of British ColumbiaVancouverBCCanada
| | - Tavis Allison
- Research Assistant, Department of RadiologyColumbia University College of Physicians and SurgeonsNew YorkNY
- Research Assistant, Department of RadiologyNew York Presbyterian HospitalNew YorkNY
| | - Omar Arnaout
- Assistant Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Christopher Abbosh
- Research Fellow, The Francis Crick InstituteLondonUnited Kingdom
- Research Fellow, University College London Cancer InstituteLondonUnited Kingdom
| | - Ian F. Dunn
- Associate Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Raymond H. Mak
- Associate Professor, Department of Radiation Oncology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Rulla M. Tamimi
- Associate Professor, Department of MedicineBrigham and Women’s Hospital, Dana‐Farber Cancer Institute, Harvard Medical SchoolBostonMA
| | - Clare M. Tempany
- Professor of Radiology, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Charles Swanton
- Professor, The Francis Crick InstituteLondonUnited Kingdom
- Professor, University College London Cancer InstituteLondonUnited Kingdom
| | - Udo Hoffmann
- Professor of Radiology, Department of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Lawrence H. Schwartz
- Professor of Radiology, Department of RadiologyColumbia University College of Physicians and SurgeonsNew YorkNY
- Chair, Department of RadiologyNew York Presbyterian HospitalNew YorkNY
| | - Robert J. Gillies
- Professor of Radiology, Department of Cancer PhysiologyH. Lee Moffitt Cancer Center and Research InstituteTampaFL
| | - Raymond Y. Huang
- Assistant Professor, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Hugo J. W. L. Aerts
- Associate Professor, Departments of Radiation Oncology and Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
- Professor in AI in Medicine, Radiology and Nuclear Medicine, GROWMaastricht University Medical Centre (MUMC+)MaastrichtThe Netherlands
| |
Collapse
|
46
|
Crivelli P, Ledda RE, Parascandolo N, Fara A, Soro D, Conti M. A New Challenge for Radiologists: Radiomics in Breast Cancer. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6120703. [PMID: 30402486 PMCID: PMC6196984 DOI: 10.1155/2018/6120703] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 08/24/2018] [Accepted: 09/09/2018] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Over the last decade, the field of medical imaging experienced an exponential growth, leading to the development of radiomics, with which innumerable quantitative features are obtained from digital medical images, providing a comprehensive characterization of the tumor. This review aims to assess the role of this emerging diagnostic tool in breast cancer, focusing on the ability of radiomics to predict malignancy, response to neoadjuvant chemotherapy, prognostic factors, molecular subtypes, and risk of recurrence. EVIDENCE ACQUISITION A literature search on PubMed and on Cochrane database websites to retrieve English-written systematic reviews, review articles, meta-analyses, and randomized clinical trials published from August 2013 up to July 2018 was carried out. RESULTS Twenty papers (19 retrospective and 1 prospective studies) conducted with different conventional imaging modalities were included. DISCUSSION The integration of quantitative information with clinical, histological, and genomic data could enable clinicians to provide personalized treatments for breast cancer patients. Current limitations of a routinely application of radiomics are represented by the limited knowledge of its basics concepts among radiologists and by the lack of efficient and standardized systems of feature extraction and data sharing.
Collapse
Affiliation(s)
- Paola Crivelli
- Department of Biomedical Sciences, Institute of Radiological Sciences, University of Sassari, Sassari, Italy
| | - Roberta Eufrasia Ledda
- Department of Clinical and Experimental Medicine, Institute of Radiological Sciences, University of Sassari, Sassari, Italy
| | - Nicola Parascandolo
- Department of Clinical and Experimental Medicine, Institute of Radiological Sciences, University of Sassari, Sassari, Italy
| | - Alberto Fara
- Department of Clinical and Experimental Medicine, Institute of Radiological Sciences, University of Sassari, Sassari, Italy
| | - Daniela Soro
- Department of Clinical and Experimental Medicine, Institute of Radiological Sciences, University of Sassari, Sassari, Italy
| | - Maurizio Conti
- Department of Clinical and Experimental Medicine, Institute of Radiological Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
47
|
Pujara AC, Kim E, Axelrod D, Melsaether AN. PET/MRI in Breast Cancer. J Magn Reson Imaging 2018; 49:328-342. [DOI: 10.1002/jmri.26298] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Akshat C. Pujara
- Department of Radiology, Division of Breast Imaging; University of Michigan Health System; Ann Arbor Michigan USA
| | - Eric Kim
- Department of Radiology; NYU School of Medicine; New York New York USA
| | - Deborah Axelrod
- Department of Surgery; Perlmutter Cancer Center, NYU School of Medicine; New York New York USA
| | - Amy N. Melsaether
- Department of Radiology; NYU School of Medicine; New York New York USA
| |
Collapse
|
48
|
Morin O, Vallières M, Jochems A, Woodruff HC, Valdes G, Braunstein SE, Wildberger JE, Villanueva-Meyer JE, Kearney V, Yom SS, Solberg TD, Lambin P. A Deep Look Into the Future of Quantitative Imaging in Oncology: A Statement of Working Principles and Proposal for Change. Int J Radiat Oncol Biol Phys 2018; 102:1074-1082. [PMID: 30170101 DOI: 10.1016/j.ijrobp.2018.08.032] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/21/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022]
Abstract
The adoption of enterprise digital imaging, along with the development of quantitative imaging methods and the re-emergence of statistical learning, has opened the opportunity for more personalized cancer treatments through transformative data science research. In the last 5 years, accumulating evidence has indicated that noninvasive advanced imaging analytics (i.e., radiomics) can reveal key components of tumor phenotype for multiple lesions at multiple time points over the course of treatment. Many groups using homegrown software have extracted engineered and deep quantitative features on 3-dimensional medical images for better spatial and longitudinal understanding of tumor biology and for the prediction of diverse outcomes. These developments could augment patient stratification and prognostication, buttressing emerging targeted therapeutic approaches. Unfortunately, the rapid growth in popularity of this immature scientific discipline has resulted in many early publications that miss key information or use underpowered patient data sets, without production of generalizable results. Quantitative imaging research is complex, and key principles should be followed to realize its full potential. The fields of quantitative imaging and radiomics in particular require a renewed focus on optimal study design and reporting practices, standardization, interpretability, data sharing, and clinical trials. Standardization of image acquisition, feature calculation, and statistical analysis (i.e., machine learning) are required for the field to move forward. A new data-sharing paradigm enacted among open and diverse participants (medical institutions, vendors and associations) should be embraced for faster development and comprehensive clinical validation of imaging biomarkers. In this review and critique of the field, we propose working principles and fundamental changes to the current scientific approach, with the goal of high-impact research and development of actionable prediction models that will yield more meaningful applications of precision cancer medicine.
Collapse
Affiliation(s)
- Olivier Morin
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California.
| | | | - Arthur Jochems
- The D-Lab, Grow Research Institute for Oncology, Maastricht University, Maastricht, The Netherlands
| | - Henry C Woodruff
- The D-Lab, Grow Research Institute for Oncology, Maastricht University, Maastricht, The Netherlands
| | - Gilmer Valdes
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Steve E Braunstein
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Joachim E Wildberger
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Vasant Kearney
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Sue S Yom
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Timothy D Solberg
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Philippe Lambin
- The D-Lab, Grow Research Institute for Oncology, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|