1
|
Kai C, Tamori H, Ohtsuka T, Nara M, Yoshida A, Sato I, Futamura H, Kodama N, Kasai S. Classifying the molecular subtype of breast cancer using vision transformer and convolutional neural network features. Breast Cancer Res Treat 2025; 210:771-782. [PMID: 39841349 DOI: 10.1007/s10549-025-07614-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/10/2025] [Indexed: 01/23/2025]
Abstract
PURPOSE Identification of the molecular subtypes in breast cancer allows to optimize treatment strategies, but usually requires invasive needle biopsy. Recently, non-invasive imaging has emerged as promising means to classify them. Magnetic resonance imaging is often used for this purpose because it is three-dimensional and highly informative. Instead, only a few reports have documented the use of mammograms. Given that mammography is the first choice for breast cancer screening, using it to classify molecular subtypes would allow for early intervention on a much wider scale. Here, we aimed to evaluate the effectiveness of combining global and local mammographic features by using Vision Transformer (ViT) and Convolutional Neural Network (CNN) to classify molecular subtypes in breast cancer. METHODS The feature values for binary classification were calculated using the ViT and EfficientnetV2 feature extractors, followed by dimensional compression via principal component analysis. LightGBM was used to perform binary classification of each molecular subtype: triple-negative, HER2-enriched, luminal A, and luminal B. RESULTS The combination of ViT and CNN achieved higher accuracy than ViT or CNN alone. The sensitivity for triple-negative subtypes was very high (0.900, with F-value = 0.818); whereas F-value and sensitivity were 0.720 and 0.750 for HER2-enriched, 0.765 and 0.867 for luminal A, and 0.614 and 0.711 for luminal B subtypes, respectively. CONCLUSION Features obtained from mammograms by combining ViT and CNN allow the classification of molecular subtypes with high accuracy. This approach could streamline early treatment workflows and triage, especially for poor prognosis subtypes such as triple-negative breast cancer.
Collapse
Affiliation(s)
- Chiharu Kai
- Department of Radiological Technology, Faculty of Medical Technology, Niigata University of Health and Welfare, 1398 Shimamichou, Kita-Ku, Niigata, Japan
- Major in Health and Welfare, Graduate School of Niigata, University of Health and Welfare, Niigata, Japan
| | | | | | | | - Akifumi Yoshida
- Department of Radiological Technology, Faculty of Medical Technology, Niigata University of Health and Welfare, 1398 Shimamichou, Kita-Ku, Niigata, Japan
| | - Ikumi Sato
- Major in Health and Welfare, Graduate School of Niigata, University of Health and Welfare, Niigata, Japan
- Department of Nursing, Faculty of Nursing, Niigata University of Health and Welfare, Niigata City, Niigata, Japan
| | | | - Naoki Kodama
- Department of Radiological Technology, Faculty of Medical Technology, Niigata University of Health and Welfare, 1398 Shimamichou, Kita-Ku, Niigata, Japan
| | - Satoshi Kasai
- Department of Radiological Technology, Faculty of Medical Technology, Niigata University of Health and Welfare, 1398 Shimamichou, Kita-Ku, Niigata, Japan.
| |
Collapse
|
2
|
Tsarouchi M, Hoxhaj A, Portaluri A, Sung J, Sechopoulos I, Pinker-Domenig K, Mann RM. Breast cancer staging with contrast-enhanced imaging. The benefits and drawbacks of MRI, CEM, and dedicated breast CT. Eur J Radiol 2025; 185:112013. [PMID: 40036929 DOI: 10.1016/j.ejrad.2025.112013] [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: 12/18/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
Pretherapeutic breast cancer staging is pivotal for patient-centered disease management, guiding treatment stratification and assessing prognostic outcomes. Breast imaging plays a key role in both anatomic and prognostic staging by providing comprehensive insights into disease extent, localization, and tumor aggressiveness characteristics. To date, clinical image-based staging relies on conventional modalities such as digital mammography (DM), digital breast tomosynthesis (DBT), and ultrasound. Considering the phenotypic disparities of breast cancer and their relation to treatment response, other imaging techniques based on contrast-enhanced mechanisms, which highlight areas of increased neovascularity, appear indispensable in breast cancer staging. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) offers a comprehensive 3-dimensional assessment of extent of disease and characterization of lesions, capturing both morphological and functional aspects which are crucial for treatment and monitoring of the disease. Based on this established approach, contrast-enhanced x-ray-based techniques, with high spatial resolution, such as contrast-enhanced mammography (CEM) and dedicated contrast-enhanced breast computed tomography (dCEBCT), have emerged. This review outlines the current status, limitations, and ongoing challenges associated with each one contrast-enhanced imaging modality, while emphasis is given to key breast cancer manifestations and the optimal interpretation of their imaging phenotypes, in the current era of image-based (anatomic and prognostic) breast cancer staging.
Collapse
Affiliation(s)
- Marialena Tsarouchi
- Department of Radiology and Nuclear Medicine, Radboudumc, Geert Grooteplein 10, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| | - Alma Hoxhaj
- Department of Radiology and Nuclear Medicine, Radboudumc, Geert Grooteplein 10, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Antonio Portaluri
- Department of Biomedical Sciences and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
| | - Janice Sung
- Department of Radiology, Columbia University Irving Medical Center 161 Fort Washington Avenue, New York, NY 10032, The United States
| | - Ioannis Sechopoulos
- Department of Radiology and Nuclear Medicine, Radboudumc, Geert Grooteplein 10, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Katja Pinker-Domenig
- Department of Radiology, Columbia University Irving Medical Center 161 Fort Washington Avenue, New York, NY 10032, The United States
| | - Ritse M Mann
- Department of Radiology and Nuclear Medicine, Radboudumc, Geert Grooteplein 10, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| |
Collapse
|
3
|
Ventura C, Fogante M, Marconi E, Franca Simonetti B, Borgoforte Gradassi S, Carboni N, Lenti E, Argalia G. Morphodynamic Features of Contrast-Enhanced Mammography and Their Correlation with Breast Cancer Histopathology. J Imaging 2025; 11:80. [PMID: 40137192 PMCID: PMC11942963 DOI: 10.3390/jimaging11030080] [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: 02/15/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Contrast-enhanced mammography (CEM) combines morphological and functional imaging, enhancing breast cancer (BC) diagnosis. This study investigates the relationship between CEM morphodynamic features and histopathological characteristics of BC. In this prospective study, 50 female patients (mean age: 57.2 ± 13.7 years) with BI-RADS 4-5 lesions underwent CEM followed by surgical excision between December 2022 and May 2024. Low-energy and recombined CEM images were analyzed for breast composition, lesion characteristics, and enhancement patterns, while histopathological evaluation included tumor size, histotype, grade, lymphovascular invasion, and immunophenotype. Spearman rank correlation and multivariable regression analysis were used to evaluate the relationship between CEM findings and histopathological characteristics. Tumor size on CEM strongly correlated with histopathological tumor size (ρ = 0.788, p < 0.001) and was associated with high-grade lesions (p = 0.017). Non-circumscribed margins were linked to a Luminal-B subtype (p = 0.001), while high lesion conspicuity was associated with Luminal-B and triple-negative BC (p = 0.001) and correlated with larger tumors (ρ = 0.517, p < 0.001). Background parenchymal enhancement was negatively correlated with age (ρ = -0.286, p = 0.049). CEM provides critical insights into BC, demonstrating significant relationship between imaging features and histopathological characteristics. These findings highlight CEM's potential as a reliable tool for tumor size estimation, subtype characterization, and prognostic assessment, suggesting its role as an alternative to MRI, particularly for patients with contraindications.
Collapse
Affiliation(s)
- Claudio Ventura
- SOD Radiologia Materno Infantile, Senologica, Cardiologica ed Ecografica Ambulatoriale, Azienda Ospedaliero Universitaria delle Marche, Via Conca 71, 60126 Ancona, Italy; (C.V.); (E.M.); (B.F.S.); (S.B.G.); (N.C.); (G.A.)
| | - Marco Fogante
- SOD Radiologia Materno Infantile, Senologica, Cardiologica ed Ecografica Ambulatoriale, Azienda Ospedaliero Universitaria delle Marche, Via Conca 71, 60126 Ancona, Italy; (C.V.); (E.M.); (B.F.S.); (S.B.G.); (N.C.); (G.A.)
| | - Elisabetta Marconi
- SOD Radiologia Materno Infantile, Senologica, Cardiologica ed Ecografica Ambulatoriale, Azienda Ospedaliero Universitaria delle Marche, Via Conca 71, 60126 Ancona, Italy; (C.V.); (E.M.); (B.F.S.); (S.B.G.); (N.C.); (G.A.)
| | - Barbara Franca Simonetti
- SOD Radiologia Materno Infantile, Senologica, Cardiologica ed Ecografica Ambulatoriale, Azienda Ospedaliero Universitaria delle Marche, Via Conca 71, 60126 Ancona, Italy; (C.V.); (E.M.); (B.F.S.); (S.B.G.); (N.C.); (G.A.)
| | - Silvia Borgoforte Gradassi
- SOD Radiologia Materno Infantile, Senologica, Cardiologica ed Ecografica Ambulatoriale, Azienda Ospedaliero Universitaria delle Marche, Via Conca 71, 60126 Ancona, Italy; (C.V.); (E.M.); (B.F.S.); (S.B.G.); (N.C.); (G.A.)
| | - Nicola Carboni
- SOD Radiologia Materno Infantile, Senologica, Cardiologica ed Ecografica Ambulatoriale, Azienda Ospedaliero Universitaria delle Marche, Via Conca 71, 60126 Ancona, Italy; (C.V.); (E.M.); (B.F.S.); (S.B.G.); (N.C.); (G.A.)
| | - Enrico Lenti
- SOD Chirurgia Senologica, Azienda Ospedaliero Universitaria delle Marche, Via Conca 71, 60126 Ancona, Italy;
| | - Giulio Argalia
- SOD Radiologia Materno Infantile, Senologica, Cardiologica ed Ecografica Ambulatoriale, Azienda Ospedaliero Universitaria delle Marche, Via Conca 71, 60126 Ancona, Italy; (C.V.); (E.M.); (B.F.S.); (S.B.G.); (N.C.); (G.A.)
| |
Collapse
|
4
|
Han F, Li W, Hu Y, Wang H, Liu T, Wu J. MRI Radiomics-Based Machine Learning to Predict Lymphovascular Invasion of HER2-Positive Breast Cancer. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024:10.1007/s10278-024-01329-x. [PMID: 39538052 DOI: 10.1007/s10278-024-01329-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/23/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
This study aims to develop and prospectively validate radiomic models based on MRI to predict lymphovascular invasion (LVI) status in patients with HER2-positive breast cancer. A total of 225 patients with HER2-positive breast cancer who preoperatively underwent breast MRI were selected, forming the training set (n = 99 LVI-positive, n = 126 LVI-negative). A prospective validation cohort included 130 patients with breast cancer from the Affiliated Zhongshan Hospital of Dalian University (n = 57 LVI-positive, n = 73 LVI-negative). A total of 390 radiomic features and eight conventional radiological characteristics were extracted. For the optimum feature selection phase, the LASSO regression model with tenfold cross-validation (CV) was employed to identify features with non-zero coefficients. The conventional radiological (CR) model was determined based on visual morphological (VM) features and the optimal radiomic features correlated with LVI, identified through multivariate logistic analyses. Subsequently, various machine learning (ML) models were developed using algorithms such as support vector machine (SVM), k-nearest neighbor (KNN), gradient boosting machine (GBM), and random forest (RF). The performance of ML and CR models. The results show that the AUC of the CR model in the training and validation sets were 0.81 (95% confidence interval [CI], 0.74-0.86) and 0.82 (95% CI, 0.69-0.89), respectively. The ML model achieved the best performance, with AUCs of 0.96 (95% CI, 0.99-1.00) in the training set and 0.95 (95% CI, 0.89-0.96) in the validation set. There were significant differences between the CR and ML models in predicting LVI status. Our study demonstrated that the machine learning models exhibited superior performance in predicting LVI status based on pretreatment MRI compared to the CR model, which does not necessarily rely on a priori knowledge of visual morphology.
Collapse
Affiliation(s)
- Fang Han
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Wenfei Li
- Department of Radiology, First Hospital of Qinhuangdao, Qinhuangdao, 066000, Hebei, China.
| | - Yurui Hu
- Department of Radiology, First Hospital of Qinhuangdao, Qinhuangdao, 066000, Hebei, China
| | - Huiping Wang
- Department of Radiology, People's Hospital of Pingyao County, Jinzhong, 031100, Shanxi, China
| | - Tianyu Liu
- Department of Breast Surgery, First Hospital of Qinhuangdao, Qinhuangdao, 066000, Hebei, China
| | - Jianlin Wu
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| |
Collapse
|
5
|
Wang S, Wang T, Guo S, Zhu S, Chen R, Zheng J, Jiang T, Li R, Li J, Li J, Shen X, Qian M, Yang M, Yu S, You C, Gu Y. Whole tumour- and subregion-based radiomics of contrast-enhanced mammography in differentiating HER2 expression status of invasive breast cancers: A double-centre pilot study. Br J Cancer 2024; 131:1613-1622. [PMID: 39379571 PMCID: PMC11554679 DOI: 10.1038/s41416-024-02871-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
OBJECTIVES To explore the value of whole tumour- and subregion-based radiomics of contrast-enhanced mammography (CEM) in differentiating the HER2 expression status of breast cancers. METHODS 352 patients underwent preoperative CEM from two centres were consecutively enroled and divided into the training, internal validation, and external validation cohorts. The lesions were divided into HER2-positive and HER2-negative groups. Besides the radiological features, radiomics features capturing the whole tumour-based (wITH) and subregion-based intratumoral heterogeneity (sITH) were extracted from the craniocaudal view of CEM recombined images. The XGBoost classifier was applied to develop the radiological, sITH, and wITH models. A combined model was constructed by fusing the prediction results of the three models. RESULTS The mean age of the patients was 51.1 ± 10.7 years. Two radiological features, four wITH features, and three sITH features were selected to establish the models. The combined model significantly improved the AUC to 0.80 ± 0.03 (95% CI: 0.73-0.86), 0.79 ± 0.06 (95% CI: 0.67-0.90), and 0.79 ± 0.05 (95% CI: 0.69-0.89) in the training, internal validation, and external validation cohorts, respectively (All P < 0.05). The combined model showed good agreement between the predicted and observed probabilities and favourable net clinical benefit in the validation cohorts. CONCLUSIONS Both whole tumour- and subregion-based ITH radiomics features of CEM exhibited potential for differentiating the HER2 expression status. Combining conventional radiological features and ITH features can improve the model's performance.
Collapse
Affiliation(s)
- Simin Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ting Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sailing Guo
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Shuangshuang Zhu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Ruchuan Chen
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
| | - Jinlong Zheng
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
| | - Tingting Jiang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruimin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinhui Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
| | - Jiawei Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xigang Shen
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Min Qian
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Meng Yang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shengnan Yu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Chao You
- 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
|
6
|
Filippone F, Boudagga Z, Frattini F, Fortuna GF, Razzini D, Tambasco A, Menardi V, Balbiano di Colcavagno A, Carriero S, Gambaro ACL, Carriero A. Contrast Enhancement in Breast Cancer: Magnetic Resonance vs. Mammography: A 10-Year Systematic Review. Diagnostics (Basel) 2024; 14:2400. [PMID: 39518367 PMCID: PMC11545212 DOI: 10.3390/diagnostics14212400] [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: 05/20/2024] [Revised: 09/25/2024] [Accepted: 10/05/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE Contrast Enhancement Magnetic Resonance (CEMR) and Contrast-Enhanced Mammography (CEM) are important diagnostic tools to evaluate breast cancer patients, and both are objects of interest in the literature. The purpose of this systematic review was to select publications from the last ten years in order to evaluate the literature contributions related to the frequency of contrast agents used, administration techniques and the presence of adverse reactions. METHODS We have selected, according to the PRISMA statement, publications reviewed on Pub Med in the period from 1 January 2012 to 31 December 2022. The search engine was activated using the following keywords: "CESM", "CEM", "CEDM", "Contrast mammography" for CEM, "DCE-MRI", "Contrast Enhancement MRI" for CEMR, excluding reviews, book chapters and meta-analyses. From the total number of publications, we made a preliminary selection based on titles and abstracts and excluded all articles published in languages other than English and all experimental studies performed on surgical specimen or animal population, as well as all articles for which the extended version was not available. Two readers evaluated all the articles and compiled a pre-compiled form accordingly. RESULTS After a preliminary collection of 571 CEM publications, 118 articles were selected, relating to an overall population of 21,178 patients. From a total of 3063 CEMR publications, 356 articles relating to an overall population of 45,649 patients were selected. The most used contrast agents are Iohexol for CEM (39.83%) and Gadopentetic acid (Gd-DTPA) for CEMR (32.5%). Regarding the CEM contrast administration protocol, in 84.7% of cases a dose of 1.5 mL/kg was used with an infusion rate of 2-3 mL/s. Regarding the CEMR infusion protocol, in 71% of cases a dose of 1 mmol/kg was used at an infusion rate of 2-4 mL/s. Twelve out of 118 CEM articles reported allergic reactions, involving 29 patients (0.13%). In DCE-MRI, only one out of 356 articles reported allergic reactions, involving two patients (0.004%). No severe reactions were observed in either cohort of exams. CONCLUSIONS CEM and CEMR are essential contrast methods to evaluate breast diseases. However, from the literature analysis, although there are preferences on the uses of the contrast agent (Iohexol for CESM, G-DTPA for CEMR), a wide range of molecules are still used in contrast methods, with different administration protocols. Based on the collected data, it is possible to state that both methods are safe, and no severe reactions were observed in our evaluation.
Collapse
Affiliation(s)
- Francesco Filippone
- SCDU Radiology, “Maggiore della Carità” Hospital, University of Eastern Piedmont, 28100 Novara, Italy; (F.F.); (G.F.F.); (D.R.); (A.T.); (V.M.); (A.B.d.C.); (A.C.L.G.); (A.C.)
| | - Zohra Boudagga
- SCDU Radiology, “Maggiore della Carità” Hospital, University of Eastern Piedmont, 28100 Novara, Italy; (F.F.); (G.F.F.); (D.R.); (A.T.); (V.M.); (A.B.d.C.); (A.C.L.G.); (A.C.)
| | - Francesca Frattini
- SCDU Radiology, “Maggiore della Carità” Hospital, University of Eastern Piedmont, 28100 Novara, Italy; (F.F.); (G.F.F.); (D.R.); (A.T.); (V.M.); (A.B.d.C.); (A.C.L.G.); (A.C.)
| | - Gaetano Federico Fortuna
- SCDU Radiology, “Maggiore della Carità” Hospital, University of Eastern Piedmont, 28100 Novara, Italy; (F.F.); (G.F.F.); (D.R.); (A.T.); (V.M.); (A.B.d.C.); (A.C.L.G.); (A.C.)
| | - Davide Razzini
- SCDU Radiology, “Maggiore della Carità” Hospital, University of Eastern Piedmont, 28100 Novara, Italy; (F.F.); (G.F.F.); (D.R.); (A.T.); (V.M.); (A.B.d.C.); (A.C.L.G.); (A.C.)
| | - Anna Tambasco
- SCDU Radiology, “Maggiore della Carità” Hospital, University of Eastern Piedmont, 28100 Novara, Italy; (F.F.); (G.F.F.); (D.R.); (A.T.); (V.M.); (A.B.d.C.); (A.C.L.G.); (A.C.)
| | - Veronica Menardi
- SCDU Radiology, “Maggiore della Carità” Hospital, University of Eastern Piedmont, 28100 Novara, Italy; (F.F.); (G.F.F.); (D.R.); (A.T.); (V.M.); (A.B.d.C.); (A.C.L.G.); (A.C.)
| | - Alessandro Balbiano di Colcavagno
- SCDU Radiology, “Maggiore della Carità” Hospital, University of Eastern Piedmont, 28100 Novara, Italy; (F.F.); (G.F.F.); (D.R.); (A.T.); (V.M.); (A.B.d.C.); (A.C.L.G.); (A.C.)
| | - Serena Carriero
- Foundation IRCCS Cà Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Anna Clelia Lucia Gambaro
- SCDU Radiology, “Maggiore della Carità” Hospital, University of Eastern Piedmont, 28100 Novara, Italy; (F.F.); (G.F.F.); (D.R.); (A.T.); (V.M.); (A.B.d.C.); (A.C.L.G.); (A.C.)
| | - Alessandro Carriero
- SCDU Radiology, “Maggiore della Carità” Hospital, University of Eastern Piedmont, 28100 Novara, Italy; (F.F.); (G.F.F.); (D.R.); (A.T.); (V.M.); (A.B.d.C.); (A.C.L.G.); (A.C.)
| |
Collapse
|
7
|
Bakker MAG, Ovalho MDL, Matela N, Mota AM. Decoding Breast Cancer: Using Radiomics to Non-Invasively Unveil Molecular Subtypes Directly from Mammographic Images. J Imaging 2024; 10:218. [PMID: 39330438 PMCID: PMC11432960 DOI: 10.3390/jimaging10090218] [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: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide. The therapy used and its success depend highly on the histology of the tumor. This study aimed to explore the potential of predicting the molecular subtype of breast cancer using radiomic features extracted from screening digital mammography (DM) images. A retrospective study was performed using the OPTIMAM Mammography Image Database (OMI-DB). Four binary classification tasks were performed: luminal A vs. non-luminal A, luminal B vs. non-luminal B, TNBC vs. non-TNBC, and HER2 vs. non-HER2. Feature selection was carried out by Pearson correlation and LASSO. The support vector machine (SVM) and naive Bayes (NB) ML classifiers were used, and their performance was evaluated with the accuracy and the area under the receiver operating characteristic curve (AUC). A total of 186 patients were included in the study: 58 luminal A, 35 luminal B, 52 TNBC, and 41 HER2. The SVM classifier resulted in AUCs during testing of 0.855 for luminal A, 0.812 for luminal B, 0.789 for TNBC, and 0.755 for HER2, respectively. The NB classifier showed AUCs during testing of 0.714 for luminal A, 0.746 for luminal B, 0.593 for TNBC, and 0.714 for HER2. The SVM classifier outperformed NB with statistical significance for luminal A (p = 0.0268) and TNBC (p = 0.0073). Our study showed the potential of radiomics for non-invasive breast cancer subtype classification.
Collapse
Affiliation(s)
- Manon A. G. Bakker
- Faculty of Science and Engineering, University of Groningen, 9700 AS Groningen, The Netherlands
| | - Maria de Lurdes Ovalho
- Departamento de Radiologia, Hospital da Luz Lisboa, Luz Saúde, 1500-650 Lisboa, Portugal
| | - Nuno Matela
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1649-004 Lisbon, Portugal
- Departamento de Física, Faculdade de Ciências, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Ana M. Mota
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1649-004 Lisbon, Portugal
- Departamento de Física, Faculdade de Ciências, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| |
Collapse
|
8
|
Baniasadi A, Das JP, Prendergast CM, Beizavi Z, Ma HY, Jaber MY, Capaccione KM. Imaging at the nexus: how state of the art imaging techniques can enhance our understanding of cancer and fibrosis. J Transl Med 2024; 22:567. [PMID: 38872212 PMCID: PMC11177383 DOI: 10.1186/s12967-024-05379-1] [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: 02/11/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Both cancer and fibrosis are diseases involving dysregulation of cell signaling pathways resulting in an altered cellular microenvironment which ultimately leads to progression of the condition. The two disease entities share common molecular pathophysiology and recent research has illuminated the how each promotes the other. Multiple imaging techniques have been developed to aid in the early and accurate diagnosis of each disease, and given the commonalities between the pathophysiology of the conditions, advances in imaging one disease have opened new avenues to study the other. Here, we detail the most up-to-date advances in imaging techniques for each disease and how they have crossed over to improve detection and monitoring of the other. We explore techniques in positron emission tomography (PET), magnetic resonance imaging (MRI), second generation harmonic Imaging (SGHI), ultrasound (US), radiomics, and artificial intelligence (AI). A new diagnostic imaging tool in PET/computed tomography (CT) is the use of radiolabeled fibroblast activation protein inhibitor (FAPI). SGHI uses high-frequency sound waves to penetrate deeper into the tissue, providing a more detailed view of the tumor microenvironment. Artificial intelligence with the aid of advanced deep learning (DL) algorithms has been highly effective in training computer systems to diagnose and classify neoplastic lesions in multiple organs. Ultimately, advancing imaging techniques in cancer and fibrosis can lead to significantly more timely and accurate diagnoses of both diseases resulting in better patient outcomes.
Collapse
Affiliation(s)
- Alireza Baniasadi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA.
| | - Jeeban P Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Conor M Prendergast
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Zahra Beizavi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Hong Y Ma
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | | | - Kathleen M Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| |
Collapse
|
9
|
Zhu S, Wang S, Guo S, Wu R, Zhang J, Kong M, Pan L, Gu Y, Yu S. Contrast-Enhanced Mammography Radiomics Analysis for Preoperative Prediction of Breast Cancer Molecular Subtypes. Acad Radiol 2024; 31:2228-2238. [PMID: 38142176 DOI: 10.1016/j.acra.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND Predicting breast cancer molecular subtypes can help guide individualised clinical treatment of patients who need the rational preoperative treatment. This study aimed to investigate the efficacy of preoperative prediction of breast cancer molecular subtypes by contrast-enhanced mammography (CEM) radiomic features. METHODS This retrospective two-centre study included women with breast cancer who underwent CEM preoperatively between August 2016 and May 2022. We included 356 patients with 386 lesions, which were grouped into training (n = 162), internal test (n = 160) and external test sets (n = 64). Radiomics features were extracted from low-energy (LE) images and recombined (RC) images and selected. Three dichotomous tasks were established according to postoperative immunohistochemical results: Luminal vs. non-Luminal, human epidermal growth factor receptor (HER2)-enriched vs. non-HER2-enriched, and triple-negative breast cancer (TNBC) vs. non-TNBC. For each dichotomous task, the LE, RC, and LE+RC radiomics models were built by the support vector machine classifier. The prediction performance of the models was assessed by the area under the receiver operating characteristic curve (AUC). Then, the accuracy, sensitivity, specificity, positive predictive value, and negative predictive value were calculated for the models. DeLong's test was utilised to compare the AUCs. RESULTS Radiomics models based on CEM are valuable for predicting breast cancer molecular subtypes. The LE+RC model achieved the best performance in the test set. The LE+RC model predicted Luminal, HER2-enriched, and TNBC subtypes with AUCs of 0.93, 0.89, and 0.87 in the internal test set and 0.82, 0.83, and 0.69 in the external test set, respectively. In addition, the LE model performed more satisfactorily than the RC model. CONCLUSION CEM radiomics features can effectively predict breast cancer molecular subtypes preoperatively, and the LE+RC model has the best predictive performance.
Collapse
Affiliation(s)
- Shuangshuang Zhu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China (S.Z., S.G., R.W., J.Z., M.K., L.P., S.Y.)
| | - Simin Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China (S.W., Y.G.)
| | - Sailing Guo
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China (S.Z., S.G., R.W., J.Z., M.K., L.P., S.Y.)
| | - Ruoxi Wu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China (S.Z., S.G., R.W., J.Z., M.K., L.P., S.Y.)
| | - Jinggang Zhang
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China (S.Z., S.G., R.W., J.Z., M.K., L.P., S.Y.)
| | - Mengyu Kong
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China (S.Z., S.G., R.W., J.Z., M.K., L.P., S.Y.)
| | - Liang Pan
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China (S.Z., S.G., R.W., J.Z., M.K., L.P., S.Y.)
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China (S.W., Y.G.)
| | - Shengnan Yu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China (S.Z., S.G., R.W., J.Z., M.K., L.P., S.Y.).
| |
Collapse
|
10
|
Petrillo A, Fusco R, Petrosino T, Vallone P, Granata V, Rubulotta MR, Pariante P, Raiano N, Scognamiglio G, Fanizzi A, Massafra R, Lafranceschina M, La Forgia D, Greco L, Ferranti FR, De Soccio V, Vidiri A, Botta F, Dominelli V, Cassano E, Sorgente E, Pecori B, Cerciello V, Boldrini L. A multicentric study of radiomics and artificial intelligence analysis on contrast-enhanced mammography to identify different histotypes of breast cancer. LA RADIOLOGIA MEDICA 2024; 129:864-878. [PMID: 38755477 DOI: 10.1007/s11547-024-01817-8] [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: 09/27/2023] [Accepted: 04/16/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVE To evaluate the performance of radiomic analysis on contrast-enhanced mammography images to identify different histotypes of breast cancer mainly in order to predict grading, to identify hormone receptors, to discriminate human epidermal growth factor receptor 2 (HER2) and to identify luminal histotype of the breast cancer. METHODS From four Italian centers were recruited 180 malignant lesions and 68 benign lesions. However, only the malignant lesions were considered for the analysis. All patients underwent contrast-enhanced mammography in cranium caudal (CC) and medium lateral oblique (MLO) view. Considering histological findings as the ground truth, four outcomes were considered: (1) G1 + G2 vs. G3; (2) HER2 + vs. HER2 - ; (3) HR + vs. HR - ; and (4) non-luminal vs. luminal A or HR + /HER2- and luminal B or HR + /HER2 + . For multivariate analysis feature selection, balancing techniques and patter recognition approaches were considered. RESULTS The univariate findings showed that the diagnostic performance is low for each outcome, while the results of the multivariate analysis showed that better performances can be obtained. In the HER2 + detection, the best performance (73% of accuracy and AUC = 0.77) was obtained using a linear regression model (LRM) with 12 features extracted by MLO view. In the HR + detection, the best performance (77% of accuracy and AUC = 0.80) was obtained using a LRM with 14 features extracted by MLO view. In grading classification, the best performance was obtained by a decision tree trained with three predictors extracted by MLO view reaching an accuracy of 82% on validation set. In the luminal versus non-luminal histotype classification, the best performance was obtained by a bagged tree trained with 15 predictors extracted by CC view reaching an accuracy of 94% on validation set. CONCLUSIONS The results suggest that radiomics analysis can be effectively applied to design a tool to support physician decision making in breast cancer classification. In particular, the classification of luminal versus non-luminal histotypes can be performed with high accuracy.
Collapse
Affiliation(s)
- Antonella Petrillo
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy.
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 80013, Naples, Italy
| | - Teresa Petrosino
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Paolo Vallone
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Vincenza Granata
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Maria Rosaria Rubulotta
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Paolo Pariante
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Nicola Raiano
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Giosuè Scognamiglio
- Pathology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Annarita Fanizzi
- Direzione Scientifica, IRCCS Istituto Tumori Giovanni Paolo II, Via Orazio Flacco 65, 70124, Bari, Italy
| | - Raffaella Massafra
- SSD Fisica Sanitaria, IRCCS Istituto Tumori Giovanni Paolo II, Via Orazio Flacco 65, 70124, Bari, Italy
| | - Miria Lafranceschina
- Struttura Semplice Dipartimentale Di Radiodiagnostica Senologica, IRCCS Istituto Tumori Giovanni Paolo II, Via Orazio Flacco 65, 70124, Bari, Italy
| | - Daniele La Forgia
- Struttura Semplice Dipartimentale Di Radiodiagnostica Senologica, IRCCS Istituto Tumori Giovanni Paolo II, Via Orazio Flacco 65, 70124, Bari, Italy
| | - Laura Greco
- Radiology and Diagnostic Imaging, Istituto Di Ricovero E Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Romana Ferranti
- Radiology and Diagnostic Imaging, Istituto Di Ricovero E Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Valeria De Soccio
- Radiology and Diagnostic Imaging, Istituto Di Ricovero E Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging, Istituto Di Ricovero E Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Botta
- Breast Imaging Division, IEO Istituto Europeo Di Oncologia, 20141, Milan, Italy
| | - Valeria Dominelli
- Breast Imaging Division, IEO Istituto Europeo Di Oncologia, 20141, Milan, Italy
| | - Enrico Cassano
- Breast Imaging Division, IEO Istituto Europeo Di Oncologia, 20141, Milan, Italy
| | - Eugenio Sorgente
- Radiation Protection and Innovative Technology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Biagio Pecori
- Radiation Protection and Innovative Technology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Vincenzo Cerciello
- Medical Physics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Luca Boldrini
- Dipartimento Di Diagnostica Per Immagini, Radioterapia Oncologica Ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| |
Collapse
|
11
|
Kang D, Wang C, Han Z, Zheng L, Guo W, Fu F, Qiu L, Han X, He J, Li L, Chen J. Exploration of the relationship between tumor-infiltrating lymphocyte score and histological grade in breast cancer. BMC Cancer 2024; 24:318. [PMID: 38454386 PMCID: PMC10921807 DOI: 10.1186/s12885-024-12069-0] [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: 09/21/2023] [Accepted: 02/28/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND The histological grade is an important factor in the prognosis of invasive breast cancer and is vital to accurately identify the histological grade and reclassify of Grade2 status in breast cancer patients. METHODS In this study, data were collected from 556 invasive breast cancer patients, and then randomly divided into training cohort (n = 335) and validation cohort (n = 221). All patients were divided into actual low risk group (Grade1) and high risk group (Grade2/3) based on traditional histological grade, and tumor-infiltrating lymphocyte score (TILs-score) obtained from multiphoton images, and the TILs assessment method proposed by International Immuno-Oncology Biomarker Working Group (TILs-WG) were also used to differentiate between high risk group and low risk group of histological grade in patients with invasive breast cancer. Furthermore, TILs-score was used to reclassify Grade2 (G2) into G2 /Low risk and G2/High risk. The coefficients for each TILs in the training cohort were retrieved using ridge regression and TILs-score was created based on the coefficients of the three kinds of TILs. RESULTS Statistical analysis shows that TILs-score is significantly correlated with histological grade, and is an independent predictor of histological grade (odds ratio [OR], 2.548; 95%CI, 1.648-3.941; P < 0.0001), but TILs-WG is not an independent predictive factor for grade (P > 0.05 in the univariate analysis). Moreover, the risk of G2/High risk group is higher than that of G2/Low risk group, and the survival rate of patients with G2/Low risk is similar to that of Grade1, while the survival rate of patients with G2/High risk is even worse than that of patients with G3. CONCLUSION Our results suggest that TILs-score can be used to predict the histological grade of breast cancer and potentially to guide the therapeutic management of breast cancer patients.
Collapse
Affiliation(s)
- Deyong Kang
- Department of Pathology, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Chuan Wang
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Zhonghua Han
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Liqin Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China
| | - Wenhui Guo
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Fangmeng Fu
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Lida Qiu
- College of Physics and Electronic Information Engineering, Minjiang University, 350108, Fuzhou, P. R. China
| | - Xiahui Han
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China
| | - Jiajia He
- School of Science, Jimei University, 361021, Xiamen, P. R. China.
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China.
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China.
| |
Collapse
|
12
|
Bhalla K, Xiao Q, Luna JM, Podany E, Ahmad T, Ademuyiwa FO, Davis A, Bennett DL, Gastounioti A. Radiologic imaging biomarkers in triple-negative breast cancer: a literature review about the role of artificial intelligence and the way forward. BJR ARTIFICIAL INTELLIGENCE 2024; 1:ubae016. [PMID: 40201726 PMCID: PMC11974408 DOI: 10.1093/bjrai/ubae016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/27/2024] [Accepted: 11/10/2024] [Indexed: 04/10/2025]
Abstract
Breast cancer is one of the most common and deadly cancers in women. Triple-negative breast cancer (TNBC) accounts for approximately 10%-15% of breast cancer diagnoses and is an aggressive molecular breast cancer subtype associated with important challenges in its diagnosis, treatment, and prognostication. This poses an urgent need for developing more effective and personalized imaging biomarkers for TNBC. Towards this direction, artificial intelligence (AI) for radiologic imaging holds a prominent role, leveraging unique advantages of radiologic breast images, being used routinely for TNBC diagnosis, staging, and treatment planning, and offering high-resolution whole-tumour visualization, combined with the immense potential of AI to elucidate anatomical and functional properties of tumours that may not be easily perceived by the human eye. In this review, we synthesize the current state-of-the-art radiologic imaging applications of AI in assisting TNBC diagnosis, treatment, and prognostication. Our goal is to provide a comprehensive overview of radiomic and deep learning-based AI developments and their impact on advancing TNBC management over the last decade (2013-2024). For completeness of the review, we start with a brief introduction of AI, radiomics, and deep learning. Next, we focus on clinically relevant AI-based diagnostic, predictive, and prognostic models for radiologic breast images evaluated in TNBC. We conclude with opportunities and future directions for AI towards advancing diagnosis, treatment response predictions, and prognostic evaluations for TNBC.
Collapse
Affiliation(s)
- Kanika Bhalla
- Breast Image Computing Lab, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
| | - Qi Xiao
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
| | - José Marcio Luna
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
- Alvin J. Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
| | - Emily Podany
- Division of Hematology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
- Division of Oncology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
| | - Tabassum Ahmad
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
| | - Foluso O Ademuyiwa
- Alvin J. Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
- Division of Oncology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
| | - Andrew Davis
- Alvin J. Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
- Division of Oncology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
| | - Debbie Lee Bennett
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
- Alvin J. Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
| | - Aimilia Gastounioti
- Breast Image Computing Lab, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
- Alvin J. Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
| |
Collapse
|
13
|
Li N, Gong W, Xie Y, Sheng L. Correlation between the CEM imaging characteristics and different molecular subtypes of breast cancer. Breast 2023; 72:103595. [PMID: 37925875 PMCID: PMC10661457 DOI: 10.1016/j.breast.2023.103595] [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: 06/28/2023] [Revised: 09/09/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023] Open
Abstract
PURPOSE To investigate the correlation between the contrast-enhanced mammography (CEM) imaging characteristics and different molecular subtypes of breast cancer (BC). METHODS We retrospectively included 313 eligible female patients who underwent CEM examination and surgery in our hospital from July 2017 to July 2021. Their lesions were confirmed on histopathological examination and immunohistochemical analysis. BC was divided into luminal A, luminal B, HER2-enriched, and triple-negative BC (TNBC) subtypes according to immunohistochemical markers. Nine features were extracted from CEM images, including tumor shape, margins, spiculated mass, lobulated mass, malignant calcification, lesion conspicuity, internal enhancement pattern, multifocal mass, and swollen axillary lymph nodes. Statistical analysis was performed using SPSS 25.0. Univariate analysis and binomial regression were used to analyze the correlation between CEM imaging features and BC molecular subtypes. RESULTS There were 184 (58.8 %) Luminal A, 44 (14.1 %) Luminal B, 47 (15.0 %) HER-2-enriched and 38 (12.1 %) TNBC, respectively. Molecular subtypes were significantly related to the tumor shape, margins, spiculated mass, internal enhancement pattern, malignant calcification and swollen axillary lymph nodes. Spiculated and calcified tumors were associated with Luminal subtypes, especially Luminal B (P < 0.05). Irregular tumor shape and malignant calcification were associated with HER-2-enriched subtype (P < 0.05). Oval or round tumor shape, rim enhancement, and swollen axillary lymph nodes were associated with TNBC (P < 0.05). CONCLUSION CEM imaging features could distinguish BC molecular subtypes. In particular, TNBC showed oval or round tumor shape, rim enhancement, and swollen axillary lymph nodes, providing insights into the diagnosis and prognosis of TNBC.
Collapse
Affiliation(s)
- Na Li
- Department of Medical Imaging, Affiliated Hospital of Jining Medical University, Jining, 272000, China.
| | - Weiyun Gong
- Clinic Imaging Center, The Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
| | - Yuanzhong Xie
- Clinic Imaging Center, The Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
| | - Lei Sheng
- Clinic Imaging Center, The Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China.
| |
Collapse
|
14
|
Petrillo A, Fusco R, Barretta ML, Granata V, Mattace Raso M, Porto A, Sorgente E, Fanizzi A, Massafra R, Lafranceschina M, La Forgia D, Trombadori CML, Belli P, Trecate G, Tenconi C, De Santis MC, Greco L, Ferranti FR, De Soccio V, Vidiri A, Botta F, Dominelli V, Cassano E, Boldrini L. Radiomics and artificial intelligence analysis by T2-weighted imaging and dynamic contrast-enhanced magnetic resonance imaging to predict Breast Cancer Histological Outcome. LA RADIOLOGIA MEDICA 2023; 128:1347-1371. [PMID: 37801198 DOI: 10.1007/s11547-023-01718-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/01/2023] [Indexed: 10/07/2023]
Abstract
OBJECTIVE The objective of the study was to evaluate the accuracy of radiomics features obtained by MR images to predict Breast Cancer Histological Outcome. METHODS A total of 217 patients with malignant lesions were analysed underwent MRI examinations. Considering histological findings as the ground truth, four different types of findings were used in both univariate and multivariate analyses: (1) G1 + G2 vs G3 classification; (2) presence of human epidermal growth factor receptor 2 (HER2 + vs HER2 -); (3) presence of the hormone receptor (HR + vs HR -); and (4) presence of luminal subtypes of breast cancer. RESULTS The best accuracy for discriminating HER2 + versus HER2 - breast cancers was obtained considering nine predictors by early phase T1-weighted subtraction images and a decision tree (accuracy of 88% on validation set). The best accuracy for discriminating HR + versus HR - breast cancers was obtained considering nine predictors by T2-weighted subtraction images and a decision tree (accuracy of 90% on validation set). The best accuracy for discriminating G1 + G2 versus G3 breast cancers was obtained considering 16 predictors by early phase T1-weighted subtraction images in a linear regression model with an accuracy of 75%. The best accuracy for discriminating luminal versus non-luminal breast cancers was obtained considering 27 predictors by early phase T1-weighted subtraction images and a decision tree (accuracy of 94% on validation set). CONCLUSIONS The combination of radiomics analysis and artificial intelligence techniques could be used to support physician decision-making in prediction of Breast Cancer Histological Outcome.
Collapse
Affiliation(s)
- Antonella Petrillo
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy.
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 80013, Naples, Italy
| | - Maria Luisa Barretta
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Vincenza Granata
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Mauro Mattace Raso
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Annamaria Porto
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Eugenio Sorgente
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Annarita Fanizzi
- Direzione Scientifica-IRCCS, Istituto Tumori Giovanni Paolo II-Via Orazio Flacco 65, 70124, Bari, Italy
| | - Raffaella Massafra
- SSD Fisica Sanitaria-IRCCS Istituto Tumori Giovanni Paolo II-Via Orazio Flacco 65, 70124, Bari, Italy
| | - Miria Lafranceschina
- Struttura Semplice Dipartimentale di Radiodiagnostica Senologica-IRCCS Istituto Tumori Giovanni Paolo II-Via Orazio Flacco 65, 70124, Bari, Italy
| | - Daniele La Forgia
- Struttura Semplice Dipartimentale di Radiodiagnostica Senologica-IRCCS Istituto Tumori Giovanni Paolo II-Via Orazio Flacco 65, 70124, Bari, Italy
| | | | - Paolo Belli
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Giovanna Trecate
- Department of Radiodiagnostic and Magnetic Resonance, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Chiara Tenconi
- Department of Medical Physics, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Maria Carmen De Santis
- De Santis Radiation Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Laura Greco
- Radiology and Diagnostic Imaging, Istituto di Ricovero E Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Romana Ferranti
- Radiology and Diagnostic Imaging, Istituto di Ricovero E Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Valeria De Soccio
- Radiology and Diagnostic Imaging, Istituto di Ricovero E Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging, Istituto di Ricovero E Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Botta
- Breast Imaging Division, IEO Istituto Europeo di Oncologia, 20141, Milan, Italy
| | - Valeria Dominelli
- Breast Imaging Division, IEO Istituto Europeo di Oncologia, 20141, Milan, Italy
| | - Enrico Cassano
- Breast Imaging Division, IEO Istituto Europeo di Oncologia, 20141, Milan, Italy
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| |
Collapse
|
15
|
Zhang J, Wu J, Zhou XS, Shi F, Shen D. Recent advancements in artificial intelligence for breast cancer: Image augmentation, segmentation, diagnosis, and prognosis approaches. Semin Cancer Biol 2023; 96:11-25. [PMID: 37704183 DOI: 10.1016/j.semcancer.2023.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/03/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Breast cancer is a significant global health burden, with increasing morbidity and mortality worldwide. Early screening and accurate diagnosis are crucial for improving prognosis. Radiographic imaging modalities such as digital mammography (DM), digital breast tomosynthesis (DBT), magnetic resonance imaging (MRI), ultrasound (US), and nuclear medicine techniques, are commonly used for breast cancer assessment. And histopathology (HP) serves as the gold standard for confirming malignancy. Artificial intelligence (AI) technologies show great potential for quantitative representation of medical images to effectively assist in segmentation, diagnosis, and prognosis of breast cancer. In this review, we overview the recent advancements of AI technologies for breast cancer, including 1) improving image quality by data augmentation, 2) fast detection and segmentation of breast lesions and diagnosis of malignancy, 3) biological characterization of the cancer such as staging and subtyping by AI-based classification technologies, 4) prediction of clinical outcomes such as metastasis, treatment response, and survival by integrating multi-omics data. Then, we then summarize large-scale databases available to help train robust, generalizable, and reproducible deep learning models. Furthermore, we conclude the challenges faced by AI in real-world applications, including data curating, model interpretability, and practice regulations. Besides, we expect that clinical implementation of AI will provide important guidance for the patient-tailored management.
Collapse
Affiliation(s)
- Jiadong Zhang
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China
| | - Jiaojiao Wu
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Xiang Sean Zhou
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Feng Shi
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China.
| | - Dinggang Shen
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China; Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China; Shanghai Clinical Research and Trial Center, Shanghai, China.
| |
Collapse
|
16
|
Yao X, Ao W, Zhu X, Tian S, Han X, Hu J, Xu W, Mao G, Deng S. A novel radiomics based on multi-parametric magnetic resonance imaging for predicting Ki-67 expression in rectal cancer: a multicenter study. BMC Med Imaging 2023; 23:168. [PMID: 37891502 PMCID: PMC10612175 DOI: 10.1186/s12880-023-01123-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND To explore the value of multiparametric MRI markers for preoperative prediction of Ki-67 expression among patients with rectal cancer. METHODS Data from 259 patients with postoperative pathological confirmation of rectal adenocarcinoma who had received enhanced MRI and Ki-67 detection was divided into 4 cohorts: training (139 cases), internal validation (in-valid, 60 cases), and external validation (ex-valid, 60 cases) cohorts. The patients were divided into low and high Ki-67 expression groups. In the training cohort, DWI, T2WI, and contrast enhancement T1WI (CE-T1) sequence radiomics features were extracted from MRI images. Radiomics marker scores and regression coefficient were then calculated for data fitting to construct a radscore model. Subsequently, clinical features with statistical significance were selected to construct a combined model for preoperative individualized prediction of rectal cancer Ki-67 expression. The models were internally and externally validated, and the AUC of each model was calculated. Calibration and decision curves were used to evaluate the clinical practicality of nomograms. RESULTS Three models for predicting rectal cancer Ki-67 expression were constructed. The AUC and Delong test results revealed that the combined model had better prediction performance than other models in three chohrts. A decision curve analysis revealed that the nomogram based on the combined model had relatively good clinical performance, which can be an intuitive prediction tool for clinicians. CONCLUSION The multiparametric MRI radiomics model can provide a noninvasive and accurate auxiliary tool for preoperative evaluation of Ki-67 expression in patients with rectal cancer and can support clinical decision-making.
Collapse
Affiliation(s)
- Xiuzhen Yao
- Department of Ultrasound, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weiqun Ao
- Department of Radiology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Hangzhou, Zhejiang Province, 310012, China
| | - Xiandi Zhu
- Department of Radiology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Hangzhou, Zhejiang Province, 310012, China
| | - Shuyuan Tian
- Department of Ultrasound, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Xiaoyu Han
- Department of Pathology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Jinwen Hu
- Department of Radiology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenjie Xu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Guoqun Mao
- Department of Radiology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Hangzhou, Zhejiang Province, 310012, China.
| | - Shuitang Deng
- Department of Radiology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Hangzhou, Zhejiang Province, 310012, China.
| |
Collapse
|
17
|
Ma X, Pierce E, Anand H, Aviles N, Kunk P, Alemazkoor N. Early prediction of response to palliative chemotherapy in patients with stage-IV gastric and esophageal cancer. BMC Cancer 2023; 23:910. [PMID: 37759332 PMCID: PMC10536729 DOI: 10.1186/s12885-023-11422-z] [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: 04/17/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The goal of therapy for many patients with advanced stage malignancies, including those with metastatic gastric and esophageal cancers, is to extend overall survival while also maintaining quality of life. After weighing the risks and benefits of treatment with palliative chemotherapy (PC) with non-curative intent, many patients decide to pursue treatment. It is known that a subset of patients who are treated with PC experience significant side effects without clinically significant survival benefits from PC. METHODS We use data from 150 patients with stage-IV gastric and esophageal cancers to train machine learning models that predict whether a patient with stage-IV gastric or esophageal cancers would benefit from PC, in terms of increased survival duration, at very early stages of the treatment. RESULTS Our findings show that machine learning can predict with high accuracy whether a patient will benefit from PC at the time of diagnosis. More accurate predictions can be obtained after only two cycles of PC (i.e., about 4 weeks after diagnosis). The results from this study are promising with regard to potential improvements in quality of life for patients near the end of life and a potential overall survival benefit by optimizing systemic therapy earlier in the treatment course of patients.
Collapse
Affiliation(s)
- Xiaoyuan Ma
- Department of Statistics, University of Virginia, Charlottesville, USA
| | - Eric Pierce
- School of Medicine, University of Virginia, Charlottesville, USA
| | - Harsh Anand
- System and Information Engineering, University of Virginia, Charlottesville, USA
| | - Natalie Aviles
- Department of Sociology, University of Virginia, Charlottesville, USA
| | - Paul Kunk
- School of Medicine, University of Virginia, Charlottesville, USA
| | - Negin Alemazkoor
- System and Information Engineering, University of Virginia, Charlottesville, USA.
| |
Collapse
|
18
|
Siviengphanom S, Gandomkar Z, Lewis SJ, Brennan PC. Global Radiomic Features from Mammography for Predicting Difficult-To-Interpret Normal Cases. J Digit Imaging 2023; 36:1541-1552. [PMID: 37253894 PMCID: PMC10406750 DOI: 10.1007/s10278-023-00836-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 06/01/2023] Open
Abstract
This work aimed to investigate whether global radiomic features (GRFs) from mammograms can predict difficult-to-interpret normal cases (NCs). Assessments from 537 readers interpreting 239 normal mammograms were used to categorise cases as 120 difficult-to-interpret and 119 easy-to-interpret based on cases having the highest and lowest difficulty scores, respectively. Using lattice- and squared-based approaches, 34 handcrafted GRFs per image were extracted and normalised. Three classifiers were constructed: (i) CC and (ii) MLO using the GRFs from corresponding craniocaudal and mediolateral oblique images only, based on the random forest technique for distinguishing difficult- from easy-to-interpret NCs, and (iii) CC + MLO using the median predictive scores from both CC and MLO models. Useful GRFs for the CC and MLO models were recognised using a scree test. The CC and MLO models were trained and validated using the leave-one-out-cross-validation. The models' performances were assessed by the AUC and compared using the DeLong test. A Kruskal-Wallis test was used to examine if the 34 GRFs differed between difficult- and easy-to-interpret NCs and if difficulty level based on the traditional breast density (BD) categories differed among 115 low-BD and 124 high-BD NCs. The CC + MLO model achieved higher performance (0.71 AUC) than the individual CC and MLO model alone (0.66 each), but statistically non-significant difference was found (all p > 0.05). Six GRFs were identified to be valuable in describing difficult-to-interpret NCs. Twenty features, when compared between difficult- and easy-to-interpret NCs, differed significantly (p < 0.05). No statistically significant difference was observed in difficulty between low- and high-BD NCs (p = 0.709). GRF mammographic analysis can predict difficult-to-interpret NCs.
Collapse
Affiliation(s)
- Somphone Siviengphanom
- Medical Image Optimisation and Perception Group, Discipline of Medical Imaging Science, Sydney School of Health Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW, 2006, Australia.
| | - Ziba Gandomkar
- Medical Image Optimisation and Perception Group, Discipline of Medical Imaging Science, Sydney School of Health Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW, 2006, Australia
| | - Sarah J Lewis
- Medical Image Optimisation and Perception Group, Discipline of Medical Imaging Science, Sydney School of Health Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW, 2006, Australia
| | - Patrick C Brennan
- Medical Image Optimisation and Perception Group, Discipline of Medical Imaging Science, Sydney School of Health Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
19
|
Szep M, Pintican R, Boca B, Perja A, Duma M, Feier D, Epure F, Fetica B, Eniu D, Roman A, Dudea SM, Chiorean A. Whole-Tumor ADC Texture Analysis Is Able to Predict Breast Cancer Receptor Status. Diagnostics (Basel) 2023; 13:diagnostics13081414. [PMID: 37189515 DOI: 10.3390/diagnostics13081414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
There are different breast cancer molecular subtypes with differences in incidence, treatment response and outcome. They are roughly divided into estrogen and progesterone receptor (ER and PR) negative and positive cancers. In this retrospective study, we included 185 patients augmented with 25 SMOTE patients and divided them into two groups: the training group consisted of 150 patients and the validation cohort consisted of 60 patients. Tumors were manually delineated and whole-volume tumor segmentation was used to extract first-order radiomic features. The ADC-based radiomics model reached an AUC of 0.81 in the training cohort and was confirmed in the validation set, which yielded an AUC of 0.93, in differentiating ER/PR positive from ER/PR negative status. We also tested a combined model using radiomics data together with ki67% proliferation index and histological grade, and obtained a higher AUC of 0.93, which was also confirmed in the validation group. In conclusion, whole-volume ADC texture analysis is able to predict hormonal status in breast cancer masses.
Collapse
Affiliation(s)
- Madalina Szep
- Department of Radiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Roxana Pintican
- Department of Radiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Bianca Boca
- Department of Medical Imaging, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Andra Perja
- Department of Radiology and Medical Imaging, County Clinical Emergency Hospital, 400347 Cluj-Napoca, Romania
| | | | - Diana Feier
- Department of Radiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- Medimages Breast Center, 400462 Cluj-Napoca, Romania
| | - Flavia Epure
- Medical Imaging Department, Medisprof Cancer Center, 400641 Cluj Napoca, Romania
| | - Bogdan Fetica
- Department of Pathology, "Ion Chiricuţă" Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Dan Eniu
- Department of Surgical Oncology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Andrei Roman
- Department of Radiology, "Ion Chiricuță" Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Sorin Marian Dudea
- Department of Radiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | | |
Collapse
|
20
|
Miller MM, Rubaiyat AHM, Rohde GK. Predicting Malignancy of Breast Imaging Findings Using Quantitative Analysis of Contrast-Enhanced Mammography (CEM). Diagnostics (Basel) 2023; 13:diagnostics13061129. [PMID: 36980437 PMCID: PMC10047016 DOI: 10.3390/diagnostics13061129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
We sought to develop new quantitative approaches to characterize the spatial distribution of mammographic density and contrast enhancement of suspicious contrast-enhanced mammography (CEM) findings to improve malignant vs. benign classifications of breast lesions. We retrospectively analyzed all breast lesions that underwent CEM imaging and tissue sampling at our institution from 2014–2020 in this IRB-approved study. A penalized linear discriminant analysis was used to classify lesions based on the averaged histograms of radial distributions of mammographic density and contrast enhancement. T-tests were used to compare the classification accuracies of density, contrast, and concatenated density and contrast histograms. Logistic regression and AUC-ROC analyses were used to assess if adding demographic and clinical data improved the model accuracy. A total of 159 suspicious findings were evaluated. Density histograms were more accurate in classifying lesions as malignant or benign than a random classifier (62.37% vs. 48%; p < 0.001), but the concatenated density and contrast histograms demonstrated a higher accuracy (71.25%; p < 0.001) than the density histograms alone. Including the demographic and clinical data in our models led to a higher AUC-ROC than concatenated density and contrast images (0.81 vs. 0.70; p < 0.001). In the classification of invasive vs. non-invasive malignancy, the concatenated density and contrast histograms demonstrated no significant improvement in accuracy over the density histograms alone (77.63% vs. 78.59%; p = 0.504). Our findings suggest that quantitative differences in the radial distribution of mammographic density could be used to discriminate malignant from benign breast findings; however, classification accuracy was significantly improved with the addition of contrast-enhanced imaging data from CEM. Adding patient demographic and clinical information further improved the classification accuracy.
Collapse
Affiliation(s)
- Matthew M. Miller
- Department of Radiology and Medical Imaging, University of Virginia Health System, 1215 Lee St., Charlottesville, VA 22903, USA
- Correspondence:
| | - Abu Hasnat Mohammad Rubaiyat
- Department of Electrical and Computer Engineering, University of Virginia, 415 Lane Rd., Charlottesville, VA 22903, USA
| | - Gustavo K. Rohde
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd., Charlottesville, VA 22903, USA
| |
Collapse
|
21
|
Zheng D, He X, Jing J. Overview of Artificial Intelligence in Breast Cancer Medical Imaging. J Clin Med 2023; 12:419. [PMID: 36675348 PMCID: PMC9864608 DOI: 10.3390/jcm12020419] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/07/2023] Open
Abstract
The heavy global burden and mortality of breast cancer emphasize the importance of early diagnosis and treatment. Imaging detection is one of the main tools used in clinical practice for screening, diagnosis, and treatment efficacy evaluation, and can visualize changes in tumor size and texture before and after treatment. The overwhelming number of images, which lead to a heavy workload for radiologists and a sluggish reporting period, suggests the need for computer-aid detection techniques and platform. In addition, complex and changeable image features, heterogeneous quality of images, and inconsistent interpretation by different radiologists and medical institutions constitute the primary difficulties in breast cancer screening and imaging diagnosis. The advancement of imaging-based artificial intelligence (AI)-assisted tumor diagnosis is an ideal strategy for improving imaging diagnosis efficient and accuracy. By learning from image data input and constructing algorithm models, AI is able to recognize, segment, and diagnose tumor lesion automatically, showing promising application prospects. Furthermore, the rapid advancement of "omics" promotes a deeper and more comprehensive recognition of the nature of cancer. The fascinating relationship between tumor image and molecular characteristics has attracted attention to the radiomic and radiogenomics, which allow us to perform analysis and detection on the molecular level with no need for invasive operations. In this review, we integrate the current developments in AI-assisted imaging diagnosis and discuss the advances of AI-based breast cancer precise diagnosis from a clinical point of view. Although AI-assisted imaging breast cancer screening and detection is an emerging field and draws much attention, the clinical application of AI in tumor lesion recognition, segmentation, and diagnosis is still limited to research or in limited patients' cohort. Randomized clinical trials based on large and high-quality cohort are lacking. This review aims to describe the progress of the imaging-based AI application in breast cancer screening and diagnosis for clinicians.
Collapse
Affiliation(s)
| | | | - Jing Jing
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| |
Collapse
|
22
|
Nicosia L, Bozzini AC, Ballerini D, Palma S, Pesapane F, Raimondi S, Gaeta A, Bellerba F, Origgi D, De Marco P, Castiglione Minischetti G, Sangalli C, Meneghetti L, Curigliano G, Cassano E. Radiomic Features Applied to Contrast Enhancement Spectral Mammography: Possibility to Predict Breast Cancer Molecular Subtypes in a Non-Invasive Manner. Int J Mol Sci 2022; 23:ijms232315322. [PMID: 36499648 PMCID: PMC9740943 DOI: 10.3390/ijms232315322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
We aimed to investigate the association between the radiomic features of contrast-enhanced spectral mammography (CESM) images and a specific receptor pattern of breast neoplasms. In this single-center retrospective study, we selected patients with neoplastic breast lesions who underwent CESM before a biopsy and surgical assessment between January 2013 and February 2022. Radiomic analysis was performed on regions of interest selected from recombined CESM images. The association between the features and each evaluated endpoint (ER, PR, Ki-67, HER2+, triple negative, G2-G3 expressions) was investigated through univariate logistic regression. Among the significant and highly correlated radiomic features, we selected only the one most associated with the endpoint. From a group of 321 patients, we enrolled 205 malignant breast lesions. The median age at the exam was 50 years (interquartile range (IQR) 45-58). NGLDM_Contrast was the only feature that was positively associated with both ER and PR expression (p-values = 0.01). NGLDM_Coarseness was negatively associated with Ki-67 expression (p-value = 0.02). Five features SHAPE Volume(mL), SHAPE_Volume(vx), GLRLM_RLNU, NGLDM_Busyness and GLZLM_GLNU were all positively and significantly associated with HER2+; however, all of them were highly correlated. Radiomic features of CESM images could be helpful to predict particular molecular subtypes before a biopsy.
Collapse
Affiliation(s)
- Luca Nicosia
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
- Correspondence:
| | - Anna Carla Bozzini
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Daniela Ballerini
- Breast Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy
| | - Simone Palma
- Department of Radiological and Hematological Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Filippo Pesapane
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Sara Raimondi
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO IRCCS, 20139 Milan, Italy
| | - Aurora Gaeta
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO IRCCS, 20139 Milan, Italy
| | - Federica Bellerba
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO IRCCS, 20139 Milan, Italy
| | - Daniela Origgi
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Paolo De Marco
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Giuseppe Castiglione Minischetti
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- School of Medical Physics, University of Milan, Via Celoria 16, 20133 Milan, Italy
| | - Claudia Sangalli
- Data Management, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lorenza Meneghetti
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milano, 20122 Milano, Italy
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Enrico Cassano
- Breast Imaging Division, Radiology Department, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| |
Collapse
|
23
|
Multiregional Radiomic Signatures Based on Functional Parametric Maps from DCE-MRI for Preoperative Identification of Estrogen Receptor and Progesterone Receptor Status in Breast Cancer. Diagnostics (Basel) 2022; 12:diagnostics12102558. [PMID: 36292247 PMCID: PMC9601361 DOI: 10.3390/diagnostics12102558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/01/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Radiomics based on dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) has been used for breast estrogen receptor (ER) and progesterone receptor (PR) status evaluation. However, the radiomic features of peritumoral regions were not thoroughly analyzed. This study aimed to establish and validate the multiregional radiomic signatures (RSs) for the preoperative identification of the ER and PR status in breast cancer. A total of 443 patients with breast cancer were divided into training (n = 356) and validation (n = 87) sets. Radiomic features were extracted from intra- and peritumoral regions on six functional parametric maps from DCE-MRI. A two-sample t-test, least absolute shrinkage and selection operator regression, and stepwise were used for feature selections. Three RSs for predicting the ER and PR status were constructed using a logistic regression model based on selected intratumoral, peritumoral, and combined intra- and peritumoral radiomic features. The area under the receiver operator characteristic curve (AUC) was used to assess the discriminative performance of three RSs. The AUCs of intra- and peritumoral RSs for identifying the ER status were 0.828/0.791 and 0.755/0.733 in the training and validation sets, respectively. For predicting the PR status, intra- and peritumoral RSs resulted in AUCs of 0.816/0.749 and 0.806/0.708 in the training and validation sets, respectively. Multiregional RSs achieved the best AUCs among three RSs for evaluating the ER (0.851 and 0.833) and PR (0.848 and 0.763) status. In conclusion, multiregional RSs based on functional parametric maps from DCE-MRI showed promising results for preoperatively evaluating the ER and PR status in breast cancer patients. Further studies using a larger cohort from multiple centers are necessary to confirm the reliability of the established models before clinical application.
Collapse
|
24
|
Luczynska E, Piegza T, Szpor J, Heinze S, Popiela T, Kargol J, Rudnicki W. Contrast-Enhanced Mammography (CEM) Capability to Distinguish Molecular Breast Cancer Subtypes. Biomedicines 2022; 10:2384. [PMID: 36289645 PMCID: PMC9598186 DOI: 10.3390/biomedicines10102384] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/14/2022] [Accepted: 09/22/2022] [Indexed: 11/20/2022] Open
Abstract
With breast cancer ranking first among the most common malignant neoplasms in the world, new techniques of early detection are in even more demand than before. Our awareness of tumors' biology is expanding and may be used to treat patients more efficiently. A link between radiology and pathology was searched for in our study, as well as the answer to the question of whether a tumor type can be seen on contrast-enhanced mammography and if such knowledge may serve as part of precision medicine.
Collapse
Affiliation(s)
- Elzbieta Luczynska
- Department of Electroradiology, Jagiellonian University Medical College, 31-008 Cracow, Poland
| | - Tomasz Piegza
- Department of Radiology, 5th Military Clinical Hospital in Cracow, 30-901 Cracow, Poland
| | - Joanna Szpor
- Department of Pathomorphology, Jagiellonian University Medical College, 30-688 Cracow, Poland
| | - Sylwia Heinze
- Department of Radiology, Maria Sklodowska-Curie National Research Institute of Oncology in Cracow, 31-115 Cracow, Poland
| | - Tadeusz Popiela
- Department of Radiology, Jagiellonian University Medical College, 30-688 Cracow, Poland
| | - Jaromir Kargol
- Institute of Medical Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Wojciech Rudnicki
- Department of Electroradiology, Jagiellonian University Medical College, 31-008 Cracow, Poland
| |
Collapse
|
25
|
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: 5] [Impact Index Per Article: 1.7] [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
|
26
|
Radiomic Signatures Derived from Hybrid Contrast-Enhanced Ultrasound Images (CEUS) for the Assessment of Histological Characteristics of Breast Cancer: A Pilot Study. Cancers (Basel) 2022; 14:cancers14163905. [PMID: 36010897 PMCID: PMC9405598 DOI: 10.3390/cancers14163905] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 12/30/2022] Open
Abstract
The purpose of this study was to evaluate the diagnostic performance of radiomic features extracted from standardized hybrid contrast-enhanced ultrasound (CEUS) data for the assessment of hormone receptor status, human epidermal growth factor receptor 2 (HER2) status, tumor grade and Ki-67 in patients with primary breast cancer. METHODS This prospective study included 72 patients with biopsy-proven breast cancer who underwent CEUS examinations between October 2020 and September 2021. RESULTS A radiomic analysis found the WavEnHH_s_4 parameter as an independent predictor associated with the HER2+ status with 76.92% sensitivity, and 64.41% specificity and a prediction model that could differentiate between the HER2 entities with 76.92% sensitivity and 84.75% specificity. The RWavEnLH_s-4 parameter was an independent predictor for estrogen receptor (ER) status with 55.93% sensitivity and 84.62% specificity, while a prediction model (RPerc01, RPerc10 and RWavEnLH_s_4) could differentiate between the progesterone receptor (PR) status with 44.74% sensitivity and 88.24% specificity. No texture parameter showed statistically significant results at the univariate analysis when comparing the Nottingham grade and the Ki-67 status. CONCLUSION Our preliminary data indicate a potential that hybrid CEUS radiomic features allow the discrimination between breast cancers of different receptor and HER2 statuses with high specificity. Hybrid CEUS radiomic features might have the potential to provide a noninvasive, easily accessible and contrast-agent-safe method to assess tumor biology before and during treatment.
Collapse
|
27
|
Xu A, Chu X, Zhang S, Zheng J, Shi D, Lv S, Li F, Weng X. Development and validation of a clinicoradiomic nomogram to assess the HER2 status of patients with invasive ductal carcinoma. BMC Cancer 2022; 22:872. [PMID: 35945526 PMCID: PMC9364617 DOI: 10.1186/s12885-022-09967-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Background The determination of HER2 expression status contributes significantly to HER2-targeted therapy in breast carcinoma. However, an economical, efficient, and non-invasive assessment of HER2 is lacking. We aimed to develop a clinicoradiomic nomogram based on radiomics scores extracted from multiparametric MRI (mpMRI, including ADC-map, T2W1, DCE-T1WI) and clinical risk factors to assess HER2 status. Methods We retrospectively collected 214 patients with pathologically confirmed invasive ductal carcinoma between January 2018 to March 2021 from Fudan University Shanghai Cancer Center, and randomly divided this cohort into training set (n = 128, 42 HER2-positive and 86 HER2-negative cases) and validation set (n = 86, 28 HER2-positive and 58 HER2-negative cases) at a ratio of 6:4. The original and transformed pretherapy mpMRI images were treated by semi-automated segmentation and manual modification on the DeepWise scientific research platform v1.6 (http://keyan.deepwise.com/), then radiomics feature extraction was implemented with PyRadiomics library. Recursive feature elimination (RFE) based on logistic regression (LR) and LASSO regression were adpoted to identify optimal features before modeling. LR, Linear Discriminant Analysis (LDA), support vector machine (SVM), random forest (RF), naive Bayesian (NB) and XGBoost (XGB) algorithms were used to construct the radiomics signatures. Independent clinical predictors were identified through univariate logistic analysis (age, tumor location, ki-67 index, histological grade, and lymph node metastasis). Then, the radiomics signature with the best diagnostic performance (Rad score) was further combined with significant clinical risk factors to develop a clinicoradiomic model (nomogram) using multivariate logistic regression. The discriminative power of the constructed models were evaluated by AUC, DeLong test, calibration curve, and decision curve analysis (DCA). Results 70 (32.71%) of the enrolled 214 cases were HER2-positive, while 144 (67.29%) were HER2-negative. Eleven best radiomics features were retained to develop 6 radiomcis classifiers in which RF classifier showed the highest AUC of 0.887 (95%CI: 0.827–0.947) in the training set and acheived the AUC of 0.840 (95%CI: 0.758–0.922) in the validation set. A nomogram that incorporated the Rad score with two selected clinical factors (Ki-67 index and histological grade) was constructed and yielded better discrimination compared with Rad score (p = 0.374, Delong test), with an AUC of 0.945 (95%CI: 0.904–0.987) in the training set and 0.868 (95%CI: 0.789–0.948; p = 0.123) in the validation set. Moreover, calibration with the p-value of 0.732 using Hosmer–Lemeshow test demonstrated good agreement, and the DCA verified the benefits of the nomogram. Conclusion Post largescale validation, the clinicoradiomic nomogram may have the potential to be used as a non-invasive tool for determination of HER2 expression status in clinical HER2-targeted therapy prediction. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09967-6.
Collapse
Affiliation(s)
- Aqiao Xu
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, 312030, China.
| | - Xiufeng Chu
- Department of Surgical, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, 312030, China
| | - Shengjian Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jing Zheng
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, 312030, China
| | - Dabao Shi
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, 312030, China
| | - Shasha Lv
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, 312030, China
| | - Feng Li
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd, Beijing, 100080, P.R. China
| | - Xiaobo Weng
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, 312030, China.
| |
Collapse
|
28
|
Vasselli F, Fabi A, Ferranti FR, Barba M, Botti C, Vidiri A, Tommasin S. How Dual-Energy Contrast-Enhanced Spectral Mammography Can Provide Useful Clinical Information About Prognostic Factors in Breast Cancer Patients: A Systematic Review of Literature. Front Oncol 2022; 12:859838. [PMID: 35941874 PMCID: PMC9355886 DOI: 10.3389/fonc.2022.859838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/27/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction In the past decade, a new technique derived from full-field digital mammography has been developed, named contrast-enhanced spectral mammography (CESM). The aim of this study was to define the association between CESM findings and usual prognostic factors, such as estrogen receptors, progesterone receptors, HER2, and Ki67, in order to offer an updated overview of the state of the art for the early differential diagnosis of breast cancer and following personalized treatments. Materials and Methods According to the PRISMA guidelines, two electronic databases (PubMed and Scopus) were investigated, using the following keywords: breast cancer AND (CESM OR contrast enhanced spectral mammography OR contrast enhanced dual energy mammography) AND (receptors OR prognostic factors OR HER2 OR progesterone OR estrogen OR Ki67). The search was concluded in August 2021. No restriction was applied to publication dates. Results We obtained 28 articles from the research in PubMed and 114 articles from Scopus. After the removal of six replicas that were counted only once, out of 136 articles, 37 articles were reviews. Eight articles alone have tackled the relation between CESM imaging and ER, PR, HER2, and Ki67. When comparing radiological characterization of the lesions obtained by either CESM or contrast-enhanced MRI, they have a similar association with the proliferation of tumoral cells, as expressed by Ki-67. In CESM-enhanced lesions, the expression was found to be 100% for ER and 77.4% for PR, while moderate or high HER2 positivity was found in lesions with non-mass enhancement and with mass closely associated with a non-mass enhancement component. Conversely, the non-enhancing breast cancer lesions were not associated with any prognostic factor, such as ER, PR, HER2, and Ki67, which may be associated with the probability of showing enhancement. Radiomics on CESM images has the potential for non-invasive characterization of potentially heterogeneous tumors with different hormone receptor status. Conclusions CESM enhancement is associated with the proliferation of tumoral cells, as well as to the expression of estrogen and progesterone receptors. As CESM is a relatively young imaging technique, a few related works were found; this may be due to the “off-label” modality. In the next few years, the role of CESM in breast cancer diagnostics will be more thoroughly investigated.
Collapse
Affiliation(s)
- Federica Vasselli
- Radiology and Diagnostic Imaging, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Alessandra Fabi
- Precision Medicine in Breast Cancer Unit, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Francesca Romana Ferranti
- Radiology and Diagnostic Imaging, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology 2, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Claudio Botti
- Division of Breast Surgery, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
- *Correspondence: Antonello Vidiri,
| | - Silvia Tommasin
- Human Neuroscience Department, Sapienza University of Rome, Rome, Italy
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
29
|
Prediction of Breast Cancer Histological Outcome by Radiomics and Artificial Intelligence Analysis in Contrast-Enhanced Mammography. Cancers (Basel) 2022; 14:cancers14092132. [PMID: 35565261 PMCID: PMC9102628 DOI: 10.3390/cancers14092132] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/30/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The assessment of breast lesions through mammographic images is currently challenging, especially in dense breasts. Contrast-enhanced mammography has been shown to overcome the limitations of standard mammography but it greatly depends on the interpretative skills of the physician. The aim of this study was to evaluate the potentialities of statistical and artificial intelligence algorithms as a tool for helping the radiologists in the interpretation of images. The most remarkable results were achieved in discriminating benign from malignant lesions and in the identification of the presence of the hormone receptor. A tool to support the physician’s decision-making process may be designed starting from simple logistic regression and tree-based algorithms. This type of tool may help the radiologist in assessing the investigated breast and in choosing the appropriate follow-up without resorting to histology. Abstract Purpose: To evaluate radiomics features in order to: differentiate malignant versus benign lesions; predict low versus moderate and high grading; identify positive or negative hormone receptors; and discriminate positive versus negative human epidermal growth factor receptor 2 related to breast cancer. Methods: A total of 182 patients with known breast lesions and that underwent Contrast-Enhanced Mammography were enrolled in this retrospective study. The reference standard was pathology (118 malignant lesions and 64 benign lesions). A total of 837 textural metrics were extracted by manually segmenting the region of interest from both craniocaudally (CC) and mediolateral oblique (MLO) views. Non-parametric Wilcoxon–Mann–Whitney test, receiver operating characteristic, logistic regression and tree-based machine learning algorithms were used. The Adaptive Synthetic Sampling balancing approach was used and a feature selection process was implemented. Results: In univariate analysis, the classification of malignant versus benign lesions achieved the best performance when considering the original_gldm_DependenceNonUniformity feature extracted on CC view (accuracy of 88.98%). An accuracy of 83.65% was reached in the classification of grading, whereas a slightly lower value of accuracy (81.65%) was found in the classification of the presence of the hormone receptor; the features extracted were the original_glrlm_RunEntropy and the original_gldm_DependenceNonUniformity, respectively. The results of multivariate analysis achieved the best performances when using two or more features as predictors for classifying malignant versus benign lesions from CC view images (max test accuracy of 95.83% with a non-regularized logistic regression). Considering the features extracted from MLO view images, the best test accuracy (91.67%) was obtained when predicting the grading using a classification-tree algorithm. Combinations of only two features, extracted from both CC and MLO views, always showed test accuracy values greater than or equal to 90.00%, with the only exception being the prediction of the human epidermal growth factor receptor 2, where the best performance (test accuracy of 89.29%) was obtained with the random forest algorithm. Conclusions: The results confirm that the identification of malignant breast lesions and the differentiation of histological outcomes and some molecular subtypes of tumors (mainly positive hormone receptor tumors) can be obtained with satisfactory accuracy through both univariate and multivariate analysis of textural features extracted from Contrast-Enhanced Mammography images.
Collapse
|
30
|
Massafra R, Catino A, Perrotti PMS, Pizzutilo P, Fanizzi A, Montrone M, Galetta D. Informative Power Evaluation of Clinical Parameters to Predict Initial Therapeutic Response in Patients with Advanced Pleural Mesothelioma: A Machine Learning Approach. J Clin Med 2022; 11:jcm11061659. [PMID: 35329985 PMCID: PMC8950691 DOI: 10.3390/jcm11061659] [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: 02/07/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/10/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare neoplasm whose early diagnosis is challenging and systemic treatments are generally administered as first line in the advanced disease stage. The initial clinical response may represent a useful parameter in terms of identifying patients with a better long-term outcome. In this report, the initial therapeutical response in 46 patients affected with advanced/unresectable pleural mesothelioma was investigated. The initial therapeutic response was assessed by CT scan and clinical examination after 2–3 treatment cycles. Our preliminary evaluation shows that the group of patients treated with regimens including antiangiogenetics and/or immunotherapy had a significantly better initial response as compared to patients only treated with standard chemotherapy, exhibiting a disease control rate (DCR) of 100% (95% IC, 79.40–100%) and 80.0% (95% IC, 61.40–92.30%), respectively. Furthermore, the therapeutic response was correlated with the disease stage, blood leukocytes and neutrophils, high albumin serum levels, and basal body mass index (BMI). Specifically, the patients with disease stage III showed a DCR of 95.7% (95% IC, 78.1–99.9%), whereas for disease stage IV the DCR decreased to 66.7% (95% IC, 34.9–9.1%). Moreover, a better initial response was observed in patients with a higher BMI, who reached a DCR of 96.10% (95% IC, 80.36–99.90%). Furthermore, in order to evaluate in the predictive power of the collected features a multivariate way, we report the preliminary results of a machine learning model for predicting the initial therapeutic response. We trained a state-of-the-art algorithm combined to a sequential forward feature selection procedure. The model reached a median AUC value, accuracy, sensitivity, and specificity of 77.0%, 75%, 74.8%, and 83.3%, respectively. The features with greater informational power were gender, histotype, BMI, smoking habits, packs/year, and disease stage. Our preliminary data support the possible favorable correlation between innovative treatments and therapeutic response in patients with unresectable/advanced pleural mesothelioma. The small sample size does not allow concrete conclusions to be drawn; nevertheless, this work is the basis of an ongoing study that will also involve radiomics in a larger dataset.
Collapse
Affiliation(s)
- 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;
| | - Annamaria Catino
- Struttura Semplice Dipartimentale di Oncologia Medica per la Patologia Toracica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (P.P.); (M.M.); (D.G.)
| | - Pia Maria Soccorsa Perrotti
- Struttura Semplice Dipartimentale di Radiologia, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Pamela Pizzutilo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Patologia Toracica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (P.P.); (M.M.); (D.G.)
| | - 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;
- Correspondence: ; Tel.: +39-080-555-5111
| | - Michele Montrone
- Struttura Semplice Dipartimentale di Oncologia Medica per la Patologia Toracica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (P.P.); (M.M.); (D.G.)
| | - Domenico Galetta
- Struttura Semplice Dipartimentale di Oncologia Medica per la Patologia Toracica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (P.P.); (M.M.); (D.G.)
| |
Collapse
|
31
|
Fusco R, Di Bernardo E, Piccirillo A, Rubulotta MR, Petrosino T, Barretta ML, Mattace Raso M, Vallone P, Raiano C, Di Giacomo R, Siani C, Avino F, Scognamiglio G, Di Bonito M, Granata V, Petrillo A. Radiomic and Artificial Intelligence Analysis with Textural Metrics Extracted by Contrast-Enhanced Mammography and Dynamic Contrast Magnetic Resonance Imaging to Detect Breast Malignant Lesions. Curr Oncol 2022; 29:1947-1966. [PMID: 35323359 PMCID: PMC8947713 DOI: 10.3390/curroncol29030159] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose:The purpose of this study was to discriminate between benign and malignant breast lesions through several classifiers using, as predictors, radiomic metrics extracted from CEM and DCE-MRI images. In order to optimize the analysis, balancing and feature selection procedures were performed. Methods: Fifty-four patients with 79 histo-pathologically proven breast lesions (48 malignant lesions and 31 benign lesions) underwent both CEM and DCE-MRI. The lesions were retrospectively analyzed with radiomic and artificial intelligence approaches. Forty-eight textural metrics were extracted, and univariate and multivariate analyses were performed: non-parametric statistical test, receiver operating characteristic (ROC) and machine learning classifiers. Results: Considering the single metrics extracted from CEM, the best predictors were KURTOSIS (area under ROC curve (AUC) = 0.71) and SKEWNESS (AUC = 0.71) calculated on late MLO view. Considering the features calculated from DCE-MRI, the best predictors were RANGE (AUC = 0.72), ENERGY (AUC = 0.72), ENTROPY (AUC = 0.70) and GLN (gray-level nonuniformity) of the gray-level run-length matrix (AUC = 0.72). Considering the analysis with classifiers and an unbalanced dataset, no significant results were obtained. After the balancing and feature selection procedures, higher values of accuracy, specificity and AUC were reached. The best performance was obtained considering 18 robust features among all metrics derived from CEM and DCE-MRI, using a linear discriminant analysis (accuracy of 0.84 and AUC = 0.88). Conclusions: Classifiers, adjusted with adaptive synthetic sampling and feature selection, allowed for increased diagnostic performance of CEM and DCE-MRI in the differentiation between benign and malignant lesions.
Collapse
Affiliation(s)
- Roberta Fusco
- Medical Oncolody Division, Igea SpA, 80013 Naples, Italy; (R.F.); (E.D.B.)
| | - Elio Di Bernardo
- Medical Oncolody Division, Igea SpA, 80013 Naples, Italy; (R.F.); (E.D.B.)
| | - Adele Piccirillo
- Department of Electrical Engineering and Information Technologies, Università degli Studi di Napoli Federico II, 80125 Naples, Italy;
| | - Maria Rosaria Rubulotta
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (M.R.R.); (T.P.); (M.L.B.); (M.M.R.); (P.V.); (C.R.); (A.P.)
| | - Teresa Petrosino
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (M.R.R.); (T.P.); (M.L.B.); (M.M.R.); (P.V.); (C.R.); (A.P.)
| | - Maria Luisa Barretta
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (M.R.R.); (T.P.); (M.L.B.); (M.M.R.); (P.V.); (C.R.); (A.P.)
| | - Mauro Mattace Raso
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (M.R.R.); (T.P.); (M.L.B.); (M.M.R.); (P.V.); (C.R.); (A.P.)
| | - Paolo Vallone
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (M.R.R.); (T.P.); (M.L.B.); (M.M.R.); (P.V.); (C.R.); (A.P.)
| | - Concetta Raiano
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (M.R.R.); (T.P.); (M.L.B.); (M.M.R.); (P.V.); (C.R.); (A.P.)
| | - Raimondo Di Giacomo
- Senology Surgical Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.G.); (C.S.); (F.A.)
| | - Claudio Siani
- Senology Surgical Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.G.); (C.S.); (F.A.)
| | - Franca Avino
- Senology Surgical Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.G.); (C.S.); (F.A.)
| | - Giosuè Scognamiglio
- Pathology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (G.S.); (M.D.B.)
| | - Maurizio Di Bonito
- Pathology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (G.S.); (M.D.B.)
| | - Vincenza Granata
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (M.R.R.); (T.P.); (M.L.B.); (M.M.R.); (P.V.); (C.R.); (A.P.)
- Correspondence: ; Tel.: +39-081-590-714; Fax: +39-081-590-3825
| | - Antonella Petrillo
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (M.R.R.); (T.P.); (M.L.B.); (M.M.R.); (P.V.); (C.R.); (A.P.)
| |
Collapse
|
32
|
Ferranti FR, Vasselli F, Barba M, Sperati F, Terrenato I, Graziano F, Vici P, Botti C, Vidiri A. Diagnostic Accuracy of Contrast-Enhanced, Spectral Mammography (CESM) and 3T Magnetic Resonance Compared to Full-Field Digital Mammography plus Ultrasound in Breast Lesions: Results of a (Pilot) Open-Label, Single-Centre Prospective Study. Cancers (Basel) 2022; 14:cancers14051351. [PMID: 35267659 PMCID: PMC8909837 DOI: 10.3390/cancers14051351] [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: 02/24/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: To assess the diagnostic accuracy of CESM and 3T MRI compared to full-field digital mammography (FFDM), plus US, in the evaluation of advanced breast lesions. Materials and Methods: Consenting women with suspicious findings underwent FFDM, US, CESM and 3T MRI. Breast lesions were histologically assessed, with histology being the gold standard. Two experienced breast radiologists, blinded to cancer status, read the images. Diagnostic accuracy of (1) CESM as an adjunct to FFDM and US, and (2) 3T MRI as an adjunct to CESM compared to FFDM and US, was assessed. Measures of accuracy were sensitivity (Se), specificity (Sp), positive predictive value (PPV) and negative predictive value (NPV). Results: There were 118 patients included along with 142 histologically characterized lesions. K agreement values were 0.69, 0.68, 0.63 and 0.56 for concordance between the gold standard and FFDM, FFDM + US, CESM and MRI, respectively (p < 0.001, for all). K concordance for CESM was 0.81 with FFDM + US and 0.73 with MRI (p value < 0.001 for all). Conclusions: CESM may represent a valuable alternative and/or an integrating technique to MRI in the evaluation of breast cancer patients.
Collapse
Affiliation(s)
- Francesca Romana Ferranti
- Radiology and Diagnostic Imaging Department, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (F.R.F.); (F.V.)
| | - Federica Vasselli
- Radiology and Diagnostic Imaging Department, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (F.R.F.); (F.V.)
| | - Maddalena Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
- Correspondence: (M.B.); (A.V.); Tel.: +39-0652665419 (M.B.); +39-0652662731 (A.V.)
| | - Francesca Sperati
- Biostatistics-Scientific Direction, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (F.S.); (I.T.)
| | - Irene Terrenato
- Biostatistics-Scientific Direction, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (F.S.); (I.T.)
| | - Franco Graziano
- Division of Breast Surgery, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (F.G.); (C.B.)
| | - Patrizia Vici
- Sperimentazioni di Fase IV, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Claudio Botti
- Division of Breast Surgery, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (F.G.); (C.B.)
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging Department, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; (F.R.F.); (F.V.)
- Correspondence: (M.B.); (A.V.); Tel.: +39-0652665419 (M.B.); +39-0652662731 (A.V.)
| |
Collapse
|
33
|
Li JW, Cao YC, Zhao ZJ, Shi ZT, Duan XQ, Chang C, Chen JG. Prediction for pathological and immunohistochemical characteristics of triple-negative invasive breast carcinomas: the performance comparison between quantitative and qualitative sonographic feature analysis. Eur Radiol 2022; 32:1590-1600. [PMID: 34519862 DOI: 10.1007/s00330-021-08224-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 06/28/2021] [Accepted: 07/15/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Sonographic features are associated with pathological and immunohistochemical characteristics of triple-negative breast cancer (TNBC). To predict the biological property of TNBC, the performance using quantitative high-throughput sonographic feature analysis was compared with that using qualitative feature assessment. METHODS We retrospectively reviewed ultrasound images, clinical, pathological, and immunohistochemical (IHC) data of 252 female TNBC patients. All patients were subgrouped according to the histological grade, Ki67 expression level, and human epidermal growth factor receptor 2 (HER2) score. Qualitative sonographic feature assessment included shape, margin, posterior acoustic pattern, and calcification referring to the Breast Imaging Reporting and Data System (BI-RADS). Quantitative sonographic features were acquired based on the computer-aided radiomics analysis. Breast cancer masses were manually segmented from the surrounding breast tissues. For each ultrasound image, 1688 radiomics features of 7 feature classes were extracted. The principal component analysis (PCA), least absolute shrinkage and selection operator (LASSO), and support vector machine (SVM) were used to determine the high-throughput radiomics features that were highly correlated to biological properties. The performance using both quantitative and qualitative sonographic features to predict biological properties of TNBC was represented by the area under the receiver operating characteristic curve (AUC). RESULTS In the qualitative assessment, regular tumor shape, no angular or spiculated margin, posterior acoustic enhancement, and no calcification were used as the independent sonographic features for TNBC. Using the combination of these four features to predict the histological grade, Ki67, HER2, axillary lymph node metastasis (ALNM), and lymphovascular invasion (LVI), the AUC was 0.673, 0.680, 0.651, 0.587, and 0.566, respectively. The number of high-throughput features that closely correlated with biological properties was 34 for histological grade (AUC 0.942), 27 for Ki67 (AUC 0.732), 25 for HER2 (AUC 0.730), 34 for ALNM (AUC 0.804), and 34 for LVI (AUC 0.795). CONCLUSION High-throughput quantitative sonographic features are superior to traditional qualitative ultrasound features in predicting the biological behavior of TNBC. KEY POINTS • Sonographic appearances of TNBCs showed a great variety in accordance with its biological and clinical characteristics. • Both qualitative and quantitative sonographic features of TNBCs are associated with tumor biological characteristics. • The quantitative high-throughput feature analysis is superior to two-dimensional sonographic feature assessment in predicting tumor biological property.
Collapse
Affiliation(s)
- Jia-Wei Li
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Yu-Cheng Cao
- Shanghai Key Laboratory of Multidimensional Information Processing, East China Normal University, #500 Dongchuan Rd., Shanghai, 200241, China
| | - Zhi-Jin Zhao
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Zhao-Ting Shi
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Xiao-Qian Duan
- Shanghai Key Laboratory of Multidimensional Information Processing, East China Normal University, #500 Dongchuan Rd., Shanghai, 200241, China
| | - Cai Chang
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.
| | - Jian-Gang Chen
- Shanghai Key Laboratory of Multidimensional Information Processing, East China Normal University, #500 Dongchuan Rd., Shanghai, 200241, China.
| |
Collapse
|
34
|
Wang S, Wang Z, Li R, You C, Mao N, Jiang T, Wang Z, Xie H, Gu Y. Association between quantitative and qualitative image features of contrast-enhanced mammography and molecular subtypes of breast cancer. Quant Imaging Med Surg 2022; 12:1270-1280. [PMID: 35111622 DOI: 10.21037/qims-21-589] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 09/24/2021] [Indexed: 01/21/2023]
Abstract
Background The molecular subtype of breast cancer is one of the most important factors affecting patient prognosis. The study aimed to analyze the association between quantitative and qualitative features of contrast-enhanced mammography (CEM) images and breast cancer molecular subtypes. Methods This retrospective double-center study included women who underwent CEM between November 2017 and April 2020. Each patient had at least 1 malignant lesion confirmed by pathology. The CEM images were evaluated by 2 radiologists to obtain quantitative and qualitative image features. The molecular subtypes were studied as dichotomous outcomes, including luminal versus non-luminal, human epidermal growth factor receptor (HER2)-enriched versus non-HER2-enriched, and triple-negative breast cancer (TNBC) versus non-TNBC subtypes. The association between the image features and molecular subtypes was analyzed by multivariate logistic regression, with odds ratios (ORs) and 95% confidence intervals (CIs) provided. Results A total of 151 patients with 160 malignant lesions were included in the study. For quantitative features, a higher standard deviation of lesion density was associated with non-luminal (OR =0.88, 95% CI: 0.81 to 0.96, P=0.004) and HER2-enriched breast cancers (OR =1.16, 95% CI: 1.04 to 1.28, P=0.006). The relative degree of enhancement (RDE) and contrast-to-noise ratio (CNR) were not associated with molecular subtypes. However, a higher CNR/lesion size (OR =1.06, 95% CI: 1.01 to 1.12, P=0.012) was associated with luminal subtype cancers, and a higher RDE/lesion size (OR =0.94, 95% CI: 0.88 to 1.00, P=0.035) or a higher CNR/lesion size (OR =0.94, 95% CI: 0.88-1.00, P=0.038) was associated with non-TNBCs. For qualitative features, the presence of calcification was associated with HER2-enriched breast cancers (OR =2.91, 95% CI: 1.10 to 7.67, P=0.031). The presence of architectural distortion was associated with luminal cancer (OR =14.50, 95% CI: 1.91 to 110.14, P=0.010) and non-TNBC (OR =0.05, 95% CI: 0.00 to 0.43, P=0.022). Non-mass enhancement (OR =2.78, 95% CI: 1.08 to 7.14, P=0.033) was associated with HER2-enriched breast cancers. An association remained after adjustments for age, breast thickness, and breast density (all adjusted P<0.050). Conclusions The quantitative and qualitative imaging features of CEM could contribute to distinguishing breast cancer molecular subtypes.
Collapse
Affiliation(s)
- Simin Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | - Ruimin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ning Mao
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, China
| | - Tingting Jiang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhongyi Wang
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, China
| | - Haizhu Xie
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Mireștean CC, Volovăț C, Iancu RI, Iancu DPT. Radiomics in Triple Negative Breast Cancer: New Horizons in an Aggressive Subtype of the Disease. J Clin Med 2022; 11:jcm11030616. [PMID: 35160069 PMCID: PMC8836903 DOI: 10.3390/jcm11030616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 12/17/2022] Open
Abstract
In the last decade, the analysis of the medical images has evolved significantly, applications and tools capable to extract quantitative characteristics of the images beyond the discrimination capacity of the investigator's eye being developed. The applications of this new research field, called radiomics, presented an exponential growth with direct implications in the diagnosis and prediction of response to therapy. Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype with a severe prognosis, despite the aggressive multimodal treatments applied according to the guidelines. Radiomics has already proven the ability to differentiate TNBC from fibroadenoma. Radiomics features extracted from digital mammography may also distinguish between TNBC and non-TNBC. Recent research has identified three distinct subtypes of TNBC using IRM breast images voxel-level radiomics features (size/shape related features, texture features, sharpness). The correlation of these TNBC subtypes with the clinical response to neoadjuvant therapy may lead to the identification of biomarkers in order to guide the clinical decision. Furthermore, the variation of some radiomics features in the neoadjuvant settings provides a tool for the rapid evaluation of treatment efficacy. The association of radiomics features with already identified biomarkers can generate complex predictive and prognostic models. Standardization of image acquisition and also of radiomics feature extraction is required to validate this method in clinical practice.
Collapse
Affiliation(s)
- Camil Ciprian Mireștean
- Department of Oncology and Radiotherapy, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
- Department of Surgery, Railways Clinical Hospital, 700506 Iasi, Romania
| | - Constantin Volovăț
- Department of Medical Oncology-Radiotherapy, Faculty of Medicine, “Gr. T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.V.); (D.P.T.I.)
- Euroclinic Oncological Hospital, 700110 Iasi, Romania
| | - Roxana Irina Iancu
- Department of Oral Pathology, Faculty of Dentistry, “Gr. T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Laboratory Department, “St. Spiridon” Emergency Hospital, 700111 Iasi, Romania
- Correspondence: ; Tel.: +40-232-301-603
| | - Dragoș Petru Teodor Iancu
- Department of Medical Oncology-Radiotherapy, Faculty of Medicine, “Gr. T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.V.); (D.P.T.I.)
- Department of Radiotherapy, Regional Institute of Oncology, 700483 Iasi, Romania
| |
Collapse
|
36
|
Mao N, Shi Y, Lian C, Wang Z, Zhang K, Xie H, Zhang H, Chen Q, Cheng G, Xu C, Dai Y. Intratumoral and peritumoral radiomics for preoperative prediction of neoadjuvant chemotherapy effect in breast cancer based on contrast-enhanced spectral mammography. Eur Radiol 2022; 32:3207-3219. [DOI: 10.1007/s00330-021-08414-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/26/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022]
|
37
|
Zhang Y, Liu F, Zhang H, Ma H, Sun J, Zhang R, Song L, Shi H. Diagnostic Value of Radiomics Analysis in Contrast-Enhanced Spectral Mammography for Identifying Triple-Negative Breast Cancer. Front Oncol 2022; 11:773196. [PMID: 35004294 PMCID: PMC8733550 DOI: 10.3389/fonc.2021.773196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/02/2021] [Indexed: 11/29/2022] Open
Abstract
Purpose To evaluate the value of radiomics analysis in contrast-enhanced spectral mammography (CESM) for the identification of triple-negative breast cancer (TNBC). Method CESM images of 367 pathologically confirmed breast cancer patients (training set: 218, testing set: 149) were retrospectively analyzed. Cranial caudal (CC), mediolateral oblique (MLO), and combined models were built on the basis of the features extracted from subtracted images on CC, MLO, and the combination of CC and MLO, respectively, in the tumour region. The performance of the models was evaluated through receiver operating characteristic (ROC) curve analysis, the Hosmer-Lemeshow test, and decision curve analysis (DCA). The areas under ROC curves (AUCs) were compared through the DeLong test. Results The combined CC and MLO model had the best AUC and sensitivity of 0.90 (95% confidence interval: 0.85–0.96) and 0.97, respectively. The Hosmer–Lemeshow test yielded a non-significant statistic with p-value of 0.59. The clinical usefulness of the combined CC and MLO model was confirmed if the threshold was between 0.02 and 0.81 in the DCA. Conclusions Machine learning models based on subtracted images in CESM images were valuable for distinguishing TNBC and NTNBC. The model with the combined CC and MLO features had the best performance compared with models that used CC or MLO features alone.
Collapse
Affiliation(s)
- Yongxia Zhang
- Department of Medical Imaging, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Fengjie Liu
- Department of Radiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Han Zhang
- Department of Radiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Heng Ma
- Department of Radiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Jian Sun
- Department of Radiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Ran Zhang
- Marketing Research Department, Huiying Medical Technology Co. Ltd, Beijing, China
| | - Lei Song
- Department of Geratology, Yantai Yuhuangding Hospital, Yantai, China
| | - Hao Shi
- Department of Medical Imaging, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
38
|
Park HS, Lee KS, Seo BK, Kim ES, Cho KR, Woo OH, Song SE, Lee JY, Cha J. Machine Learning Models That Integrate Tumor Texture and Perfusion Characteristics Using Low-Dose Breast Computed Tomography Are Promising for Predicting Histological Biomarkers and Treatment Failure in Breast Cancer Patients. Cancers (Basel) 2021; 13:cancers13236013. [PMID: 34885124 PMCID: PMC8656976 DOI: 10.3390/cancers13236013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/17/2021] [Accepted: 11/27/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Tumor angiogenesis and heterogeneity are associated with poor prognosis for breast cancer. Advances in computer technology have made it possible to noninvasively quantify tumor angiogenesis and heterogeneity appearing in imaging data. We investigated whether low-dose CT could be used as a method for functional oncology imaging to assess tumor heterogeneity and angiogenesis in breast cancer and to predict noninvasively histological biomarkers and molecular subtypes of breast cancer. Low-dose breast CT has advantages in terms of radiation safety and patient convenience. Our study produced promising results for the use of machine learning with low-dose breast CT to identify histological prognostic factors including hormone receptor and human epidermal growth factor receptor 2 status, grade, and molecular subtype in patients with invasive breast cancer. Machine learning that integrates texture and perfusion features of breast cancer with low-dose CT can provide valuable information for the realization of precision medicine. Abstract This prospective study enrolled 147 women with invasive breast cancer who underwent low-dose breast CT (80 kVp, 25 mAs, 1.01–1.38 mSv) before treatment. From each tumor, we extracted eight perfusion parameters using the maximum slope algorithm and 36 texture parameters using the filtered histogram technique. Relationships between CT parameters and histological factors were analyzed using five machine learning algorithms. Performance was compared using the area under the receiver-operating characteristic curve (AUC) with the DeLong test. The AUCs of the machine learning models increased when using both features instead of the perfusion or texture features alone. The random forest model that integrated texture and perfusion features was the best model for prediction (AUC = 0.76). In the integrated random forest model, the AUCs for predicting human epidermal growth factor receptor 2 positivity, estrogen receptor positivity, progesterone receptor positivity, ki67 positivity, high tumor grade, and molecular subtype were 0.86, 0.76, 0.69, 0.65, 0.75, and 0.79, respectively. Entropy of pre- and postcontrast images and perfusion, time to peak, and peak enhancement intensity of hot spots are the five most important CT parameters for prediction. In conclusion, machine learning using texture and perfusion characteristics of breast cancer with low-dose CT has potential value for predicting prognostic factors and risk stratification in breast cancer patients.
Collapse
Affiliation(s)
- Hyun-Soo Park
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan-si 15355, Korea; (H.-S.P.); (E.-S.K.); (J.C.)
| | - Kwang-sig Lee
- AI Center, Korea University Anam Hospital, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea;
| | - Bo-Kyoung Seo
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan-si 15355, Korea; (H.-S.P.); (E.-S.K.); (J.C.)
- Correspondence:
| | - Eun-Sil Kim
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan-si 15355, Korea; (H.-S.P.); (E.-S.K.); (J.C.)
| | - Kyu-Ran Cho
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, Korea; (K.-R.C.); (S.-E.S.)
| | - Ok-Hee Woo
- Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul 08308, Korea;
| | - Sung-Eun Song
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, Korea; (K.-R.C.); (S.-E.S.)
| | - Ji-Young Lee
- Department of Radiology, Ilsan Paik Hospital, Inje University College of Medicine, 170 Juhwa-ro, Ilsanseo-gu, Goyang 10380, Korea;
| | - Jaehyung Cha
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan-si 15355, Korea; (H.-S.P.); (E.-S.K.); (J.C.)
- Cheng Hyang NF Co., Ltd., 44-5 Daehak-ro, Jongno-gu, Seoul 03122, Korea
| |
Collapse
|
39
|
Peng S, Chen L, Tao J, Liu J, Zhu W, Liu H, Yang F. Radiomics Analysis of Multi-Phase DCE-MRI in Predicting Tumor Response to Neoadjuvant Therapy in Breast Cancer. Diagnostics (Basel) 2021; 11:diagnostics11112086. [PMID: 34829433 PMCID: PMC8625316 DOI: 10.3390/diagnostics11112086] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/19/2022] Open
Abstract
Objective: To explore whether the pretreatment dynamic contrast enhancement magnetic resonance imaging (DCE-MRI) and radiomics signatures were associated with pathologic complete response (pCR) to neoadjuvant therapy (NAT) in breast cancer. Method: A retrospective review of 70 patients with breast invasive carcinomas proved by biopsy between June 2017 and October 2020 (26 patients were pathological complete response, and 44 patients were non-pathological complete response). Within the pre-contrast and five post-contrast dynamic series, a total of 1037 quantitative imaging features were extracted from in each phase. Additionally, the Δfeatures (the difference between the features before and after the comparison) were used for subsequent analysis. The least absolute shrinkage and selection operator (LASSO) regression method was used to select features related to pCR, and then use these features to train multiple machine learning classifiers to predict the probability of pCR for a given patient. The area under the curve (AUC), accuracy, sensitivity, and specificity were calculated to assess the predictive performances of the radiomics model for each of the five phases of time points. Result: Among the five phases, each individual phase performed with AUCs ranging from 0.845 to 0.919 in predicting pCR. The best single phases performance was given by the 3rd phase (AUC = 0.919, sensitivity 0.885, specificity 0.864). 5 of the features have significant differences between pCR and non-pCR groups in each phase, most features reach their maximum or minimum in the 2nd or 3rd phase. Conclusion: The radiomic features extracted from each phase of pre-treatment DCE-MRI possess discriminatory power to predict tumor response.
Collapse
Affiliation(s)
- Shuyi Peng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (S.P.); (L.C.); (J.T.); (J.L.); (W.Z.)
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Leqing Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (S.P.); (L.C.); (J.T.); (J.L.); (W.Z.)
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Juan Tao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (S.P.); (L.C.); (J.T.); (J.L.); (W.Z.)
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Jie Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (S.P.); (L.C.); (J.T.); (J.L.); (W.Z.)
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Wenying Zhu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (S.P.); (L.C.); (J.T.); (J.L.); (W.Z.)
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Huan Liu
- Precision Healthcare Institute, GE Healthcare, Shanghai 201203, China;
| | - Fan Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (S.P.); (L.C.); (J.T.); (J.L.); (W.Z.)
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Correspondence: ; Tel.: +86-027-85726392
| |
Collapse
|
40
|
Sentinel Lymph Node Metastasis on Clinically Negative Patients: Preliminary Results of a Machine Learning Model Based on Histopathological Features. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112110372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The reported incidence of node metastasis at sentinel lymph node biopsy is generally low, so that the majority of women underwent unnecessary invasive axilla surgery. Although the sentinel lymph node biopsy is time consuming and expensive, it is still the intra-operative exam with the highest performance, but sometimes surgery is achieved without a clear diagnosis and also with possible serious complications. In this work, we developed a machine learning model to predict the sentinel lymph nodes positivity in clinically negative patients. Breast cancer clinical and immunohistochemical features of 907 patients characterized by a clinically negative lymph node status were collected. We trained different machine learning algorithms on the retrospective collected data and selected an optimal subset of features through a sequential forward procedure. We found comparable performances for different classification algorithms: on a hold-out training set, the logistics regression classifier with seven features, i.e., tumor diameter, age, histologic type, grading, multiplicity, in situ component and Her2-neu status reached an AUC value of 71.5% and showed a better trade-off between sensitivity and specificity (69.4 and 66.9%, respectively) compared to other two classifiers. On the hold-out test set, the performance dropped by five percentage points in terms of accuracy. Overall, the histological characteristics alone did not allow us to develop a support tool suitable for actual clinical application, but it showed the maximum informative power contained in the same for the resolution of the clinical problem. The proposed study represents a starting point for future development of predictive models to obtain the probability for lymph node metastases by using histopathological features combined with other features of a different nature.
Collapse
|
41
|
Rana SP, Dey M, Loretoni R, Duranti M, Sani L, Vispa A, Ghavami M, Dudley S, Tiberi G. Radial Basis Function for Breast Lesion Detection from MammoWave Clinical Data. Diagnostics (Basel) 2021; 11:1930. [PMID: 34679628 PMCID: PMC8534354 DOI: 10.3390/diagnostics11101930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 11/22/2022] Open
Abstract
Recently, a novel microwave apparatus for breast lesion detection (MammoWave), uniquely able to function in air with 2 antennas rotating in the azimuth plane and operating within the band 1-9 GHz has been developed. Machine learning (ML) has been implemented to understand information from the frequency spectrum collected through MammoWave in response to the stimulus, segregating breasts with and without lesions. The study comprises 61 breasts (from 35 patients), each one with the correspondent output of the radiologist's conclusion (i.e., gold standard) obtained from echography and/or mammography and/or MRI, plus pathology or 1-year clinical follow-up when required. The MammoWave examinations are performed, recording the frequency spectrum, where the magnitudes show substantial discrepancy and reveals dissimilar behaviours when reflected from tissues with/without lesions. Principal component analysis is implemented to extract the unique quantitative response from the frequency response for automated breast lesion identification, engaging the support vector machine (SVM) with a radial basis function kernel. In-vivo feasibility validation (now ended) of MammoWave was approved in 2015 by the Ethical Committee of Umbria, Italy (N. 6845/15/AV/DM of 14 October 2015, N. 10352/17/NCAV of 16 March 2017, N 13203/18/NCAV of 17 April 2018). Here, we used a set of 35 patients. According to the radiologists conclusions, 25 breasts without lesions and 36 breasts with lesions underwent a MammoWave examination. The proposed SVM model achieved the accuracy, sensitivity, and specificity of 91%, 84.40%, and 97.20%. The proposed ML augmented MammoWave can identify breast lesions with high accuracy.
Collapse
Affiliation(s)
- Soumya Prakash Rana
- School of Engineering, London South Bank University, London SE1 0AA, UK; (M.D.); (M.G.); (S.D.); (G.T.)
| | - Maitreyee Dey
- School of Engineering, London South Bank University, London SE1 0AA, UK; (M.D.); (M.G.); (S.D.); (G.T.)
| | - Riccardo Loretoni
- Breast Screening and Diagnostic Breast Cancer Unit, AUSL Umbria 2, 06034 Foligno, Italy;
| | - Michele Duranti
- Department of Diagnostic Imaging, Perugia Hospital, 06156 Perugia, Italy;
| | - Lorenzo Sani
- UBT-Umbria Bioengineering Technologies, 06081 Perugia, Italy; (L.S.); (A.V.)
| | - Alessandro Vispa
- UBT-Umbria Bioengineering Technologies, 06081 Perugia, Italy; (L.S.); (A.V.)
| | - Mohammad Ghavami
- School of Engineering, London South Bank University, London SE1 0AA, UK; (M.D.); (M.G.); (S.D.); (G.T.)
| | - Sandra Dudley
- School of Engineering, London South Bank University, London SE1 0AA, UK; (M.D.); (M.G.); (S.D.); (G.T.)
| | - Gianluigi Tiberi
- School of Engineering, London South Bank University, London SE1 0AA, UK; (M.D.); (M.G.); (S.D.); (G.T.)
- UBT-Umbria Bioengineering Technologies, 06081 Perugia, Italy; (L.S.); (A.V.)
| |
Collapse
|
42
|
Wang S, Sun Y, Mao N, Duan S, Li Q, Li R, Jiang T, Wang Z, Xie H, Gu Y. Incorporating the clinical and radiomics features of contrast-enhanced mammography to classify breast lesions: a retrospective study. Quant Imaging Med Surg 2021; 11:4418-4430. [PMID: 34603996 DOI: 10.21037/qims-21-103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/11/2021] [Indexed: 12/21/2022]
Abstract
Background Contrast-enhanced mammography (CEM) is a promising breast imaging technique. A limited number of studies have focused on the radiomics analysis of CEM. We intended to explore whether a model constructed with both clinical and radiomics features of CEM can better classify benign and malignant breast lesions. Methods This retrospective, double-center study included women who underwent CEM between August 2017 and February 2020. The data from Center 1 were used as training set and the data from Center 2 were used as external testing set (training: testing =2:1). Models were constructed with the clinical, radiomics, and clinical + radiomics features of CEM. The clinical features included patient age and clinical image features interpreted by the radiologists. The radiomics features were extracted from high-energy (HE), low-energy (LE), and dual-energy subtraction (DES) images of CEM. The Mann-Whitney U test, Pearson correlation and Boruta's approach were used to select the radiomics features. Random Forest (RF) and logistic regression were used to establish the models. For the testing set, the areas under the curve (AUCs) and 95% confidence intervals (CIs) were employed to evaluate the performance of the models. For the training set, the mean AUCs were obtained by performing internal validation for 100 iterations and then compared by the Kruskal-Wallis and Mann-Whitney U tests. Results A total of 226 women (mean age: 47.4±10.1 years) with 226 pathologically proven breast lesions (101 benign; 125 malignant) were included. For the external testing set, the AUCs were 0.964 (95% CI: 0.918-1.000) for the combined model, 0.947 (95% CI: 0.891-0.997) for the radiomics model, and 0.882 (95% CI: 0.803-0.962) for the clinical model. In the internal validation process, the combined model achieved a mean AUC of 0.934±0.030, which was significantly higher than those of the radiomics (mean AUC =0.921±0.031, adjusted P<0.050) and clinical models (mean AUC =0.907±0.036; adjusted P<0.050). Conclusions Incorporating both clinical and radiomics features of CEM may achieve better classification results for breast lesions.
Collapse
Affiliation(s)
- Simin Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuqi Sun
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Ning Mao
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, China
| | | | - Qin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruimin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tingting Jiang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhongyi Wang
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, China
| | - Haizhu Xie
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
43
|
Xi G, He J, Kang D, Xu S, Guo W, Fu F, Liu Y, Zheng L, Qiu L, Li L, Wang C, Chen J. Nomogram model combining macro and micro tumor-associated collagen signatures obtained from multiphoton images to predict the histologic grade in breast cancer. BIOMEDICAL OPTICS EXPRESS 2021; 12:6558-6570. [PMID: 34745756 PMCID: PMC8548007 DOI: 10.1364/boe.433281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 06/13/2023]
Abstract
The purpose of this study is to develop and validate a new nomogram model combining macro and micro tumor-associated collagen signatures obtained from multiphoton images to differentiate tumor grade in patients with invasive breast cancer. A total of 543 patients were included in this study. We used computer-generated random numbers to assign 328 of these patients to the training cohort and 215 patients to the validation cohort. Macroscopic tumor-associated collagen signatures (TACS1-8) were obtained by multiphoton microscopy at the invasion front and inside of the breast primary tumor. TACS corresponding microscopic features (TCMF) including morphology and texture features were extracted from the segmented regions of interest using Matlab 2016b. Using ridge regression analysis, we obtained a TACS-score for each patient based on the combined TACS1-8, and the least absolute shrinkage and selection operator (LASSO) regression was applied to select the most robust TCMF features to build a TCMF-score. Univariate logistic regression analysis demonstrates that the TACS-score and TCMF-score are significantly associated with histologic grade (odds ratio, 2.994; 95% CI, 2.013-4.452; P < 0.001; 4.245, 2.876-6.264, P < 0.001 in the training cohort). The nomogram (collagen) model combining the TACS-score and TCMF-score could stratify patients into Grade1 and Grade2/3 groups with the AUC of 0.859 and 0.863 in the training and validation cohorts. The predictive performance can be further improved by combining the clinical factors, achieving the AUC of 0.874 in both data cohorts. The nomogram model combining the TACS-score and TCMF-score can be useful in differentiating breast tumor patients with Grade1 and Grade2/3.
Collapse
Affiliation(s)
- Gangqin Xi
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou 350007, China
- These authors contributed equally to this work
| | - Jiajia He
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou 350007, China
- These authors contributed equally to this work
| | - Deyong Kang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou 350001, China
- These authors contributed equally to this work
| | - Shuoyu Xu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenhui Guo
- Department of Breast Surgery, Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Fangmeng Fu
- Department of Breast Surgery, Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Yulan Liu
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Liqin Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Lida Qiu
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou 350007, China
- College of Physics and Electronic Information Engineering, Minjiang University, Fuzhou 350108, China
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Chuan Wang
- Department of Breast Surgery, Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| |
Collapse
|
44
|
Huang Y, Wei L, Hu Y, Shao N, Lin Y, He S, Shi H, Zhang X, Lin Y. Multi-Parametric MRI-Based Radiomics Models for Predicting Molecular Subtype and Androgen Receptor Expression in Breast Cancer. Front Oncol 2021; 11:706733. [PMID: 34490107 PMCID: PMC8416497 DOI: 10.3389/fonc.2021.706733] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/28/2021] [Indexed: 12/30/2022] Open
Abstract
Objective To investigate whether radiomics features extracted from multi-parametric MRI combining machine learning approach can predict molecular subtype and androgen receptor (AR) expression of breast cancer in a non-invasive way. Materials and Methods Patients diagnosed with clinical T2–4 stage breast cancer from March 2016 to July 2020 were retrospectively enrolled. The molecular subtypes and AR expression in pre-treatment biopsy specimens were assessed. A total of 4,198 radiomics features were extracted from the pre-biopsy multi-parametric MRI (including dynamic contrast-enhancement T1-weighted images, fat-suppressed T2-weighted images, and apparent diffusion coefficient map) of each patient. We applied several feature selection strategies including the least absolute shrinkage and selection operator (LASSO), and recursive feature elimination (RFE), the maximum relevance minimum redundancy (mRMR), Boruta and Pearson correlation analysis, to select the most optimal features. We then built 120 diagnostic models using distinct classification algorithms and feature sets divided by MRI sequences and selection strategies to predict molecular subtype and AR expression of breast cancer in the testing dataset of leave-one-out cross-validation (LOOCV). The performances of binary classification models were assessed via the area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV). And the performances of multiclass classification models were assessed via AUC, overall accuracy, precision, recall rate, and F1-score. Results A total of 162 patients (mean age, 46.91 ± 10.08 years) were enrolled in this study; 30 were low-AR expression and 132 were high-AR expression. HR+/HER2− cancers were diagnosed in 56 cases (34.6%), HER2+ cancers in 81 cases (50.0%), and TNBC in 25 patients (15.4%). There was no significant difference in clinicopathologic characteristics between low-AR and high-AR groups (P > 0.05), except the menopausal status, ER, PR, HER2, and Ki-67 index (P = 0.043, <0.001, <0.001, 0.015, and 0.006, respectively). No significant difference in clinicopathologic characteristics was observed among three molecular subtypes except the AR status and Ki-67 (P = <0.001 and 0.012, respectively). The Multilayer Perceptron (MLP) showed the best performance in discriminating AR expression, with an AUC of 0.907 and an accuracy of 85.8% in the testing dataset. The highest performances were obtained for discriminating TNBC vs. non-TNBC (AUC: 0.965, accuracy: 92.6%), HER2+ vs. HER2− (AUC: 0.840, accuracy: 79.0%), and HR+/HER2− vs. others (AUC: 0.860, accuracy: 82.1%) using MLP as well. The micro-AUC of MLP multiclass classification model was 0.896, and the overall accuracy was 0.735. Conclusions Multi-parametric MRI-based radiomics combining with machine learning approaches provide a promising method to predict the molecular subtype and AR expression of breast cancer non-invasively.
Collapse
Affiliation(s)
- Yuhong Huang
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lihong Wei
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yalan Hu
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nan Shao
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingyu Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shaofu He
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huijuan Shi
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoling Zhang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Lin
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
45
|
Gatta G, Cappabianca S, La Forgia D, Massafra R, Fanizzi A, Cuccurullo V, Brunese L, Tagliafico A, Grassi R. Second-Generation 3D Automated Breast Ultrasonography (Prone ABUS) for Dense Breast Cancer Screening Integrated to Mammography: Effectiveness, Performance and Detection Rates. J Pers Med 2021; 11:875. [PMID: 34575652 PMCID: PMC8468126 DOI: 10.3390/jpm11090875] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/24/2021] [Accepted: 08/29/2021] [Indexed: 12/22/2022] Open
Abstract
In our study, we added a three-dimensional automated breast ultrasound (3D ABUS) to mammography to evaluate the performance and cancer detection rate of mammography alone or with the addition of 3D prone ABUS in women with dense breasts. Our prospective observational study was based on the screening of 1165 asymptomatic women with dense breasts who selected independent of risk factors. The results evaluated include the cancers detected between June 2017 and February 2019, and all surveys were subjected to a double reading. Mammography detected four cancers, while mammography combined with a prone Sofia system (3D ABUS) doubled the detection rate, with eight instances of cancer being found. The diagnostic yield difference was 3.4 per 1000. Mammography alone was subjected to a recall rate of 14.5 for 1000 women, while mammography combined with 3D prone ABUS resulted in a recall rate of 26.6 per 1000 women. We also observed an additional 12.1 recalls per 1000 women screened. Integrating full-field digital mammography (FFDM) with 3D prone ABUS in women with high breast density increases and improves breast cancer detection rates in a significant manner, including small and invasive cancers, and it has a tolerable impact on recall rate. Moreover, 3D prone ABUS performance results are comparable with the performance results of the supine 3D ABUS system.
Collapse
Affiliation(s)
- Gianluca Gatta
- Dipartimento di Medicina di Precisione Università della Campania “Luigi Vanvitelli”, 80131 Napoli, Italy; (G.G.); (S.C.); (V.C.); (R.G.)
| | - Salvatore Cappabianca
- Dipartimento di Medicina di Precisione Università della Campania “Luigi Vanvitelli”, 80131 Napoli, Italy; (G.G.); (S.C.); (V.C.); (R.G.)
| | - Daniele La Forgia
- IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (R.M.); (A.F.)
| | - Raffaella Massafra
- IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (R.M.); (A.F.)
| | - Annarita Fanizzi
- IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (R.M.); (A.F.)
| | - Vincenzo Cuccurullo
- Dipartimento di Medicina di Precisione Università della Campania “Luigi Vanvitelli”, 80131 Napoli, Italy; (G.G.); (S.C.); (V.C.); (R.G.)
| | - Luca Brunese
- Dipartimento di Medicina e Scienze della Salute “Vincenzo Tiberio”—Università degli Studi del Molise, 86100 Campobasso, Italy;
| | | | - Roberto Grassi
- Dipartimento di Medicina di Precisione Università della Campania “Luigi Vanvitelli”, 80131 Napoli, Italy; (G.G.); (S.C.); (V.C.); (R.G.)
| |
Collapse
|
46
|
Carbonara N, La Forgia D, Pellegrino R, Ressa C, Tommasi S. A Cost Decision Model Supporting Treatment Strategy Selection in BRCA1/2 Mutation Carriers in Breast Cancer. J Pers Med 2021; 11:847. [PMID: 34575624 PMCID: PMC8470684 DOI: 10.3390/jpm11090847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/21/2021] [Accepted: 08/25/2021] [Indexed: 01/08/2023] Open
Abstract
In this paper, a cost decision-making model that compares the healthcare costs for diverse treatment strategies is built for BRCA-mutated women with breast cancer. Moreover, this model calculates the cancer treatment costs that could potentially be prevented, if the treatment strategy with the lowest total cost, along the entire lifetime of the patient, is chosen for high-risk women with breast cancer. The benchmark of the healthcare costs for diverse treatment strategies is selected in the presence of uncertainty, i.e., considering, throughout the lifetime of the patient, the risks and complications that may arise in each strategy and, therefore, the costs associated with the management of such events. Our results reveal a clear economic advantage of adopting the cost decision-making model for benchmarking the healthcare costs for various treatment strategies for BRCA-mutated women with breast cancer. The cost savings were higher when all breast cancer patients underwent counseling and genetic testing before deciding on any diagnostic-therapeutic path, with a probability of obtaining savings of over 75%.
Collapse
Affiliation(s)
- Nunzia Carbonara
- Departments of Mechanics Mathematics and Management, Politecnico di Bari, 70126 Bari, Italy;
| | - Daniele La Forgia
- SSD Radiodiagnostica Senologica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II” di Bari, 70124 Bari, Italy;
| | - Roberta Pellegrino
- Departments of Mechanics Mathematics and Management, Politecnico di Bari, 70126 Bari, Italy;
| | - Cosmo Ressa
- S.C. Chirurgia Plastica e Ricostruttiva, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II” di Bari, 70124 Bari, Italy;
| | - Stefania Tommasi
- SSD Diagnostica Molecolare e Farmacogenetica, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II” di Bari, 70124 Bari, Italy;
| |
Collapse
|
47
|
A Multicentre Evaluation of Dosiomics Features Reproducibility, Stability and Sensitivity. Cancers (Basel) 2021; 13:cancers13153835. [PMID: 34359737 PMCID: PMC8345157 DOI: 10.3390/cancers13153835] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Dosiomics is born directly as an extension of radiomics: it entails extracting features from the patients’ three-dimensional (3D) radiotherapy dose distribution rather than from conventional medical images to obtain specific spatial and statistical information. Dosiomic studies, in a multicentre setting, require assessing the features’ stability to dose calculation settings and the features’ capability in distinguishing different dose distributions. This study provides the first multicentre evaluation of the dosiomic features in terms of reproducibility, stability and sensitivity across various dose distributions obtained from multiple technologies and techniques and considering different dose calculation algorithms of TPS and two different resolutions of the dose grid. Harmonisation strategies to account for a possible variation in the dose distribution due to these confounding factors should be adopted when investigating a correlation between dosiomic features and clinical outcomes in multicentre studies. Abstract Dosiomics is a texture analysis method to produce dose features that encode the spatial 3D distribution of radiotherapy dose. Dosiomic studies, in a multicentre setting, require assessing the features’ stability to dose calculation settings and the features’ capability in distinguishing different dose distributions. Dose distributions were generated by eight Italian centres on a shared image dataset acquired on a dedicated phantom. Treatment planning protocols, in terms of planning target volume coverage and dose–volume constraints to the organs at risk, were shared among the centres to produce comparable dose distributions for measuring reproducibility/stability and sensitivity of dosiomic features. In addition, coefficient of variation (CV) was employed to evaluate the dosiomic features’ variation. We extracted 38,160 features from 30 different dose distributions from six regions of interest, grouped by four features’ families. A selected group of features (CV < 3 for the reproducibility/stability studies, CV > 1 for the sensitivity studies) were identified to support future multicentre studies, assuring both stable features when dose distributions variation is minimal and sensitive features when dose distribution variations need to be clearly identified. Dosiomic is a promising tool that could support multicentre studies, especially for predictive models, and encode the spatial and statistical characteristics of the 3D dose distribution.
Collapse
|
48
|
Differentiating Breast Tumors from Background Parenchymal Enhancement at Contrast-Enhanced Mammography: The Role of Radiomics-A Pilot Reader Study. Diagnostics (Basel) 2021; 11:diagnostics11071248. [PMID: 34359332 PMCID: PMC8305277 DOI: 10.3390/diagnostics11071248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 12/27/2022] Open
Abstract
Background: The purpose of this study was to assess the effectiveness of the radiomic analysis of contrast-enhanced spectral mammography (CESM) in discriminating between breast cancers and background parenchymal enhancement (BPE). Methods: This retrospective study included 38 patients that underwent CESM examinations for clinical purposes between January 2019–December 2020. A total of 57 malignant breast lesions and 23 CESM examinations with 31 regions of BPE were assessed through radiomic analysis using MaZda software. The parameters that demonstrated to be independent predictors for breast malignancy were exported into the B11 program and a k-nearest neighbor classifier (k-NN) was trained on the initial groups of patients and was tested using a validation group. Histopathology results obtained after surgery were considered the gold standard. Results: Radiomic analysis found WavEnLL_s_2 parameter as an independent predictor for breast malignancies with a sensitivity of 68.42% and a specificity of 83.87%. The prediction model that included CH1D6SumAverg, CN4D6Correlat, Kurtosis, Perc01, Perc10, Skewness, and WavEnLL_s_2 parameters had a sensitivity of 73.68% and a specificity of 80.65%. Higher values were obtained of WavEnLL_s_2 and the prediction model for tumors than for BPEs. The comparison between the ROC curves provided by the WaveEnLL_s_2 and the entire prediction model did not show statistically significant results (p = 0.0943). The k-NN classifier based on the parameter WavEnLL_s_2 had a sensitivity and specificity on training and validating groups of 71.93% and 45.16% vs. 60% and 44.44%, respectively. Conclusion: Radiomic analysis has the potential to differentiate CESM between malignant lesions and BPE. Further quantitative insight into parenchymal enhancement patterns should be performed to facilitate the role of BPE in personalized clinical decision-making and risk assessment.
Collapse
|
49
|
Chakraborty D, Ivan C, Amero P, Khan M, Rodriguez-Aguayo C, Başağaoğlu H, Lopez-Berestein G. Explainable Artificial Intelligence Reveals Novel Insight into Tumor Microenvironment Conditions Linked with Better Prognosis in Patients with Breast Cancer. Cancers (Basel) 2021; 13:3450. [PMID: 34298668 PMCID: PMC8303703 DOI: 10.3390/cancers13143450] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 12/29/2022] Open
Abstract
We investigated the data-driven relationship between immune cell composition in the tumor microenvironment (TME) and the ≥5-year survival rates of breast cancer patients using explainable artificial intelligence (XAI) models. We acquired TCGA breast invasive carcinoma data from the cbioPortal and retrieved immune cell composition estimates from bulk RNA sequencing data from TIMER2.0 based on EPIC, CIBERSORT, TIMER, and xCell computational methods. Novel insights derived from our XAI model showed that B cells, CD8+ T cells, M0 macrophages, and NK T cells are the most critical TME features for enhanced prognosis of breast cancer patients. Our XAI model also revealed the inflection points of these critical TME features, above or below which ≥5-year survival rates improve. Subsequently, we ascertained the conditional probabilities of ≥5-year survival under specific conditions inferred from the inflection points. In particular, the XAI models revealed that the B cell fraction (relative to all cells in a sample) exceeding 0.025, M0 macrophage fraction (relative to the total immune cell content) below 0.05, and NK T cell and CD8+ T cell fractions (based on cancer type-specific arbitrary units) above 0.075 and 0.25, respectively, in the TME could enhance the ≥5-year survival in breast cancer patients. The findings could lead to accurate clinical predictions and enhanced immunotherapies, and to the design of innovative strategies to reprogram the breast TME.
Collapse
Affiliation(s)
- Debaditya Chakraborty
- Department of Construction Science, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.I.); (P.A.); (C.R.-A.); (G.L.-B.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.I.); (P.A.); (C.R.-A.); (G.L.-B.)
| | - Maliha Khan
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.I.); (P.A.); (C.R.-A.); (G.L.-B.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.I.); (P.A.); (C.R.-A.); (G.L.-B.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
50
|
Wang S, Sun Y, Li R, Mao N, Li Q, Jiang T, Chen Q, Duan S, Xie H, Gu Y. Diagnostic performance of perilesional radiomics analysis of contrast-enhanced mammography for the differentiation of benign and malignant breast lesions. Eur Radiol 2021; 32:639-649. [PMID: 34189600 DOI: 10.1007/s00330-021-08134-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/16/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To conduct perilesional region radiomics analysis of contrast-enhanced mammography (CEM) images to differentiate benign and malignant breast lesions. METHODS AND MATERIALS This retrospective study included patients who underwent CEM from November 2017 to February 2020. Lesion contours were manually delineated. Perilesional regions were automatically obtained. Seven regions of interest (ROIs) were obtained for each lesion, including the lesion ROI, annular perilesional ROIs (1 mm, 3 mm, 5 mm), and lesion + perilesional ROIs (1 mm, 3 mm, 5 mm). Overall, 4,098 radiomics features were extracted from each ROI. Datasets were divided into training and testing sets (1:1). Seven classification models using features from the seven ROIs were constructed using LASSO regression. Model performance was assessed by the AUC with 95% CI. RESULTS Overall, 190 women with 223 breast lesions (101 benign; 122 malignant) were enrolled. In the testing set, the annular perilesional ROI of 3-mm model showed the highest AUC of 0.930 (95% CI: 0.882-0.977), followed by the annular perilesional ROI of 1 mm model (AUC = 0.929; 95% CI: 0.881-0.978) and the lesion ROI model (AUC = 0.909; 95% CI: 0.857-0.961). A new model was generated by combining the predicted probabilities of the lesion ROI and annular perilesional ROI of 3-mm models, which achieved a higher AUC in the testing set (AUC = 0.940). CONCLUSIONS Annular perilesional radiomics analysis of CEM images is useful for diagnosing breast cancers. Adding annular perilesional information to the radiomics model built on the lesion information may improve the diagnostic performance. KEY POINTS • Radiomics analysis of the annular perilesional region of 3 mm in CEM images may provide valuable information for the differential diagnosis of benign and malignant breast lesions. • The radiomics information from the lesion region and the annular perilesional region may be complementary. Combining the predicted probabilities of the models constructed by the features from the two regions may improve the diagnostic performance of radiomics models.
Collapse
Affiliation(s)
- Simin Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuqi Sun
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Ruimin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ning Mao
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Shandong, 264000, China
| | - Qin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tingting Jiang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qianqian Chen
- GE Healthcare China, No. 1 Huatuo Road, Shanghai, 210000, China
| | - Shaofeng Duan
- GE Healthcare China, No. 1 Huatuo Road, Shanghai, 210000, China
| | - Haizhu Xie
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Shandong, 264000, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|