1
|
Gullo RL, Brunekreef J, Marcus E, Han LK, Eskreis-Winkler S, Thakur SB, Mann R, Lipman KG, Teuwen J, Pinker K. AI Applications to Breast MRI: Today and Tomorrow. J Magn Reson Imaging 2024; 60:2290-2308. [PMID: 38581127 PMCID: PMC11452568 DOI: 10.1002/jmri.29358] [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: 12/06/2023] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 04/08/2024] Open
Abstract
In breast imaging, there is an unrelenting increase in the demand for breast imaging services, partly explained by continuous expanding imaging indications in breast diagnosis and treatment. As the human workforce providing these services is not growing at the same rate, the implementation of artificial intelligence (AI) in breast imaging has gained significant momentum to maximize workflow efficiency and increase productivity while concurrently improving diagnostic accuracy and patient outcomes. Thus far, the implementation of AI in breast imaging is at the most advanced stage with mammography and digital breast tomosynthesis techniques, followed by ultrasound, whereas the implementation of AI in breast magnetic resonance imaging (MRI) is not moving along as rapidly due to the complexity of MRI examinations and fewer available dataset. Nevertheless, there is persisting interest in AI-enhanced breast MRI applications, even as the use of and indications of breast MRI continue to expand. This review presents an overview of the basic concepts of AI imaging analysis and subsequently reviews the use cases for AI-enhanced MRI interpretation, that is, breast MRI triaging and lesion detection, lesion classification, prediction of treatment response, risk assessment, and image quality. Finally, it provides an outlook on the barriers and facilitators for the adoption of AI in breast MRI. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY: Stage 6.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joren Brunekreef
- AI for Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Eric Marcus
- AI for Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lynn K Han
- Weill Cornell Medical College, New York-Presbyterian Hospital, New York, NY, USA
| | - Sarah Eskreis-Winkler
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sunitha B Thakur
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ritse Mann
- AI for Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kevin Groot Lipman
- AI for Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jonas Teuwen
- AI for Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Katja Pinker
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
2
|
Chen Y, Chen S, Tang W, Konge Q, Zhong Z, Yu X, Sui Y, Hu W, Jiang X, Guo Y. Multiparametric MRI Radiomics With Machine Learning for Differentiating HER2-Zero, -Low, and -Positive Breast Cancer: Model Development, Testing, and Interpretability Analysis. AJR Am J Roentgenol 2024. [PMID: 39413232 DOI: 10.2214/ajr.24.31717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
BACKGROUND: MRI radiomics has been explored for three-tiered classification of breast cancer HER2 expression (i.e., HER2-zero, HER2-low, or HER2-positive), although understanding of how such models reach their predictions is lacking. OBJECTIVE: To develop and test multiparametric MRI radiomics machine-learning models for differentiating three-tiered HER2 expression levels in patients with breast cancer, and to explain the contributions of model features through local and global interpretations using SHapley Additive exPlanation (SHAP) analysis. METHODS: This retrospective study included 737 patients (mean age, 54.1±10.6 years) with breast cancer from two centers (center 1: n=578; center 2: n=159), who underwent breast MRI and had HER2 expression determined after excisional biopsy. Analysis entailed two tasks: differentiating HER2-negative (i.e., HER2-zero or HER2-low) from HER2-positive tumors (task 1), and differentiating HER2-zero from HER2-low tumors (task 2). For each task, patients from center 1 were randomly assigned in 7:3 ratio to training (task 1: n=405; task 2: n=284) or internal test (task 1: n=173; task 2: n=122) sets; those from center 2 formed an external test set (task 1: n=159; task 2: n=105). Radiomics features were extracted from early-phase dynamic contrast-enhanced images (DCE), T2-weighted images (T2WI), and DWI. For each task, a support vector machine (SVM) was used for feature selection; a multiparametric radiomics score (radscore) was computed using feature weights from SVM correlation coefficients; conventional MRI and combined models were constructed; and model performances were evaluated. SHAP analysis was used to provide local and global interpretations for model outputs. RESULTS: In the external test set, for task 1, AUCs for the conventional MRI model, radscore, and combined model were 0.624, 0.757, and 0.762, respectively; for task 2, AUC for radscore was 0.754, and no conventional MRI model or combined model could be constructed. SHAP analysis identified early-phase DCE features as having the strongest influence for both tasks; T2WI features also had a prominent role for task 2. CONCLUSION: The findings indicate suboptimal performance of MRI radiomics models for noninvasive characterization of HER2 expression. CLINICAL IMPACT: The study provides an example of the use of SHAP interpretation analysis to better understand predictions of imaging-based machine learning models.
Collapse
Affiliation(s)
- Yongxin Chen
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Siyi Chen
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Wenjie Tang
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Qingcong Konge
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Zhidan Zhong
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Xiaomeng Yu
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Yi Sui
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510005, China
| | - Wenke Hu
- Department of Radiology, Shenzhen Guangming District People's Hospital, Shenzhen, Guangdong 518000, China
| | - Xinqing Jiang
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Yuan Guo
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, 510180, China
| |
Collapse
|
3
|
Lo Gullo R, Ochoa-Albiztegui RE, Chakraborty J, Thakur SB, Robson M, Jochelson MS, Varela K, Resch D, Eskreis-Winkler S, Pinker K. Development of an MRI Radiomic Machine-Learning Model to Predict Triple-Negative Breast Cancer Based on Fibroglandular Tissue of the Contralateral Unaffected Breast in Breast Cancer Patients. Cancers (Basel) 2024; 16:3480. [PMID: 39456574 PMCID: PMC11506272 DOI: 10.3390/cancers16203480] [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: 07/25/2024] [Revised: 10/05/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
AIM The purpose of this study was to develop a radiomic-based machine-learning model to predict triple-negative breast cancer (TNBC) based on the contralateral unaffected breast's fibroglandular tissue (FGT) in breast cancer patients. MATERIALS AND METHODS This study retrospectively included 541 patients (mean age, 51 years; range, 26-82) who underwent a screening breast MRI between November 2016 and September 2018 and who were subsequently diagnosed with biopsy-confirmed, treatment-naïve breast cancer. Patients were divided into training (n = 250) and validation (n = 291) sets. In the training set, 132 radiomic features were extracted using the open-source CERR platform. Following feature selection, the final prediction model was created, based on a support vector machine with a polynomial kernel of order 2. RESULTS In the validation set, the final prediction model, which included four radiomic features, achieved an F1 score of 0.66, an area under the curve of 0.71, a sensitivity of 54% [47-60%], a specificity of 74% [65-84%], a positive predictive value of 84% [78-90%], and a negative predictive value of 39% [31-47%]. CONCLUSIONS TNBC can be predicted based on radiomic features extracted from the FGT of the contralateral unaffected breast of patients, suggesting the potential for risk prediction specific to TNBC.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Department of Radiology, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, New York, NY 10065, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (S.B.T.)
| | | | - Jayasree Chakraborty
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sunitha B. Thakur
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (S.B.T.)
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mark Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maxine S. Jochelson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (S.B.T.)
| | | | - Daphne Resch
- Medical School, Sigmund Freud University, A-1020 Vienna, Austria
| | - Sarah Eskreis-Winkler
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (S.B.T.)
| | - Katja Pinker
- Department of Radiology, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, New York, NY 10065, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (S.B.T.)
| |
Collapse
|
4
|
Murmu A, Győrffy B. Artificial intelligence methods available for cancer research. Front Med 2024; 18:778-797. [PMID: 39115792 DOI: 10.1007/s11684-024-1085-3] [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: 01/03/2024] [Accepted: 05/17/2024] [Indexed: 11/01/2024]
Abstract
Cancer is a heterogeneous and multifaceted disease with a significant global footprint. Despite substantial technological advancements for battling cancer, early diagnosis and selection of effective treatment remains a challenge. With the convenience of large-scale datasets including multiple levels of data, new bioinformatic tools are needed to transform this wealth of information into clinically useful decision-support tools. In this field, artificial intelligence (AI) technologies with their highly diverse applications are rapidly gaining ground. Machine learning methods, such as Bayesian networks, support vector machines, decision trees, random forests, gradient boosting, and K-nearest neighbors, including neural network models like deep learning, have proven valuable in predictive, prognostic, and diagnostic studies. Researchers have recently employed large language models to tackle new dimensions of problems. However, leveraging the opportunity to utilize AI in clinical settings will require surpassing significant obstacles-a major issue is the lack of use of the available reporting guidelines obstructing the reproducibility of published studies. In this review, we discuss the applications of AI methods and explore their benefits and limitations. We summarize the available guidelines for AI in healthcare and highlight the potential role and impact of AI models on future directions in cancer research.
Collapse
Affiliation(s)
- Ankita Murmu
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- National Laboratory for Drug Research and Development, Budapest, 1117, Hungary
- Department of Bioinformatics, Semmelweis University, Budapest, 1094, Hungary
| | - Balázs Győrffy
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary.
- Department of Bioinformatics, Semmelweis University, Budapest, 1094, Hungary.
- Department of Biophysics, University of Pecs, Pecs, 7624, Hungary.
| |
Collapse
|
5
|
Huang Z, Zhang X, Ju Y, Zhang G, Chang W, Song H, Gao Y. Explainable breast cancer molecular expression prediction using multi-task deep-learning based on 3D whole breast ultrasound. Insights Imaging 2024; 15:227. [PMID: 39320560 PMCID: PMC11424596 DOI: 10.1186/s13244-024-01810-9] [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: 09/19/2023] [Accepted: 09/03/2024] [Indexed: 09/26/2024] Open
Abstract
OBJECTIVES To noninvasively estimate three breast cancer biomarkers, estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) and enhance performance and interpretability via multi-task deep learning. METHODS The study included 388 breast cancer patients who received the 3D whole breast ultrasound system (3DWBUS) examinations at Xijing Hospital between October 2020 and September 2021. Two predictive models, a single-task and a multi-task, were developed; the former predicts biomarker expression, while the latter combines tumor segmentation with biomarker prediction to enhance interpretability. Performance evaluation included individual and overall prediction metrics, and Delong's test was used for performance comparison. The models' attention regions were visualized using Grad-CAM + + technology. RESULTS All patients were randomly split into a training set (n = 240, 62%), a validation set (n = 60, 15%), and a test set (n = 88, 23%). In the individual evaluation of ER, PR, and HER2 expression prediction, the single-task and multi-task models achieved respective AUCs of 0.809 and 0.735 for ER, 0.688 and 0.767 for PR, and 0.626 and 0.697 for HER2, as observed in the test set. In the overall evaluation, the multi-task model demonstrated superior performance in the test set, achieving a higher macro AUC of 0.733, in contrast to 0.708 for the single-task model. The Grad-CAM + + method revealed that the multi-task model exhibited a stronger focus on diseased tissue areas, improving the interpretability of how the model worked. CONCLUSION Both models demonstrated impressive performance, with the multi-task model excelling in accuracy and offering improved interpretability on noninvasive 3DWBUS images using Grad-CAM + + technology. CRITICAL RELEVANCE STATEMENT The multi-task deep learning model exhibits effective prediction for breast cancer biomarkers, offering direct biomarker identification and improved clinical interpretability, potentially boosting the efficiency of targeted drug screening. KEY POINTS Tumoral biomarkers are paramount for determining breast cancer treatment. The multi-task model can improve prediction performance, and improve interpretability in clinical practice. The 3D whole breast ultrasound system-based deep learning models excelled in predicting breast cancer biomarkers.
Collapse
Affiliation(s)
- Zengan Huang
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Xin Zhang
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Yan Ju
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Ge Zhang
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Wanying Chang
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Hongping Song
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China.
| | - Yi Gao
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
6
|
Qi YJ, Su GH, You C, Zhang X, Xiao Y, Jiang YZ, Shao ZM. Radiomics in breast cancer: Current advances and future directions. Cell Rep Med 2024; 5:101719. [PMID: 39293402 PMCID: PMC11528234 DOI: 10.1016/j.xcrm.2024.101719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/10/2024] [Accepted: 08/14/2024] [Indexed: 09/20/2024]
Abstract
Breast cancer is a common disease that causes great health concerns to women worldwide. During the diagnosis and treatment of breast cancer, medical imaging plays an essential role, but its interpretation relies on radiologists or clinical doctors. Radiomics can extract high-throughput quantitative imaging features from images of various modalities via traditional machine learning or deep learning methods following a series of standard processes. Hopefully, radiomic models may aid various processes in clinical practice. In this review, we summarize the current utilization of radiomics for predicting clinicopathological indices and clinical outcomes. We also focus on radio-multi-omics studies that bridge the gap between phenotypic and microscopic scale information. Acknowledging the deficiencies that currently hinder the clinical adoption of radiomic models, we discuss the underlying causes of this situation and propose future directions for advancing radiomics in breast cancer research.
Collapse
Affiliation(s)
- Ying-Jia Qi
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Guan-Hua Su
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xu Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
7
|
Shokeir FA, Elmokadem AH, Soliman N, Khater A, Bayoumi D. Texture analysis using non-contrast MRI to predict estrogen receptor status in breast cancer lesions: Is it feasible? Clin Radiol 2024; 79:e892-e899. [PMID: 38719689 DOI: 10.1016/j.crad.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 06/02/2024]
Abstract
PURPOSE We aimed to evaluate the feasibility of non-contrast-enhanced T1 sequence in texture analysis of breast cancer lesions to predict their estrogen receptor status. METHODS The study included 85 pathologically proven breast cancer lesions in 53 patients. Immunohistochemical studies were performed to determine the estrogen receptor status (ER). Lesions were divided into two groups: ER + ve status and ER-ve status. Texture analysis using the second-order analysis features [The Co-occurrence matrix (11 features)] was applied on both T1 and dynamic contrast-enhanced (DCE) MRI images for each lesion. Texture features gained from both T1 and DCE images were analyzed to obtain cut-off values using ROC curves to sort lesions according to their estrogen receptor status. RESULTS Angular second momentum and some of the entropy-based features showed statistically significant cut-off values in differentiation between the two groups [P-values for pre- and post-contrast images for AngSecMom (0.001, 0.008), sum entropy (0.003,0.005), and entropy (0.033,0.019), respectively]. On comparing the AUCs between pre- and post-contrast images, we found that differences were statistically insignificant. Sum of squares, sum variance, and sum average showed statistically significant cut-off points only on pre-contrast images [P-values for sum of squares (0.018), sum variance (0.024), and sum average (0.039)]. CONCLUSIONS Texture analysis features showed promising results in predicting estrogen receptor status of breast cancer lesions on non-contrast T1 images.
Collapse
Affiliation(s)
- F A Shokeir
- Department of Radiology, Mansoura University, Elgomhoria St. 35516, Egypt.
| | - A H Elmokadem
- Department of Radiology, Mansoura University, Elgomhoria St. 35516, Egypt.
| | - N Soliman
- Department of Radiology, Mansoura University, Elgomhoria St. 35516, Egypt.
| | - A Khater
- Mansoura University Oncology Center, Elgomhoria St. 35516, Egypt.
| | - D Bayoumi
- Department of Radiology, Mansoura University, Elgomhoria St. 35516, Egypt.
| |
Collapse
|
8
|
Lin JY, Ye JY, Chen JG, Lin ST, Lin S, Cai SQ. Prediction of Receptor Status in Radiomics: Recent Advances in Breast Cancer Research. Acad Radiol 2024; 31:3004-3014. [PMID: 38151383 DOI: 10.1016/j.acra.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 12/29/2023]
Abstract
Breast cancer is a multifactorial heterogeneous disease and the leading cause of cancer-related deaths in women; its diagnosis and treatment require clinical sensitivity and a comprehensive disciplinary research approach. The expression of different receptors on tumor cells not only provides the basis for molecular typing of breast cancer but also has a decisive role in the diagnosis, treatment, and prognosis of breast cancer. To date, immunohistochemistry (IHC), which uses invasive histological sampling, has been extensively used in clinical practice to analyze the status of receptors and to make an accurate diagnosis of breast cancer. As an invasive assay, IHC can provide important biological information on tumors at a single point in time, but cannot predict future changes (due to treatment or tumor mutations) without additional invasive procedures. These issues highlight the need to develop a non-invasive method for predicting receptor status. The emerging field of radiomics may offer a non-invasive approach to identification of receptor status without requiring biopsy. In this paper, we present a review of the latest research results in radiomics for predicting the status of breast cancer receptors, with potential important clinical applications.
Collapse
Affiliation(s)
- Jun-Yuan Lin
- Department of Radiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.L., S.Q.C.)
| | - Jia-Yi Ye
- Department of Radiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.L., S.Q.C.)
| | - Jin-Guo Chen
- Department of Radiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.L., S.Q.C.)
| | - Shu-Ting Lin
- Department of Radiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.L., S.Q.C.)
| | - Shu Lin
- Center of Neurological and Metabolic Research, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.Y., J.G.C., S.T.L., S.L.); Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia (S.L.)
| | - Si-Qing Cai
- Department of Radiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.L., S.Q.C.).
| |
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
|
Zhou J, Yu X, Wu Q, Wu Y, Fu C, Wang Y, Hai M, Tan H, Wang M. Radiomics analysis of intratumoral and different peritumoral regions from multiparametric MRI for evaluating HER2 status of breast cancer: A comparative study. Heliyon 2024; 10:e28722. [PMID: 38623231 PMCID: PMC11016612 DOI: 10.1016/j.heliyon.2024.e28722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
Purpose To investigate the potential of radiomics signatures (RSs) from intratumoral and peritumoral regions on multiparametric magnetic resonance imaging (MRI) to noninvasively evaluate HER2 status in breast cancer. Method In this retrospective study, 992 patients with pathologically confirmed breast cancers who underwent preoperative MRI were enrolled. The breast cancer lesions were segmented manually, and the intratumor region of interest (ROIIntra) was dilated by 2, 4, 6 and 8 mm (ROIPeri2mm, ROIPeri4mm, ROIPeri6mm, and ROIPeri8mm, respectively). Quantitative radiomics features were extracted from dynamic contrast-enhanced T1-weighted imaging (DCE-T1), fat-saturated T2-weighted imaging (T2) and diffusion-weighted imaging (DWI). A three-step procedure was performed for feature selection, and RSs were constructed using a support vector machine (SVM) to predict HER2 status. Result The best single-area RSs for predicting HER2 status were DCE_Peri4mm-RS, T2_Peri4mm-RS, and DWI_Peri4mm-RS, yielding areas under the curve (AUCs) of 0.716 (95% confidence interval (CI), 0.648-0.778), 0.706 (95% CI, 0.637-0.768), and 0.719 (95% CI, 0.651-0.780), respectively, in the test set. The optimal RSs combining intratumoral and peritumoral regions for evaluating HER2 status were DCE-T1_Intra + DCE_Peri4mm-RS, T2_Intra + T2_Peri6mm-RS and DWI_Intra + DWI_Peri4mm-RS, with AUCs of 0.752 (95% CI, 0.686-0.810), 0.754 (95% CI, 0.688-0.812) and 0.725 (95% CI, 0.657-0.786), respectively, in the test set. Combining three sequences in the ROIIntra, ROIPeri2mm, ROIPeri4mm, ROIPeri6mm and ROIPeri8mm areas, the optimal RS was DCE-T1_Peri4mm + T2_Peri4mm + DWI_Peri4mm-RS, achieving an AUC of 0.795 (95% CI, 0.733-0.849) in the test set. Conclusion This study systematically explored the influence of the intratumoral region, different peritumoral sizes and their combination in radiomics analysis for predicting HER2 status in breast cancer based on multiparametric MRI and found the optimal RS.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, 450003, Henan Province, China
| | - Xuan Yu
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, 450003, Henan Province, China
| | - Qingxia Wu
- Beijing United Imaging Research Institute of Intelligent Imaging & United Imaging Intelligence (Beijing) Co., Ltd., Beijing, 100089, China
| | - Yaping Wu
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, 450003, Henan Province, China
| | - Cong Fu
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, 450003, Henan Province, China
| | - Yunxia Wang
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, 450003, Henan Province, China
| | - Menglu Hai
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, Henan Province, China
| | - Hongna Tan
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, 450003, Henan Province, China
| | - Meiyun Wang
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, 450003, Henan Province, China
| |
Collapse
|
11
|
Zhou XX, Zhang L, Cui QX, Li H, Sang XQ, Zhang HX, Zhu YM, Kuai ZX. A Channel-Dimensional Feature-Reconstructed Deep Learning Model for Predicting Breast Cancer Molecular Subtypes on Overall b-Value Diffusion-Weighted MRI. J Magn Reson Imaging 2024; 59:1425-1435. [PMID: 37403945 DOI: 10.1002/jmri.28895] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Dynamic contrast-enhanced (DCE) MRI commonly outperforms diffusion-weighted (DW) MRI in breast cancer discrimination. However, the side effects of contrast agents limit the use of DCE-MRI, particularly in patients with chronic kidney disease. PURPOSE To develop a novel deep learning model to fully exploit the potential of overall b-value DW-MRI without the need for a contrast agent in predicting breast cancer molecular subtypes and to evaluate its performance in comparison with DCE-MRI. STUDY TYPE Prospective. SUBJECTS 486 female breast cancer patients (training/validation/test: 64%/16%/20%). FIELD STRENGTH/SEQUENCE 3.0 T/DW-MRI (13 b-values) and DCE-MRI (one precontrast and five postcontrast phases). ASSESSMENT The breast cancers were divided into four subtypes: luminal A, luminal B, HER2+, and triple negative. A channel-dimensional feature-reconstructed (CDFR) deep neural network (DNN) was proposed to predict these subtypes using pathological diagnosis as the reference standard. Additionally, a non-CDFR DNN (NCDFR-DNN) was built for comparative purposes. A mixture ensemble DNN (ME-DNN) integrating two CDFR-DNNs was constructed to identify subtypes on multiparametric MRI (MP-MRI) combing DW-MRI and DCE-MRI. STATISTICAL TESTS Model performance was evaluated using accuracy, sensitivity, specificity, and area under the receiver operating characteristic curve (AUC). Model comparisons were performed using the one-way analysis of variance with least significant difference post hoc test and the DeLong test. P < 0.05 was considered significant. RESULTS The CDFR-DNN (accuracies, 0.79 ~ 0.80; AUCs, 0.93 ~ 0.94) demonstrated significantly improved predictive performance than the NCDFR-DNN (accuracies, 0.76 ~ 0.78; AUCs, 0.92 ~ 0.93) on DW-MRI. Utilizing the CDFR-DNN, DW-MRI attained the predictive performance equal (P = 0.065 ~ 1.000) to DCE-MRI (accuracies, 0.79 ~ 0.80; AUCs, 0.93 ~ 0.95). The predictive performance of the ME-DNN on MP-MRI (accuracies, 0.85 ~ 0.87; AUCs, 0.96 ~ 0.97) was superior to those of both the CDFR-DNN and NCDFR-DNN on either DW-MRI or DCE-MRI. DATA CONCLUSION The CDFR-DNN enabled overall b-value DW-MRI to achieve the predictive performance comparable to DCE-MRI. MP-MRI outperformed DW-MRI and DCE-MRI in subtype prediction. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY STAGE: 1.
Collapse
Affiliation(s)
- Xin-Xiang Zhou
- Imaging Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lan Zhang
- Imaging Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Quan-Xiang Cui
- Imaging Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hui Li
- Imaging Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xi-Qiao Sang
- Division of Respiratory Disease, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hong-Xia Zhang
- Imaging Center, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yue-Min Zhu
- CREATIS, CNRS UMR 5220-INSERM U1294-University Lyon 1-INSA Lyon-University Jean Monnet Saint-Etienne, Villeurbanne, France
| | - Zi-Xiang Kuai
- Imaging Center, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
12
|
Cai L, Sidey-Gibbons C, Nees J, Riedel F, Schaefgen B, Togawa R, Killinger K, Heil J, Pfob A, Golatta M. Ultrasound Radiomics Features to Identify Patients With Triple-Negative Breast Cancer: A Retrospective, Single-Center Study. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:467-478. [PMID: 38069582 DOI: 10.1002/jum.16377] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 11/04/2023] [Indexed: 02/08/2024]
Abstract
OBJECTIVES Patients with triple-negative breast cancer (TNBC) exhibit a fast tumor growth rate and poor survival outcomes. In this study, we aimed to develop and compare intelligent algorithms using ultrasound radiomics features in addition to clinical variables to identify patients with TNBC prior to histopathologic diagnosis. METHODS We used single-center, retrospective data of patients who underwent ultrasound before histopathologic verification and subsequent neoadjuvant systemic treatment (NAST). We developed a logistic regression with an elastic net penalty algorithm using pretreatment ultrasound radiomics features in addition to patient and tumor variables to identify patients with TNBC. Findings were compared to the histopathologic evaluation of the biopsy specimen. The main outcome measure was the area under the curve (AUC). RESULTS We included 1161 patients, 813 in the development set and 348 in the validation set. Median age was 50.1 years and 24.4% (283 of 1161) had TNBC. The integrative model using radiomics and clinical information showed significantly better performance in identifying TNBC compared to the radiomics model (AUC: 0.71, 95% confidence interval [CI]: 0.65-0.76 versus 0.64, 95% CI: 0.57-0.71, P = .004). The five most important variables were cN status, shape surface volume ratio (SA:V), gray level co-occurrence matrix (GLCM) correlation, gray level dependence matrix (GLDM) dependence nonuniformity normalized, and age. Patients with TNBC were more often categorized as BI-RADS 4 than BI-RADS 5 compared to non-TNBC patients (P = .002). CONCLUSION A machine learning algorithm showed promising potential to identify patients with TNBC using ultrasound radiomics features and clinical information prior to histopathologic evaluation.
Collapse
Affiliation(s)
- Lie Cai
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Chris Sidey-Gibbons
- MD Anderson Center for INSPiRED Cancer Care (Integrated Systems for Patient-Reported Data), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juliane Nees
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Fabian Riedel
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Benedikt Schaefgen
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Riku Togawa
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kristina Killinger
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - Joerg Heil
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| | - André Pfob
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
- MD Anderson Center for INSPiRED Cancer Care (Integrated Systems for Patient-Reported Data), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Golatta
- Department of Obstetrics and Gynecology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
13
|
Zhang W, Liang F, Zhao Y, Li J, He C, Zhao Y, Lai S, Xu Y, Ding W, Wei X, Jiang X, Yang R, Zhen X. Multiparametric MR-based feature fusion radiomics combined with ADC maps-based tumor proliferative burden in distinguishing TNBC versus non-TNBC. Phys Med Biol 2024; 69:055032. [PMID: 38306970 DOI: 10.1088/1361-6560/ad25c0] [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: 06/23/2023] [Accepted: 02/01/2024] [Indexed: 02/04/2024]
Abstract
Objective.To investigate the incremental value of quantitative stratified apparent diffusion coefficient (ADC) defined tumor habitats for differentiating triple negative breast cancer (TNBC) from non-TNBC on multiparametric MRI (mpMRI) based feature-fusion radiomics (RFF) model.Approach.466 breast cancer patients (54 TNBC, 412 non-TNBC) who underwent routine breast MRIs in our hospital were retrospectively analyzed. Radiomics features were extracted from whole tumor on T2WI, diffusion-weighted imaging, ADC maps and the 2nd phase of dynamic contrast-enhanced MRI. Four models including the RFFmodel (fused features from all MRI sequences), RADCmodel (ADC radiomics feature), StratifiedADCmodel (tumor habitas defined on stratified ADC parameters) and combinational RFF-StratifiedADCmodel were constructed to distinguish TNBC versus non-TNBC. All cases were randomly divided into a training (n= 337) and test set (n= 129). The four competing models were validated using the area under the curve (AUC), sensitivity, specificity and accuracy.Main results.Both the RFFand StratifiedADCmodels demonstrated good performance in distinguishing TNBC from non-TNBC, with best AUCs of 0.818 and 0.773 in the training and test sets. StratifiedADCmodel revealed significant different tumor habitats (necrosis/cysts habitat, chaotic habitat or proliferative tumor core) between TNBC and non-TNBC with its top three discriminative parameters (p <0.05). The integrated RFF-StratifiedADCmodel demonstrated superior accuracy over the other three models, with higher AUCs of 0.832 and 0.784 in the training and test set, respectively (p <0.05).Significance.The RFF-StratifiedADCmodel through integrating various tumor habitats' information from whole-tumor ADC maps-based StratifiedADCmodel and radiomics information from mpMRI-based RFFmodel, exhibits tremendous promise for identifying TNBC.
Collapse
Affiliation(s)
- Wanli Zhang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, People's Republic of China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Fangrong Liang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, People's Republic of China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Yue Zhao
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Jiamin Li
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, People's Republic of China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Chutong He
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, People's Republic of China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Yandong Zhao
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, People's Republic of China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Shengsheng Lai
- School of Medical Equipment, Guangdong Food and Drug Vocational College, Guangzhou, Guangdong, 510520, People's Republic of China
| | - Yongzhou Xu
- Philips Healthcare, Guangzhou, Guangdong, 510220, People's Republic of China
| | - Wenshuang Ding
- Department of Pathology, Guangzhou First People's Hospital, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Xinhua Wei
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, People's Republic of China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Xinqing Jiang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, People's Republic of China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Ruimeng Yang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, People's Republic of China
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, Guangdong, 510180, People's Republic of China
| | - Xin Zhen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| |
Collapse
|
14
|
Sang L, Liu Z, Huang C, Xu J, Wang H. Multiparametric MRI-based radiomics nomogram for predicting the hormone receptor status of HER2-positive breast cancer. Clin Radiol 2024; 79:60-66. [PMID: 37838543 DOI: 10.1016/j.crad.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/28/2023] [Accepted: 09/12/2023] [Indexed: 10/16/2023]
Abstract
AIM To investigate the value of multiparametric magnetic resonance imaging (MRI)-based radiomics nomograms for predicting the hormone receptor (HR) status of HER2-positive breast cancer. MATERIALS AND METHODS Patients with HER2-positive invasive breast cancer were divided randomly into training (68 patients) and validation (30 patients) sets. All were classified as either HR-positive (HR+) or negative (HR-) at histopathology. Two radiologists outlined the three-dimensional (3D) volumetric regions of interest (VOI) on the MRI images. Features (n=1,096) were extracted from the T2-weighted imaging (WI), apparent diffusion coefficient (ADC), and dynamic contrast-enhanced (DCE) images separately. Dimensionality was reduced using feature screening. Binary radiomics prediction models were established using a logistic regression classifier and were validated in the validation set. To construct a nomogram, independent predictors were identified using multivariate logistic regression analysis. The predictive efficacy of the model was assessed using the area under the receiver operating characteristic curve (AUC). RESULTS Ten radiomics features were obtained after feature dimensionality reduction based on the merged T2WI, ADC, and DCE images. The diagnostic efficacy of the radiomics signature using the three sequences was better than that of any single sequence (training set AUC: 0.797; validation set AUC: 0.75). Using multivariate logistic regression analysis, the independent predictors for identifying HR status were combined radiomics signature and peritumoural oedema. Nomograms constructed by combining the radiomics signature and peritumoural oedema showed good discrimination in both the training and validation sets (AUC: 0.815 and 0. 805, respectively). CONCLUSION A multiparametric MRI-based nomogram incorporating the radiomics signature and peritumoural oedema can assess the HR status of HER2-positive breast cancer. The resulting model can improve diagnostic accuracy, improving patient outcomes.
Collapse
Affiliation(s)
- L Sang
- Department of Radiology, Shandong Provincial Hospital, Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan 250012, Shandong, China
| | - Z Liu
- Department of Radiology, Shandong Provincial Hospital, Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan 250012, Shandong, China
| | - C Huang
- Department of Research Collaboration, R&D Center, Beijing Deepwise & League of, PHD Technology Co. Ltd, Beijing, China
| | - J Xu
- Department of Research Collaboration, R&D Center, Beijing Deepwise & League of, PHD Technology Co. Ltd, Beijing, China
| | - H Wang
- Department of Radiology, Shandong Provincial Hospital, Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan 250012, Shandong, China.
| |
Collapse
|
15
|
Zhang L, Fan M, Li L. Efficient estimation of pharmacokinetic parameters from breast dynamic contrast-enhanced MRI based on a convolutional neural network for predicting molecular subtypes. Phys Med Biol 2023; 68:245001. [PMID: 37983902 DOI: 10.1088/1361-6560/ad0e39] [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: 04/06/2023] [Accepted: 11/20/2023] [Indexed: 11/22/2023]
Abstract
Objective. Tracer kinetic models allow for estimating pharmacokinetic (PK) parameters, which are related to pathological characteristics, from breast dynamic contrast-enhanced magnetic resonance imaging. However, existing tracer kinetic models subject to inaccuracy are time-consuming for PK parameters estimation. This study aimed to accurately and efficiently estimate PK parameters for predicting molecular subtypes based on convolutional neural network (CNN).Approach. A CNN integrating global and local features (GL-CNN) was trained using synthetic data where known PK parameters map was used as the ground truth, and subsequently used to directly estimate PK parameters (volume transfer constantKtransand flux rate constantKep) map. The accuracy assessed by the peak signal-to-noise ratio (PSNR), structural similarity (SSIM), and concordance correlation coefficient (CCC) was compared between the GL-CNN and Tofts-based PK parameters in synthetic data. Radiomic features were calculated from the PK parameters map in 208 breast tumors. A random forest classifier was constructed to predict molecular subtypes using a discovery cohort (n= 144). The diagnostic performance evaluated on a validation cohort (n= 64) using the area under the receiver operating characteristic curve (AUC) was compared between the GL-CNN and Tofts-based PK parameters.Main results. The average PSNR (48.8884), SSIM (0.9995), and CCC (0.9995) between the GL-CNN-basedKtransmap and ground truth were significantly higher than those between the Tofts-basedKtransmap and ground truth. The GL-CNN-basedKtransobtained significantly better diagnostic performance (AUCs = 0.7658 and 0.8528) than the Tofts-basedKtransfor luminal B and HER2 tumors. The GL-CNN method accelerated the computation by speed approximately 79 times compared to the Tofts method for the whole breast of all patients.Significance. Our results indicate that the GL-CNN method can be used to accurately and efficiently estimate PK parameters for predicting molecular subtypes.
Collapse
Affiliation(s)
- Liangliang Zhang
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
- School of Computer and Information, Anqing Normal University, Anqing, 246133, People's Republic of China
| | - Ming Fan
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
| | - Lihua Li
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
| |
Collapse
|
16
|
Campana A, Gandomkar Z, Giannotti N, Reed W. The use of radiomics in magnetic resonance imaging for the pre-treatment characterisation of breast cancers: A scoping review. J Med Radiat Sci 2023; 70:462-478. [PMID: 37534540 PMCID: PMC10715343 DOI: 10.1002/jmrs.709] [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: 02/28/2023] [Accepted: 07/16/2023] [Indexed: 08/04/2023] Open
Abstract
Radiomics is an emerging field that aims to extract and analyse a comprehensive set of quantitative features from medical images. This scoping review is focused on MRI-based radiomic features for the molecular profiling of breast tumours and the implications of this work for predicting patient outcomes. A thorough systematic literature search and outcome extraction were performed to identify relevant studies published in MEDLINE/PubMed (National Centre for Biotechnology Information), EMBASE and Scopus from 2015 onwards. The following information was retrieved from each article: study purpose, study design, extracted radiomic features, machine learning technique(s), sample size/characteristics, statistical result(s) and implications on patient outcomes. Based on the study purpose, four key themes were identified in the included 63 studies: tumour subtype classification (n = 35), pathologically complete response (pCR) prediction (n = 15), lymph node metastasis (LNM) detection (n = 7) and recurrence rate prediction (n = 6). In all four themes, reported accuracies widely varied among the studies, for example, area under receiver characteristics curve (AUC) for detecting LNM ranged from 0.72 to 0.91 and the AUC for predicting pCR ranged from 0.71 to 0.99. In all four themes, combining radiomic features with clinical data improved the predictive models. Preliminary results of this study showed radiomics potential to characterise the whole tumour heterogeneity, with clear implications for individual-targeted treatment. However, radiomics is still in the pre-clinical phase, currently with an insufficient number of large multicentre studies and those existing studies are often limited by insufficient methodological transparency and standardised workflow. Consequently, the clinical translation of existing studies is currently limited.
Collapse
Affiliation(s)
- Annalise Campana
- Discipline of Medical Imaging Science, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| | - Ziba Gandomkar
- Discipline of Medical Imaging Science, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| | - Nicola Giannotti
- Discipline of Medical Imaging Science, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| | - Warren Reed
- Discipline of Medical Imaging Science, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| |
Collapse
|
17
|
Lai S, Liang F, Zhang W, Zhao Y, Li J, Zhao Y, Xu Y, Ding W, Zhan J, Zhen X, Yang R. Evaluation of molecular receptors status in breast cancer using an mpMRI-based feature fusion radiomics model: mimicking radiologists' diagnosis. Front Oncol 2023; 13:1219071. [PMID: 38074664 PMCID: PMC10698551 DOI: 10.3389/fonc.2023.1219071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 10/31/2023] [Indexed: 08/31/2024] Open
Abstract
OBJECTIVE To investigate the performance of a novel feature fusion radiomics (RFF) model that incorporates features from multiparametric MRIs (mpMRI) in distinguishing different statuses of molecular receptors in breast cancer (BC) preoperatively. METHODS 460 patients with 466 pathology-confirmed BCs who underwent breast mpMRI at 1.5T in our center were retrospectively included hormone receptor (HR) positive (HR+) (n=336) and HR negative (HR-) (n=130). The HR- patients were further categorized into human epidermal growth factor receptor 2 (HER-2) enriched BC (HEBC) (n=76) and triple negative BC (TNBC) (n=54). All lesions were divided into a training/validation cohort (n=337) and a test cohort (n=129). Volumes of interest (VOIs) delineation, followed by radiomics feature extraction, was performed on T2WI, DWI600 (b=600 s/mm2), DWI800 (b=800 s/mm2), ADC map, and DCE1-6 (six continuous DCE-MRI) images of each lesion. Simulating a radiologist's work pattern, 150 classification base models were constructed and analyzed to determine the top four optimum sequences for classifying HR+ vs. HR-, TNBC vs. HEBC, TNBC vs. non-TNBC in a random selected training cohort (n=337). Building upon these findings, the optimal single sequence models (Rss) and combined sequences models (RFF) were developed. The AUC, sensitivity, accuracy and specificity of each model for subtype differentiation were evaluated. The paired samples Wilcoxon signed rank test was used for performance comparison. RESULTS During the three classification tasks, the optimal single sequence for classifying HR+ vs. HR- was DWI600, while the ADC map, derived from DWI800 performed the best in distinguishing TNBC vs. HEBC, as well as identifying TNBC vs. non-TNBC, with corresponding training AUC values of 0.787, 0.788, and 0.809, respectively. Furthermore, the integration of the top four sequences in RFF models yielded improved performance, achieving AUC values of 0.809, 0.805 and 0.847, respectively. Consistent results was observed in both the training/validation and testing cohorts, with AUC values of 0.778, 0.787, 0.818 and 0.726, 0.773, 0.773, respectively (all p < 0.05 except HR+ vs. HR-). CONCLUSION The RFF model, integrating mpMRI radiomics features, demonstrated promising ability to mimic radiologists' diagnosis for preoperative identification of molecular receptors of BC.
Collapse
Affiliation(s)
- Shengsheng Lai
- School of Medical Equipment, Guangdong Food and Drug Vocational College, Guangzhou, Guangdong, China
| | - Fangrong Liang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Wanli Zhang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Yue Zhao
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Jiamin Li
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Yandong Zhao
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Yongzhou Xu
- Department of Clinical & Technique Support, Philips Healthcare, Guangzhou, Guangdong, China
| | - Wenshuang Ding
- Department of Pathology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Jie Zhan
- Department of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xin Zhen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
| | - Ruimeng Yang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
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: 5.0] [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
|
19
|
Panthi B, Mohamed RM, Adrada BE, Boge M, Candelaria RP, Chen H, Hunt KK, Huo L, Hwang KP, Korkut A, Lane DL, Le-Petross HC, Leung JWT, Litton JK, Pashapoor S, Perez F, Son JB, Sun J, Thompson A, Tripathy D, Valero V, Wei P, White J, Xu Z, Yang W, Zhou Z, Yam C, Rauch GM, Ma J. Longitudinal dynamic contrast-enhanced MRI radiomic models for early prediction of response to neoadjuvant systemic therapy in triple-negative breast cancer. Front Oncol 2023; 13:1264259. [PMID: 37941561 PMCID: PMC10628525 DOI: 10.3389/fonc.2023.1264259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Early prediction of neoadjuvant systemic therapy (NAST) response for triple-negative breast cancer (TNBC) patients could help oncologists select individualized treatment and avoid toxic effects associated with ineffective therapy in patients unlikely to achieve pathologic complete response (pCR). The objective of this study is to evaluate the performance of radiomic features of the peritumoral and tumoral regions from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) acquired at different time points of NAST for early treatment response prediction in TNBC. This study included 163 Stage I-III patients with TNBC undergoing NAST as part of a prospective clinical trial (NCT02276443). Peritumoral and tumoral regions of interest were segmented on DCE images at baseline (BL) and after two (C2) and four (C4) cycles of NAST. Ten first-order (FO) radiomic features and 300 gray-level-co-occurrence matrix (GLCM) features were calculated. Area under the receiver operating characteristic curve (AUC) and Wilcoxon rank sum test were used to determine the most predictive features. Multivariate logistic regression models were used for performance assessment. Pearson correlation was used to assess intrareader and interreader variability. Seventy-eight patients (48%) had pCR (52 training, 26 testing), and 85 (52%) had non-pCR (57 training, 28 testing). Forty-six radiomic features had AUC at least 0.70, and 13 multivariate models had AUC at least 0.75 for training and testing sets. The Pearson correlation showed significant correlation between readers. In conclusion, Radiomic features from DCE-MRI are useful for differentiating pCR and non-pCR. Similarly, predictive radiomic models based on these features can improve early noninvasive treatment response prediction in TNBC patients undergoing NAST.
Collapse
Affiliation(s)
- Bikash Panthi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rania M. Mohamed
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Beatriz E. Adrada
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Medine Boge
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Koc University Hospital, Istanbul, Türkiye
| | - Rosalind P. Candelaria
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Huiqin Chen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kelly K. Hunt
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ken-Pin Hwang
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Deanna L. Lane
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Huong C. Le-Petross
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jessica W. T. Leung
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jennifer K. Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sanaz Pashapoor
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Frances Perez
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jong Bum Son
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jia Sun
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alastair Thompson
- Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jason White
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zhan Xu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Wei Yang
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zijian Zhou
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Clinton Yam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gaiane M. Rauch
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
20
|
Yue WY, Zhang HT, Gao S, Li G, Sun ZY, Tang Z, Cai JM, Tian N, Zhou J, Dong JH, Liu Y, Bai X, Sheng FG. Predicting Breast Cancer Subtypes Using Magnetic Resonance Imaging Based Radiomics With Automatic Segmentation. J Comput Assist Tomogr 2023; 47:729-737. [PMID: 37707402 PMCID: PMC10510832 DOI: 10.1097/rct.0000000000001474] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/02/2023] [Indexed: 05/21/2023]
Abstract
OBJECTIVE The aim of the study is to demonstrate whether radiomics based on an automatic segmentation method is feasible for predicting molecular subtypes. METHODS This retrospective study included 516 patients with confirmed breast cancer. An automatic segmentation-3-dimensional UNet-based Convolutional Neural Networks, trained on our in-house data set-was applied to segment the regions of interest. A set of 1316 radiomics features per region of interest was extracted. Eighteen cross-combination radiomics methods-with 6 feature selection methods and 3 classifiers-were used for model selection. Model classification performance was assessed using the area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, and specificity. RESULTS The average dice similarity coefficient value of the automatic segmentation was 0.89. The radiomics models were predictive of 4 molecular subtypes with the best average: AUC = 0.8623, accuracy = 0.6596, sensitivity = 0.6383, and specificity = 0.8775. For luminal versus nonluminal subtypes, AUC = 0.8788 (95% confidence interval [CI], 0.8505-0.9071), accuracy = 0.7756, sensitivity = 0.7973, and specificity = 0.7466. For human epidermal growth factor receptor 2 (HER2)-enriched versus non-HER2-enriched subtypes, AUC = 0.8676 (95% CI, 0.8370-0.8982), accuracy = 0.7737, sensitivity = 0.8859, and specificity = 0.7283. For triple-negative breast cancer versus non-triple-negative breast cancer subtypes, AUC = 0.9335 (95% CI, 0.9027-0.9643), accuracy = 0.9110, sensitivity = 0.4444, and specificity = 0.9865. CONCLUSIONS Radiomics based on automatic segmentation of magnetic resonance imaging can predict breast cancer of 4 molecular subtypes noninvasively and is potentially applicable in large samples.
Collapse
Affiliation(s)
- Wen-Yi Yue
- From the Fifth Medical Center of Chinese PLA General Hospital
- Chinese PLA General Medical School
| | - Hong-Tao Zhang
- From the Fifth Medical Center of Chinese PLA General Hospital
| | - Shen Gao
- From the Fifth Medical Center of Chinese PLA General Hospital
| | - Guang Li
- Keya Medical Technology Co, Ltd, Beijing, China
| | - Ze-Yu Sun
- Keya Medical Technology Co, Ltd, Beijing, China
| | - Zhe Tang
- Keya Medical Technology Co, Ltd, Beijing, China
| | - Jian-Ming Cai
- From the Fifth Medical Center of Chinese PLA General Hospital
| | - Ning Tian
- From the Fifth Medical Center of Chinese PLA General Hospital
| | - Juan Zhou
- From the Fifth Medical Center of Chinese PLA General Hospital
| | - Jing-Hui Dong
- From the Fifth Medical Center of Chinese PLA General Hospital
| | - Yuan Liu
- From the Fifth Medical Center of Chinese PLA General Hospital
| | - Xu Bai
- From the Fifth Medical Center of Chinese PLA General Hospital
| | - Fu-Geng Sheng
- From the Fifth Medical Center of Chinese PLA General Hospital
| |
Collapse
|
21
|
Cho S, Joo B, Park M, Ahn SJ, Suh SH, Park YW, Ahn SS, Lee SK. A Radiomics-Based Model for Potentially More Accurate Identification of Subtypes of Breast Cancer Brain Metastases. Yonsei Med J 2023; 64:573-580. [PMID: 37634634 PMCID: PMC10462808 DOI: 10.3349/ymj.2023.0047] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 08/29/2023] Open
Abstract
PURPOSE Breast cancer brain metastases (BCBM) may involve subtypes that differ from the primary breast cancer lesion. This study aimed to develop a radiomics-based model that utilizes preoperative brain MRI for multiclass classification of BCBM subtypes and to investigate whether the model offers better prediction accuracy than the assumption that primary lesions and their BCBMs would be of the same subtype (non-conversion model) in an external validation set. MATERIALS AND METHODS The training and external validation sets each comprised 51 cases (102 cases total). Four machine learning classifiers combined with three feature selection methods were trained on radiomic features and primary lesion subtypes for prediction of the following four subtypes: 1) hormone receptor (HR)+/human epidermal growth factor receptor 2 (HER2)-, 2) HR+/HER2+, 3) HR-/HER2+, and 4) triple-negative. After training, the performance of the radiomics-based model was compared to that of the non-conversion model in an external validation set using accuracy and F1-macro scores. RESULTS The rate of discrepant subtypes between primary lesions and their respective BCBMs were 25.5% (n=13 of 51) in the training set and 23.5% (n=12 of 51) in the external validation set. In the external validation set, the accuracy and F1-macro score of the radiomics-based model were significantly higher than those of the non-conversion model (0.902 vs. 0.765, p=0.004; 0.861 vs. 0.699, p=0.002). CONCLUSION Our radiomics-based model represents an incremental advance in the classification of BCBM subtypes, thereby facilitating a more appropriate personalized therapy.
Collapse
Affiliation(s)
- Seonghyeon Cho
- Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Bio Joo
- Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| | - Mina Park
- Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Jun Ahn
- Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Hyun Suh
- Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yae Won Park
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Image Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Image Data Science, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Image Data Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Kovačević L, Štajduhar A, Stemberger K, Korša L, Marušić Z, Prutki M. Breast Cancer Surrogate Subtype Classification Using Pretreatment Multi-Phase Dynamic Contrast-Enhanced Magnetic Resonance Imaging Radiomics: A Retrospective Single-Center Study. J Pers Med 2023; 13:1150. [PMID: 37511763 PMCID: PMC10381456 DOI: 10.3390/jpm13071150] [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: 04/19/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
This study aimed to explore the potential of multi-phase dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) radiomics for classifying breast cancer surrogate subtypes. This retrospective study analyzed 360 breast cancers from 319 patients who underwent pretreatment DCE-MRI between January 2015 and January 2019. The cohort consisted of 33 triple-negative, 26 human epidermal growth factor receptor 2 (HER2)-positive, 109 luminal A-like, 144 luminal B-like HER2-negative, and 48 luminal B-like HER2-positive lesions. A total of 1781 radiomic features were extracted from manually segmented breast cancers in each DCE-MRI sequence. The model was internally validated and selected using ten times repeated five-fold cross-validation on the primary cohort, with further evaluation using a validation cohort. The most successful models were logistic regression models applied to the third post-contrast subtraction images. These models exhibited the highest area under the curve (AUC) for discriminating between luminal A like vs. others (AUC: 0.78), luminal B-like HER2 negative vs. others (AUC: 0.57), luminal B-like HER2 positive vs. others (AUC: 0.60), HER2 positive vs. others (AUC: 0.81), and triple negative vs. others (AUC: 0.83). In conclusion, the radiomic features extracted from multi-phase DCE-MRI are promising for discriminating between breast cancer subtypes. The best-performing models relied on tissue changes observed during the mid-stage of the imaging process.
Collapse
Affiliation(s)
- Lucija Kovačević
- Clinical Department of Diagnostic and Interventional Radiology, University Hospital Centre Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia; (L.K.); (M.P.)
| | - Andrija Štajduhar
- Department for Medical Statistics, Epidemiology and Medical Informatics School of Medicine, University of Zagreb, Salata 12, 10000 Zagreb, Croatia
| | - Karlo Stemberger
- Clinical Department of Diagnostic and Interventional Radiology, University Hospital Centre Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia; (L.K.); (M.P.)
| | - Lea Korša
- Clinical Department of Pathology and Cytology, University Hospital Centre Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia
| | - Zlatko Marušić
- Clinical Department of Pathology and Cytology, University Hospital Centre Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia
| | - Maja Prutki
- Clinical Department of Diagnostic and Interventional Radiology, University Hospital Centre Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia; (L.K.); (M.P.)
- School of Medicine, University of Zagreb, Salata 3, 10000 Zagreb, Croatia
| |
Collapse
|
23
|
Zhou J, Xie T, Shan H, Cheng G. HLA-DQA1 expression is associated with prognosis and predictable with radiomics in breast cancer. Radiat Oncol 2023; 18:117. [PMID: 37434241 DOI: 10.1186/s13014-023-02314-4] [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: 11/11/2022] [Accepted: 07/05/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND High HLA-DQA1 expression is associated with a better prognosis in many cancers. However, the association between HLA-DQA1 expression and prognosis of breast cancer and the noninvasive assessment of HLA-DQA1 expression are still unclear. This study aimed to reveal the association and investigate the potential of radiomics to predict HLA-DQA1 expression in breast cancer. METHODS In this retrospective study, transcriptome sequencing data, medical imaging data, clinical and follow-up data were downloaded from the TCIA ( https://www.cancerimagingarchive.net/ ) and TCGA ( https://portal.gdc.cancer.gov/ ) databases. The clinical characteristic differences between the high HLA-DQA1 expression group (HHD group) and the low HLA-DQA1 expression group were explored. Gene set enrichment analysis, Kaplan‒Meier survival analysis and Cox regression were performed. Then, 107 dynamic contrast-enhanced magnetic resonance imaging features were extracted, including size, shape and texture. Using recursive feature elimination and gradient boosting machine, a radiomics model was established to predict HLA-DQA1 expression. Receiver operating characteristic (ROC) curves, precision-recall curves, calibration curves, and decision curves were used for model evaluation. RESULTS The HHD group had better survival outcomes. The differentially expressed genes in the HHD group were significantly enriched in oxidative phosphorylation (OXPHOS) and estrogen response early and late signalling pathways. The radiomic score (RS) output from the model was associated with HLA-DQA1 expression. The area under the ROC curves (95% CI), accuracy, sensitivity, specificity, positive predictive value, and negative predictive value of the radiomic model were 0.866 (0.775-0.956), 0.825, 0.939, 0.7, 0.775, and 0.913 in the training set and 0.780 (0.629-0.931), 0.659, 0.81, 0.5, 0.63, and 0.714 in the validation set, respectively, showing a good prediction effect. CONCLUSIONS High HLA-DQA1 expression is associated with a better prognosis in breast cancer. Quantitative radiomics as a noninvasive imaging biomarker has potential value for predicting HLA-DQA1 expression.
Collapse
Affiliation(s)
- JingYu Zhou
- Department of Radiology, Peking University Shenzhen Hospital, LianHua Road, Shenzhen, 518000, Guangdong, China
| | - TingTing Xie
- Department of Radiology, Peking University Shenzhen Hospital, LianHua Road, Shenzhen, 518000, Guangdong, China
| | - HuiMing Shan
- Department of Radiology, Peking University Shenzhen Hospital, LianHua Road, Shenzhen, 518000, Guangdong, China
| | - GuanXun Cheng
- Department of Radiology, Peking University Shenzhen Hospital, LianHua Road, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
24
|
Hwang KP, Elshafeey NA, Kotrotsou A, Chen H, Son JB, Boge M, Mohamed RM, Abdelhafez AH, Adrada BE, Panthi B, Sun J, Musall BC, Zhang S, Candelaria RP, White JB, Ravenberg EE, Tripathy D, Yam C, Litton JK, Huo L, Thompson AM, Wei P, Yang WT, Pagel MD, Ma J, Rauch GM. A Radiomics Model Based on Synthetic MRI Acquisition for Predicting Neoadjuvant Systemic Treatment Response in Triple-Negative Breast Cancer. Radiol Imaging Cancer 2023; 5:e230009. [PMID: 37505106 PMCID: PMC10413296 DOI: 10.1148/rycan.230009] [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: 02/08/2023] [Revised: 04/18/2023] [Accepted: 06/03/2023] [Indexed: 07/29/2023]
Abstract
Purpose To determine if a radiomics model based on quantitative maps acquired with synthetic MRI (SyMRI) is useful for predicting neoadjuvant systemic therapy (NAST) response in triple-negative breast cancer (TNBC). Materials and Methods In this prospective study, 181 women diagnosed with stage I-III TNBC were scanned with a SyMRI sequence at baseline and at midtreatment (after four cycles of NAST), producing T1, T2, and proton density (PD) maps. Histopathologic analysis at surgery was used to determine pathologic complete response (pCR) or non-pCR status. From three-dimensional tumor contours drawn on the three maps, 310 histogram and textural features were extracted, resulting in 930 features per scan. Radiomic features were compared between pCR and non-pCR groups by using Wilcoxon rank sum test. To build a multivariable predictive model, logistic regression with elastic net regularization and cross-validation was performed for texture feature selection using 119 participants (median age, 52 years [range, 26-77 years]). An independent testing cohort of 62 participants (median age, 48 years [range, 23-74 years]) was used to evaluate and compare the models by area under the receiver operating characteristic curve (AUC). Results Univariable analysis identified 15 T1, 10 T2, and 12 PD radiomic features at midtreatment that predicted pCR with an AUC greater than 0.70 in both the training and testing cohorts. Multivariable radiomics models of maps acquired at midtreatment demonstrated superior performance over those acquired at baseline, achieving AUCs as high as 0.78 and 0.72 in the training and testing cohorts, respectively. Conclusion SyMRI-based radiomic features acquired at midtreatment are potentially useful for identifying early NAST responders in TNBC. Keywords: MR Imaging, Breast, Outcomes Analysis ClinicalTrials.gov registration no. NCT02276443 Supplemental material is available for this article. © RSNA, 2023 See also the commentary by Houser and Rapelyea in this issue.
Collapse
Affiliation(s)
- Ken-Pin Hwang
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Nabil A. Elshafeey
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Aikaterini Kotrotsou
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Huiqin Chen
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Jong Bum Son
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Medine Boge
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Rania M. Mohamed
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Abeer H. Abdelhafez
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Beatriz E. Adrada
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Bikash Panthi
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Jia Sun
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Benjamin C. Musall
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Shu Zhang
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Rosalind P. Candelaria
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Jason B. White
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Elizabeth E. Ravenberg
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Debu Tripathy
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Clinton Yam
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Jennifer K. Litton
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Lei Huo
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Alastair M. Thompson
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Peng Wei
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Wei T. Yang
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Mark D. Pagel
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Jingfei Ma
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Gaiane M. Rauch
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| |
Collapse
|
25
|
Park AY, Han MR, Seo BK, Ju HY, Son GS, Lee HY, Chang YW, Choi J, Cho KR, Song SE, Woo OH, Park HS. MRI-based breast cancer radiogenomics using RNA profiling: association with subtypes in a single-center prospective study. Breast Cancer Res 2023; 25:79. [PMID: 37391754 PMCID: PMC10311893 DOI: 10.1186/s13058-023-01668-7] [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: 01/11/2023] [Accepted: 05/31/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND There are few prospective studies on the correlations between MRI features and whole RNA-sequencing data in breast cancer according to molecular subtypes. The purpose of our study was to explore the association between genetic profiles and MRI phenotypes of breast cancer and to identify imaging markers that influences the prognosis and treatment according to subtypes. METHODS From June 2017 to August 2018, MRIs of 95 women with invasive breast cancer were prospectively analyzed, using the breast imaging-reporting and data system and texture analysis. Whole RNA obtained from surgical specimens was analyzed using next-generation sequencing. The association between MRI features and gene expression profiles was analyzed in the entire tumor and subtypes. Gene networks, enriched functions, and canonical pathways were analyzed using Ingenuity Pathway Analysis. The P value for differential expression was obtained using a parametric F test comparing nested linear models and adjusted for multiple testing by reporting Q value. RESULTS In 95 participants (mean age, 53 years ± 11 [standard deviation]), mass lesion type was associated with upregulation of CCL3L1 (sevenfold) and irregular mass shape was associated with downregulation of MIR421 (sixfold). In estrogen receptor-positive cancer with mass lesion type, CCL3L1 (21-fold), SNHG12 (11-fold), and MIR206 (sevenfold) were upregulated, and MIR597 (265-fold), MIR126 (12-fold), and SOX17 (fivefold) were downregulated. In triple-negative breast cancer with increased standard deviation of texture analysis on precontrast T1-weighted imaging, CLEC3A (23-fold), SRGN (13-fold), HSPG2 (sevenfold), KMT2D (fivefold), and VMP1 (fivefold) were upregulated, and IGLC2 (73-fold) and PRDX4 (sevenfold) were downregulated (all, P < 0.05 and Q < 0.1). Gene network and functional analysis showed that mass type estrogen receptor-positive cancers were associated with cell growth, anti-estrogen resistance, and poor survival. CONCLUSION MRI characteristics are associated with the different expressions of genes related to metastasis, anti-drug resistance, and prognosis, depending on the molecular subtypes of breast cancer.
Collapse
Affiliation(s)
- Ah Young Park
- Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Mi-Ryung Han
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Bo Kyoung Seo
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan City, Gyeonggi-do, 15355, Republic of Korea.
| | - Hye-Yeon Ju
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Gil Soo Son
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan City, Gyeonggi-do, Republic of Korea
| | - Hye Yoon Lee
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan City, Gyeonggi-do, Republic of Korea
| | - Young Woo Chang
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan City, Gyeonggi-do, Republic of Korea
| | - Jungyoon Choi
- Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan City, Gyeonggi-do, Republic of Korea
| | - Kyu Ran Cho
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sung Eun Song
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ok Hee Woo
- Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyun Soo Park
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan City, Gyeonggi-do, 15355, Republic of Korea
| |
Collapse
|
26
|
Teng X, Zhang J, Zhang X, Fan X, Zhou T, Huang YH, Wang L, Lee EYP, Yang R, Cai J. Noninvasive imaging signatures of HER2 and HR using ADC in invasive breast cancer: repeatability, reproducibility, and association with pathological complete response to neoadjuvant chemotherapy. Breast Cancer Res 2023; 25:77. [PMID: 37381020 DOI: 10.1186/s13058-023-01674-9] [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: 02/10/2023] [Accepted: 06/13/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND The immunohistochemical test (IHC) of HER2 and HR can provide prognostic information and treatment guidance for invasive breast cancer patients. We aimed to develop noninvasive image signatures ISHER2 and ISHR of HER2 and HR, respectively. We independently evaluate their repeatability, reproducibility, and association with pathological complete response (pCR) to neoadjuvant chemotherapy. METHODS Pre-treatment DWI, IHC receptor status HER2/HR, and pCR to neoadjuvant chemotherapy of 222 patients from the multi-institutional ACRIN 6698 trial were retrospectively collected. They were pre-separated for development, independent validation, and test-retest. 1316 image features were extracted from DWI-derived ADC maps within manual tumor segmentations. ISHER2 and ISHR were developed by RIDGE logistic regression using non-redundant and test-retest reproducible features relevant to IHC receptor status. We evaluated their association with pCR using area under receiver operating curve (AUC) and odds ratio (OR) after binarization. Their reproducibility was further evaluated using the test-retest set with intra-class coefficient of correlation (ICC). RESULTS A 5-feature ISHER2 targeting HER2 was developed (AUC = 0.70, 95% CI 0.59 to 0.82) and validated (AUC = 0.72, 95% CI 0.58 to 0.86) with high perturbation repeatability (ICC = 0.92) and test-retest reproducibility (ICC = 0.83). ISHR was developed using 5 features with higher association with HR during development (AUC = 0.75, 95% CI 0.66 to 0.84) and validation (AUC = 0.74, 95% CI 0.61 to 0.86) and similar repeatability (ICC = 0.91) and reproducibility (ICC = 0.82). Both image signatures showed significant associations with pCR with AUC of 0.65 (95% CI 0.50 to 0.80) for ISHER2 and 0.64 (95% CI 0.50 to 0.78) for ISHER2 in the validation cohort. Patients with high ISHER2 were more likely to achieve pCR to neoadjuvant chemotherapy with validation OR of 4.73 (95% CI 1.64 to 13.65, P value = 0.006). Low ISHR patients had higher pCR with OR = 0.29 (95% CI 0.10 to 0.81, P value = 0.021). Molecular subtypes derived from the image signatures showed comparable pCR prediction values to IHC-based molecular subtypes (P value > 0.05). CONCLUSION Robust ADC-based image signatures were developed and validated for noninvasive evaluation of IHC receptors HER2 and HR. We also confirmed their value in predicting treatment response to neoadjuvant chemotherapy. Further evaluations in treatment guidance are warranted to fully validate their potential as IHC surrogates.
Collapse
Affiliation(s)
- Xinzhi Teng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Xinyu Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Xinyu Fan
- Department of Applied Mathematics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Ta Zhou
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Yu-Hua Huang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Lu Wang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Elaine Yuen Phin Lee
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Y920, Lee Shau Kee Building, Hong Kong, China
| | - Ruijie Yang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China.
- The Hong Kong Polytechnic University Shenzhen Research Institute, Hong Kong, China.
- Research Institute for Smart Aging, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
27
|
Ji Y, Whitney HM, Li H, Liu P, Giger ML, Zhang X. Differences in Molecular Subtype Reference Standards Impact AI-based Breast Cancer Classification with Dynamic Contrast-enhanced MRI. Radiology 2023; 307:e220984. [PMID: 36594836 PMCID: PMC10068887 DOI: 10.1148/radiol.220984] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 01/04/2023]
Abstract
Background Breast cancer tumors can be identified as different luminal molecular subtypes depending on either immunohistochemical (IHC) staining or St Gallen criteria that includes Ki-67. Purpose To characterize molecular subtypes and understand the impact of disagreement among IHC and St Gallen molecular subtype reference standards on artificial intelligence classification of luminal A and luminal B tumors with use of radiomic features extracted from dynamic contrast-enhanced (DCE) MRI scans. Materials and Methods In this retrospective study, 28 radiomic features previously extracted from DCE-MRI scans of breast tumors imaged between February 2015 and October 2017 were examined in the following groups: (a) tumors classified as luminal A by both reference standards ("agreement"), (b) tumors classified as luminal A by IHC and luminal B by St Gallen ("disagreement"), and (c) tumors classified as luminal B by both ("agreement"). Luminal A or luminal B tumor classification with use of radiomic features was conducted with use of three sets: (a) IHC molecular subtyping, (b) St Gallen molecular subtyping, and (c) agreement tumors. The Kruskal-Wallis test was followed by the Mann-Whitney U test to determine pair-wise differences of radiomic features among agreement and disagreement tumors. Fivefold cross-validation with use of stepwise feature selection and linear discriminant analysis classified tumors in each set, with performance measured with use of area under the receiver operating characteristic curve (AUC). Results A total of 877 breast cancer tumors from 872 women (mean age, 48 years [range, 19-75 years]) were analyzed. Six features (sphericity, irregularity, surface area to volume ratio, variance of radial gradient histogram, sum average, volume of most enhancing voxels) were different (P ≤ .001) among agreement and disagreement tumors. AUC (median, 0.74 [95% CI: 0.68, 0.80]) was higher than when using tumors subtyped by either reference standard (IHC, 0.66 [0.60, 0.71], P = .003; St Gallen, 0.62 [0.58, 0.67], P = .001). Conclusion Differences in reference standards can hinder artificial intelligence classification performance of luminal molecular subtypes with dynamic contrast-enhanced MRI. © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Bae in this issue.
Collapse
Affiliation(s)
- Yu Ji
- From the Department of Radiology, The Second Hospital of Tianjin
Medical University, No. 23 Pingjiang Rd, Hexi District, Tianjin, China 300211
(Y.J., X.Z.); National Clinical Research Center for Cancer, Tianjin Medical
University Cancer Institute and Hospital, Tianjin, China (Y.J., P.L.);
Department of Radiology, The University of Chicago, Chicago, Ill (H.M.W., H.L.,
M.L.G.); and Department of Physics, Wheaton College, Wheaton, Ill
(H.M.W.)
| | - Heather M. Whitney
- From the Department of Radiology, The Second Hospital of Tianjin
Medical University, No. 23 Pingjiang Rd, Hexi District, Tianjin, China 300211
(Y.J., X.Z.); National Clinical Research Center for Cancer, Tianjin Medical
University Cancer Institute and Hospital, Tianjin, China (Y.J., P.L.);
Department of Radiology, The University of Chicago, Chicago, Ill (H.M.W., H.L.,
M.L.G.); and Department of Physics, Wheaton College, Wheaton, Ill
(H.M.W.)
| | - Hui Li
- From the Department of Radiology, The Second Hospital of Tianjin
Medical University, No. 23 Pingjiang Rd, Hexi District, Tianjin, China 300211
(Y.J., X.Z.); National Clinical Research Center for Cancer, Tianjin Medical
University Cancer Institute and Hospital, Tianjin, China (Y.J., P.L.);
Department of Radiology, The University of Chicago, Chicago, Ill (H.M.W., H.L.,
M.L.G.); and Department of Physics, Wheaton College, Wheaton, Ill
(H.M.W.)
| | - Peifang Liu
- From the Department of Radiology, The Second Hospital of Tianjin
Medical University, No. 23 Pingjiang Rd, Hexi District, Tianjin, China 300211
(Y.J., X.Z.); National Clinical Research Center for Cancer, Tianjin Medical
University Cancer Institute and Hospital, Tianjin, China (Y.J., P.L.);
Department of Radiology, The University of Chicago, Chicago, Ill (H.M.W., H.L.,
M.L.G.); and Department of Physics, Wheaton College, Wheaton, Ill
(H.M.W.)
| | - Maryellen L. Giger
- From the Department of Radiology, The Second Hospital of Tianjin
Medical University, No. 23 Pingjiang Rd, Hexi District, Tianjin, China 300211
(Y.J., X.Z.); National Clinical Research Center for Cancer, Tianjin Medical
University Cancer Institute and Hospital, Tianjin, China (Y.J., P.L.);
Department of Radiology, The University of Chicago, Chicago, Ill (H.M.W., H.L.,
M.L.G.); and Department of Physics, Wheaton College, Wheaton, Ill
(H.M.W.)
| | - Xuening Zhang
- From the Department of Radiology, The Second Hospital of Tianjin
Medical University, No. 23 Pingjiang Rd, Hexi District, Tianjin, China 300211
(Y.J., X.Z.); National Clinical Research Center for Cancer, Tianjin Medical
University Cancer Institute and Hospital, Tianjin, China (Y.J., P.L.);
Department of Radiology, The University of Chicago, Chicago, Ill (H.M.W., H.L.,
M.L.G.); and Department of Physics, Wheaton College, Wheaton, Ill
(H.M.W.)
| |
Collapse
|
28
|
Zhang T, Tan T, Han L, Appelman L, Veltman J, Wessels R, Duvivier KM, Loo C, Gao Y, Wang X, Horlings HM, Beets-Tan RGH, Mann RM. Predicting breast cancer types on and beyond molecular level in a multi-modal fashion. NPJ Breast Cancer 2023; 9:16. [PMID: 36949047 PMCID: PMC10033710 DOI: 10.1038/s41523-023-00517-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 02/21/2023] [Indexed: 03/24/2023] Open
Abstract
Accurately determining the molecular subtypes of breast cancer is important for the prognosis of breast cancer patients and can guide treatment selection. In this study, we develop a deep learning-based model for predicting the molecular subtypes of breast cancer directly from the diagnostic mammography and ultrasound images. Multi-modal deep learning with intra- and inter-modality attention modules (MDL-IIA) is proposed to extract important relations between mammography and ultrasound for this task. MDL-IIA leads to the best diagnostic performance compared to other cohort models in predicting 4-category molecular subtypes with Matthews correlation coefficient (MCC) of 0.837 (95% confidence interval [CI]: 0.803, 0.870). The MDL-IIA model can also discriminate between Luminal and Non-Luminal disease with an area under the receiver operating characteristic curve of 0.929 (95% CI: 0.903, 0.951). These results significantly outperform clinicians' predictions based on radiographic imaging. Beyond molecular-level test, based on gene-level ground truth, our method can bypass the inherent uncertainty from immunohistochemistry test. This work thus provides a noninvasive method to predict the molecular subtypes of breast cancer, potentially guiding treatment selection for breast cancer patients and providing decision support for clinicians.
Collapse
Affiliation(s)
- Tianyu Zhang
- Department of Radiology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- GROW School for Oncology and Development Biology, Maastricht University, P. O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Diagnostic Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Tao Tan
- Department of Radiology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
- Faculty of Applied Sciences, Macao Polytechnic University, 999078, Macao SAR, China.
| | - Luyi Han
- Department of Radiology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Diagnostic Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Linda Appelman
- Department of Diagnostic Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Jeroen Veltman
- Department of Radiology, Hospital Group Twente (ZGT), Almelo, The Netherlands
- Multi-Modality Medical Imaging Group, TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Ronni Wessels
- Department of Radiology, Haga Teaching Hospital, The Hague, The Netherlands
| | - Katya M Duvivier
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Claudette Loo
- Department of Radiology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Yuan Gao
- Department of Radiology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- GROW School for Oncology and Development Biology, Maastricht University, P. O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Diagnostic Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Xin Wang
- Department of Radiology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- GROW School for Oncology and Development Biology, Maastricht University, P. O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Diagnostic Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Hugo M Horlings
- Division of Pathology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Regina G H Beets-Tan
- Department of Radiology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- GROW School for Oncology and Development Biology, Maastricht University, P. O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Ritse M Mann
- Department of Radiology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Diagnostic Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
29
|
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: 1.0] [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
|
30
|
Lafcı O, Celepli P, Seher Öztekin P, Koşar PN. DCE-MRI Radiomics Analysis in Differentiating Luminal A and Luminal B Breast Cancer Molecular Subtypes. Acad Radiol 2023; 30:22-29. [PMID: 35595629 DOI: 10.1016/j.acra.2022.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 12/26/2022]
Abstract
RATIONALE AND OBJECTIVES The aim of the present study was to investigate the association between Luminal A and Luminal B molecular subtypes and radiomic features of dynamic contrast‑enhanced magnetic resonance imaging in patients with invasive breast cancer. MATERIALS AND METHODS Seventy-three patients with histopathologically proven invasive ductal cancer (IDC) were selected. Tumors were classified into molecular subtypes: Luminal A (estrogen receptor (ER)-positive and/or progesterone receptor (PR)-positive, human epidermal growth factor receptor type 2 (HER2) -negative, proliferation marker Ki-67<20) and Luminal B (ER-positive and/or PR-positive, HER2-positive or HER2-negative with high Ki-67 ≥20). A total of 81 tumoral lesions were evaluated on T1-weighted fat-suppressed sagittal post-contrast late-phase MRI images after the required "pre-process" steps and 3D segmentations were made. Forty-three radiomic features including: 1 conventional, 4 shape, 6 histogram, 7 Grey-Level Co-occurrence Matrix (GLCM), 11 Grey-Level Run-Length Matrix (GLRLM), 3 Neighborhood Grey-Level Difference Matrix (NGLDM), 11 Grey-Level Zone-Length Matrix (GLZLM) were extracted by using the software LIFEX. RESULTS A statistically significant difference was found in radiomic features including; a) Histogram: "skewness", b) Shape: "volume-ml, volume-voxel," c) GLCM: "entropy.log10, entropy.log2, energy", d) GLRLM: "GLNU, RLNU, HGRE," e) NGLDM: "busyness," f) GLZLM: "GLNU, HGZE, ZLNU, SZE" between two different molecular subtypes. The model combining Shape-volume (ml) and GLZLM-HGZE yielded 0.746 area under the curve (AUC), 0.744 sensitivity, 0.643 specificity and 0.694 accuracy. CONCLUSION Radiomic properties that may distinguish Luminal A and Luminal B molecular subtypes of IDC were identified. The radiomic features were thought to reflect the intratumoral heterogeneity in molecular subtypes. This study demonstrated that the characterization of Luminal A and Luminal B tumors could be made non-invasively by radiomics analysis.
Collapse
Affiliation(s)
- Oğuz Lafcı
- Department of Radiology, Ankara Training and Research Hospital, University of Health Sciences, Hacettepe Mh. Ulucanlar Cd. No:89 Altındağ, Ankara, Turkey.
| | - Pınar Celepli
- Department of Pathology, Ankara Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Pelin Seher Öztekin
- Department of Radiology, Ankara Training and Research Hospital, University of Health Sciences, Hacettepe Mh. Ulucanlar Cd. No:89 Altındağ, Ankara, Turkey
| | - Pınar Nercis Koşar
- Department of Radiology, Ankara Training and Research Hospital, University of Health Sciences, Hacettepe Mh. Ulucanlar Cd. No:89 Altındağ, Ankara, Turkey
| |
Collapse
|
31
|
Zhang L, Hao J, Guo J, Zhao X, Yin X. Predicting of Ki-67 Expression Level Using Diffusion-Weighted and Synthetic Magnetic Resonance Imaging in Invasive Ductal Breast Cancer. Breast J 2023; 2023:6746326. [PMID: 37063453 PMCID: PMC10098409 DOI: 10.1155/2023/6746326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/26/2023] [Accepted: 03/27/2023] [Indexed: 04/18/2023]
Abstract
Objectives To investigate the association between quantitative parameters generated using synthetic magnetic resonance imaging (SyMRI) and diffusion-weighted imaging (DWI) and Ki-67 expression level in patients with invasive ductal breast cancer (IDC). Method We retrospectively reviewed the records of patients with IDC who underwent SyMRI and DWI before treatment. Precontrast and postcontrast relaxation times (T1, longitudinal; T2, transverse), proton density (PD) parameters, and apparent diffusion coefficient (ADC) values were measured in breast lesions. Univariate and multivariate regression analyses were performed to screen for statistically significant variables to differentiate the high (≥30%) and low (<30%) Ki-67 expression groups. Their performance was evaluated by receiver operating characteristic (ROC) curve analysis. Results We analyzed 97 patients. Multivariate regression analysis revealed that the high Ki-67 expression group (n = 57) had significantly higher parameters generated using SyMRI (pre-T1, p=0.001) and lower ADC values (p=0.036) compared with the low Ki-67 expression group (n = 40). Pre-T1 showed the best diagnostic performance for predicting the Ki-67 expression level in patients with invasive ductal breast cancer (areas under the ROC curve (AUC), 0.711; 95% confidence interval (CI), 0.609-0.813). Conclusions Pre-T1 could be used to predict the pretreatment Ki-67 expression level in invasive ductal breast cancer.
Collapse
Affiliation(s)
- Liying Zhang
- Third Affiliated Hospital of Zhengzhou University, Department of Radiology, Zhengzhou, China
| | - Jisen Hao
- Third Affiliated Hospital of Zhengzhou University, Department of Radiology, Zhengzhou, China
| | - Jia Guo
- Third Affiliated Hospital of Zhengzhou University, Department of Radiology, Zhengzhou, China
| | - Xin Zhao
- Third Affiliated Hospital of Zhengzhou University, Department of Radiology, Zhengzhou, China
| | - Xing Yin
- Third Affiliated Hospital of Zhengzhou University, Department of Radiology, Zhengzhou, China
| |
Collapse
|
32
|
Jeon SH, Kim SW, Na K, Seo M, Sohn YM, Lim YJ. Radiomic models based on magnetic resonance imaging predict the spatial distribution of CD8 + tumor-infiltrating lymphocytes in breast cancer. Front Immunol 2022; 13:1080048. [PMID: 36601118 PMCID: PMC9806253 DOI: 10.3389/fimmu.2022.1080048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Infiltration of CD8+ T cells and their spatial contexture, represented by immunophenotype, predict the prognosis and therapeutic response in breast cancer. However, a non-surgical method using radiomics to evaluate breast cancer immunophenotype has not been explored. Here, we assessed the CD8+ T cell-based immunophenotype in patients with breast cancer undergoing upfront surgery (n = 182). We extracted radiomic features from the four phases of dynamic contrast-enhanced magnetic resonance imaging, and randomly divided the patients into training (n = 137) and validation (n = 45) cohorts. For predicting the immunophenotypes, radiomic models (RMs) that combined the four phases demonstrated superior performance to those derived from a single phase. For discriminating the inflamed tumor from the non-inflamed tumor, the feature-based combination model from the whole tumor (RM-wholeFC) showed high performance in both training (area under the receiver operating characteristic curve [AUC] = 0.973) and validation cohorts (AUC = 0.985). Similarly, the feature-based combination model from the peripheral tumor (RM-periFC) discriminated between immune-desert and excluded tumors with high performance in both training (AUC = 0.993) and validation cohorts (AUC = 0.984). Both RM-wholeFC and RM-periFC demonstrated good to excellent performance for every molecular subtype. Furthermore, in patients who underwent neoadjuvant chemotherapy (n = 64), pre-treatment images showed that tumors exhibiting complete response to neoadjuvant chemotherapy had significantly higher scores from RM-wholeFC and lower scores from RM-periFC. Our RMs predicted the immunophenotype of breast cancer based on the spatial distribution of CD8+ T cells with high accuracy. This approach can be used to stratify patients non-invasively based on the status of the tumor-immune microenvironment.
Collapse
Affiliation(s)
- Seung Hyuck Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - So-Woon Kim
- Department of Pathology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Kiyong Na
- Department of Pathology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Mirinae Seo
- Department of Radiology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Yu-Mee Sohn
- Department of Radiology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Yu Jin Lim
- Department of Radiation Oncology, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea,*Correspondence: Yu Jin Lim,
| |
Collapse
|
33
|
Fan Y, Pan X, Yang F, Liu S, Wang Z, Sun J, Chen J. Preoperative Computed Tomography Radiomics Analysis for Predicting Receptors Status and Ki-67 Levels in Breast Cancer. Am J Clin Oncol 2022; 45:526-533. [PMID: 36413682 PMCID: PMC9698095 DOI: 10.1097/coc.0000000000000951] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND To assess the prediction performance of preoperative chest computed tomography (CT) based radiomics features for estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor-2 (HER2+), and Ki-67 status of breast cancer. MATERIALS AND METHODS This study enrolled 108 breast cancer patients who received preoperative chest CT examinations in our institution from July 2018 to January 2020. Radiomics features were separately extracted from nonenhanced, arterial, and portal-venous phases CT images. The least absolute shrinkage and selection operator logistic regression was used for feature selection. Then the radiomics signatures for each phase and a combined model of 3 phases were built. Finally, the receiver operating characteristic curves and calibration curves were used to confirm the performance of the radiomics signatures and combined model. In addition, the decision curves were performed to estimate the clinical usefulness of the combined model. RESULTS The 20 most predictive features were finally selected to build radiomics signatures for each phase. The combined model achieved the overall best performance than using either of the nonenhanced, arterial and portal-venous phases alone, achieving an area under the receiver operating characteristic curve of 0.870 for ER+ versus ER-, 0.797 for PR+ versus PR-, 0.881 for HER2+ versus HER2-, and 0.726 for Ki-67. The decision curve demonstrated that the CT-based radiomics features were clinically useful. CONCLUSION This study indicated preopreative chest CT radiomics analysis might be able to assess ER, PR, HER2+, and Ki-67 status of breast cancer. The findings need further to be verified in future larger studies.
Collapse
Affiliation(s)
- Yuan Fan
- General Surgery Department, Qujing City First People’s Hospital, Qujing Yunnan
| | | | | | - Siyun Liu
- GE Healthcare life science, Shanghai, People’s Republic of China
| | - Zhu Wang
- Laboratory of Molecular Diagnosis of Cancer, Cancer Center
| | | | - Jie Chen
- Department of Breast Surgery, West China Hospital of Sichuan University, Chengdu Sichuan
| |
Collapse
|
34
|
Multiparametric MRI Features of Breast Cancer Molecular Subtypes. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58121716. [PMID: 36556918 PMCID: PMC9785392 DOI: 10.3390/medicina58121716] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/01/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022]
Abstract
Background and Objectives: Breast cancer (BC) molecular subtypes have unique incidence, survival and response to therapy. There are five BC subtypes described by immunohistochemistry: luminal A, luminal B HER2 positive and HER2 negative, triple negative (TNBC) and HER2-enriched. Multiparametric breast MRI (magnetic resonance imaging) provides morphological and functional characteristics of breast tumours and is nowadays recommended in the preoperative setting. Aim: To evaluate the multiparametric MRI features (T2-WI, ADC values and DCE) of breast tumours along with breast density and background parenchymal enhancement (BPE) features among different BC molecular subtypes. Materials and Methods: This was a retrospective study which included 344 patients. All underwent multiparametric breast MRI (T2WI, ADC and DCE sequences) and features were extracted according to the latest BIRADS lexicon. The inter-reader agreement was assessed using the intraclass coefficient (ICC) between the ROI of ADC obtained from the two breast imagers (experienced and moderately experienced). Results: The study population was divided as follows: 89 (26%) with luminal A, 39 (11.5%) luminal B HER2 positive, 168 (48.5%) luminal B HER2 negative, 41 (12%) triple negative (TNBC) and 7 (2%) with HER2 enriched. Luminal A tumours were associated with special histology type, smallest tumour size and persistent kinetic curve (all p-values < 0.05). Luminal B HER2 negative tumours were associated with lowest ADC value (0.77 × 10−3 mm2/s2), which predicts the BC molecular subtype with an accuracy of 0.583. TNBC were associated with asymmetric and moderate/marked BPE, round/oval masses with circumscribed margins and rim enhancement (all p-values < 0.05). HER2 enriched BC were associated with the largest tumour size (mean 37.28 mm, p-value = 0.02). Conclusions: BC molecular subtypes can be associated with T2WI, ADC and DCE MRI features. ADC can help predict the luminal B HER2 negative cases.
Collapse
|
35
|
Yin H, Bai L, Jia H, Lin G. Noninvasive assessment of breast cancer molecular subtypes on multiparametric MRI using convolutional neural network with transfer learning. Thorac Cancer 2022; 13:3183-3191. [PMID: 36203226 PMCID: PMC9663668 DOI: 10.1111/1759-7714.14673] [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: 07/13/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND To evaluate the performances of multiparametric MRI-based convolutional neural networks (CNNs) for the preoperative assessment of breast cancer molecular subtypes. METHODS A total of 136 patients with 136 pathologically confirmed invasive breast cancers were randomly divided into training, validation, and testing sets in this retrospective study. The CNN models were established based on contrast-enhanced T1 -weighted imaging (T1 C), Apparent diffusion coefficient (ADC), and T2 -weighted imaging (T2 W) using the training and validation sets. The performances of CNN models were evaluated on the testing set. The area under the receiver operating characteristic curve (AUC), sensitivity, specificity, and accuracy were calculated to assess the performance. RESULTS For the separation of each subtype from other subtypes on the testing set, the T1 C-based models yielded AUCs from 0.762 to 0.920; the ADC-based models yielded AUCs from 0.686 to 0.851; and the T2 W-based models achieved AUCs from 0.639 to 0.697. CONCLUSION T1 C-based models performed better than ADC-based models and T2 W-based models in assessing the breast cancer molecular subtypes. The discriminating performances of our CNN models for triple negative and human epidermal growth factor receptor 2-enriched subtypes were better than that of luminal A and luminal B subtypes.
Collapse
Affiliation(s)
- Haolin Yin
- Department of RadiologyHuadong Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Lutian Bai
- Department of RadiologyHuadong Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Huihui Jia
- Department of RadiologyHuadong Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Guangwu Lin
- Department of RadiologyHuadong Hospital Affiliated to Fudan UniversityShanghaiChina
| |
Collapse
|
36
|
Kong QC, Tang WJ, Chen SY, Hu WK, Hu Y, Liang YS, Zhang QQ, Cheng ZX, Huang D, Yang J, Guo Y. Nomogram for the prediction of triple-negative breast cancer histological heterogeneity based on multiparameter MRI features: A preliminary study including metaplastic carcinoma and non- metaplastic carcinoma. Front Oncol 2022; 12:916988. [PMID: 36212484 PMCID: PMC9533710 DOI: 10.3389/fonc.2022.916988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Objectives Triple-negative breast cancer (TNBC) is a heterogeneous disease, and different histological subtypes of TNBC have different clinicopathological features and prognoses. Therefore, this study aimed to establish a nomogram model to predict the histological heterogeneity of TNBC: including Metaplastic Carcinoma (MC) and Non-Metaplastic Carcinoma (NMC). Methods We evaluated 117 patients who had pathologically confirmed TNBC between November 2016 and December 2020 and collected preoperative multiparameter MRI and clinicopathological data. The patients were randomly assigned to a training set and a validation set at a ratio of 3:1. Based on logistic regression analysis, we established a nomogram model to predict the histopathological subtype of TNBC. Nomogram performance was assessed with the area under the receiver operating characteristic curve (AUC), calibration curve and decision curve. According to the follow-up information, disease-free survival (DFS) survival curve was estimated using the Kaplan-Meier product-limit method. Results Of the 117 TNBC patients, 29 patients had TNBC-MC (age range, 29–65 years; median age, 48.0 years), and 88 had TNBC-NMC (age range, 28–88 years; median age, 44.5 years). Multivariate logistic regression analysis demonstrated that lesion type (p = 0.001) and internal enhancement pattern (p = 0.001) were significantly predictive of TNBC subtypes in the training set. The nomogram incorporating these variables showed excellent discrimination power with an AUC of 0.849 (95% CI: 0.750−0.949) in the training set and 0.819 (95% CI: 0.693−0.946) in the validation set. Up to the cutoff date for this analysis, a total of 66 patients were enrolled in the prognostic analysis. Six of 14 TNBC-MC patients experienced recurrence, while 7 of 52 TNBC-NMC patients experienced recurrence. The DFS of the two subtypes was significantly different (p=0.035). Conclusions In conclusion, we developed a nomogram consisting of lesion type and internal enhancement pattern, which showed good discrimination ability in predicting TNBC-MC and TNBC-NMC.
Collapse
Affiliation(s)
- Qing-cong Kong
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wen-jie Tang
- Department of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Si-yi Chen
- Department of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wen-ke Hu
- Department of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yue Hu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yun-shi Liang
- Department of Pathology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiong-qiong Zhang
- Department of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zi-xuan Cheng
- Department of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Di Huang
- Department of Breast Surgery, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Di Huang, ; Jing Yang, ; Yuan Guo,
| | - Jing Yang
- Department of Pathology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Di Huang, ; Jing Yang, ; Yuan Guo,
| | - Yuan Guo
- Department of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Di Huang, ; Jing Yang, ; Yuan Guo,
| |
Collapse
|
37
|
Sheng W, Xia S, Wang Y, Yan L, Ke S, Mellisa E, Gong F, Zheng Y, Tang T. Invasive ductal breast cancer molecular subtype prediction by MRI radiomic and clinical features based on machine learning. Front Oncol 2022; 12:964605. [PMID: 36172153 PMCID: PMC9510620 DOI: 10.3389/fonc.2022.964605] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundMost studies of molecular subtype prediction in breast cancer were mainly based on two-dimensional MRI images, the predictive value of three-dimensional volumetric features from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) for predicting breast cancer molecular subtypes has not been thoroughly investigated. This study aimed to look into the role of features derived from DCE-MRI and how they could be combined with clinical data to predict invasive ductal breast cancer molecular subtypes.MethodsFrom January 2019 to December 2021, 190 Chinese women with invasive ductal breast cancer were studied (32 triple-negative, 59 HER2-enriched, and 99 luminal lesions) in this institutional review board-approved retrospective cohort study. The image processing software extracted 1130 quantitative radiomic features from the segmented lesion area, including shape-based, first-order statistical, texture, and wavelet features. Three binary classifications of the subtypes were performed: triple-negative vs. non-triple-negative, HER2-overexpressed vs. non-HER2-overexpressed, and luminal (A + B) vs. non-luminal. For the classification, five machine learning methods (random forest, logistic regression, support vector machine, naïve Bayes, and eXtreme Gradient Boosting) were employed. The classifiers were chosen using the least absolute shrinkage and selection operator method. The area evaluated classification performance under the receiver operating characteristic curve, sensitivity, specificity, accuracy, F1-Score, false positive rate, precision, and geometric mean.ResultsEXtreme Gradient Boosting model showed the best performance in luminal and non-luminal groups, with AUC, sensitivity, specificity, accuracy, F1-Score, false positive rate, precision, and geometric mean of 0.8282, 0.7524, 0.6542, 0.6964, 0.6086, 0.3458, 0.8524 and 0.7016, respectively. Meanwhile, the random forest model showed the best performance in HER2-overexpressed and non-HER2-overexpressed groups, with AUC, sensitivity, specificity, accuracy, F1-Score, false positive rate, precision, and geometric mean of 0.8054, 0.2941, 0.9744, 0.7679, 0.4348, 0.0256, 0.8333 and 0.5353, respectively. Furthermore, eXtreme Gradient Boosting model showed the best performance in the triple-negative and non-triple-negative groups, with AUC, sensitivity, specificity, accuracy, F1-Score, false positive rate, precision, and geometric mean of 0.9031, 0.9362, 0.4444, 0.8571, 0.9167, 0.5556, 0.8980 and 0.6450.ConclusionClinical data and three-dimension imaging features from DCE-MRI were identified as potential biomarkers for distinguishing between three molecular subtypes of invasive ductal carcinomas breast cancer. In the future, more extensive studies will be required to evaluate the findings.
Collapse
Affiliation(s)
- Weiyong Sheng
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Shouli Xia
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yaru Wang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lizhao Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songqing Ke
- Department of Science and Technology Research Management, Wuhan Blood Center, Wuhan, China
| | - Evelyn Mellisa
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Gong
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun Zheng
- Department of Radiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Yun Zheng, ; Tiansheng Tang,
| | - Tiansheng Tang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
- *Correspondence: Yun Zheng, ; Tiansheng Tang,
| |
Collapse
|
38
|
Wu J, Mayer AT, Li R. Integrated imaging and molecular analysis to decipher tumor microenvironment in the era of immunotherapy. Semin Cancer Biol 2022; 84:310-328. [PMID: 33290844 PMCID: PMC8319834 DOI: 10.1016/j.semcancer.2020.12.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/29/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023]
Abstract
Radiological imaging is an integral component of cancer care, including diagnosis, staging, and treatment response monitoring. It contains rich information about tumor phenotypes that are governed not only by cancer cellintrinsic biological processes but also by the tumor microenvironment, such as the composition and function of tumor-infiltrating immune cells. By analyzing the radiological scans using a quantitative radiomics approach, robust relations between specific imaging and molecular phenotypes can be established. Indeed, a number of studies have demonstrated the feasibility of radiogenomics for predicting intrinsic molecular subtypes and gene expression signatures in breast cancer based on MRI. In parallel, promising results have been shown for inferring the amount of tumor-infiltrating lymphocytes, a key factor for the efficacy of cancer immunotherapy, from standard-of-care radiological images. Compared with the biopsy-based approach, radiogenomics offers a unique avenue to profile the molecular makeup of the tumor and immune microenvironment as well as its evolution in a noninvasive and holistic manner through longitudinal imaging scans. Here, we provide a systematic review of the state of the art radiogenomics studies in the era of immunotherapy and discuss emerging paradigms and opportunities in AI and deep learning approaches. These technical advances are expected to transform the radiogenomics field, leading to the discovery of reliable imaging biomarkers. This will pave the way for their clinical translation to guide precision cancer therapy.
Collapse
Affiliation(s)
- Jia Wu
- Department of Imaging Physics, MD Anderson Cancer Center, Texas, 77030, USA; Department of Thoracic/Head & Neck Medical Oncology, MD Anderson Cancer Center, Texas, 77030, USA.
| | - Aaron T Mayer
- Department of Bioengineering, Stanford University, Stanford, California, 94305, USA; Department of Radiology, Stanford University, Stanford, California, 94305, USA; Molecular Imaging Program at Stanford, Stanford University, Stanford, California, 94305, USA; BioX Program at Stanford, Stanford University, Stanford, California, 94305, USA
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University, Stanford, California, 94305, USA
| |
Collapse
|
39
|
A Simultaneous Multiparametric 18F-FDG PET/MRI Radiomics Model for the Diagnosis of Triple Negative Breast Cancer. Cancers (Basel) 2022; 14:cancers14163944. [PMID: 36010936 PMCID: PMC9406327 DOI: 10.3390/cancers14163944] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary In this study, we aimed to build a machine-learning predictive model for the identification of triple negative breast cancer, the most aggressive subtype, using quantitative parameters and radiomics features extracted from tumor lesions on hybrid PET/MRI. The good performance of the model supports the hypothesis that hybrid PET/MRI can provide quantitative data able to non-invasively detect tumor biological characteristics using artificial intelligence software and further encourages the conduction of additional studies for this purpose. Abstract Purpose: To investigate whether a machine learning (ML)-based radiomics model applied to 18F-FDG PET/MRI is effective in molecular subtyping of breast cancer (BC) and specifically in discriminating triple negative (TN) from other molecular subtypes of BC. Methods: Eighty-six patients with 98 BC lesions (Luminal A = 10, Luminal B = 51, HER2+ = 12, TN = 25) were included and underwent simultaneous 18F-FDG PET/MRI of the breast. A 3D segmentation of BC lesion was performed on T2w, DCE, DWI and PET images. Quantitative diffusion and metabolic parameters were calculated and radiomics features extracted. Data were selected using the LASSO regression and used by a fine gaussian support vector machine (SVM) classifier with a 5-fold cross validation for identification of TNBC lesions. Results: Eight radiomics models were built based on different combinations of quantitative parameters and/or radiomic features. The best performance (AUROC 0.887, accuracy 82.8%, sensitivity 79.7%, specificity 86%, PPV 85.3%, NPV 80.8%) was found for the model combining first order, neighborhood gray level dependence matrix and size zone matrix-based radiomics features extracted from ADC and PET images. Conclusion: A ML-based radiomics model applied to 18F-FDG PET/MRI is able to non-invasively discriminate TNBC lesions from other BC molecular subtypes with high accuracy. In a future perspective, a “virtual biopsy” might be performed with radiomics signatures.
Collapse
|
40
|
Yue W, Zhang H, Zhou J, Li G, Tang Z, Sun Z, Cai J, Tian N, Gao S, Dong J, Liu Y, Bai X, Sheng F. Deep learning-based automatic segmentation for size and volumetric measurement of breast cancer on magnetic resonance imaging. Front Oncol 2022; 12:984626. [PMID: 36033453 PMCID: PMC9404224 DOI: 10.3389/fonc.2022.984626] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 07/19/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose In clinical work, accurately measuring the volume and the size of breast cancer is significant to develop a treatment plan. However, it is time-consuming, and inter- and intra-observer variations among radiologists exist. The purpose of this study was to assess the performance of a Res-UNet convolutional neural network based on automatic segmentation for size and volumetric measurement of mass enhancement breast cancer on magnetic resonance imaging (MRI). Materials and methods A total of 1,000 female breast cancer patients who underwent preoperative 1.5-T dynamic contrast-enhanced MRI prior to treatment were selected from January 2015 to October 2021 and randomly divided into a training cohort (n = 800) and a testing cohort (n = 200). Compared with the masks named ground truth delineated manually by radiologists, the model performance on segmentation was evaluated with dice similarity coefficient (DSC) and intraclass correlation coefficient (ICC). The performance of tumor (T) stage classification was evaluated with accuracy, sensitivity, and specificity. Results In the test cohort, the DSC of automatic segmentation reached 0.89. Excellent concordance (ICC > 0.95) of the maximal and minimal diameter and good concordance (ICC > 0.80) of volumetric measurement were shown between the model and the radiologists. The trained model took approximately 10–15 s to provide automatic segmentation and classified the T stage with an overall accuracy of 0.93, sensitivity of 0.94, 0.94, and 0.75, and specificity of 0.95, 0.92, and 0.99, respectively, in T1, T2, and T3. Conclusions Our model demonstrated good performance and reliability for automatic segmentation for size and volumetric measurement of breast cancer, which can be time-saving and effective in clinical decision-making.
Collapse
Affiliation(s)
- Wenyi Yue
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Chinese PLA General Medical School, Beijing, China
| | - Hongtao Zhang
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Juan Zhou
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guang Li
- Keya Medical Technology Co., Ltd., Beijing, China
| | - Zhe Tang
- Keya Medical Technology Co., Ltd., Beijing, China
| | - Zeyu Sun
- Keya Medical Technology Co., Ltd., Beijing, China
| | - Jianming Cai
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ning Tian
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shen Gao
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jinghui Dong
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuan Liu
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xu Bai
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fugeng Sheng
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Fugeng Sheng,
| |
Collapse
|
41
|
Radiogenomic analysis reveals tumor heterogeneity of triple-negative breast cancer. Cell Rep Med 2022; 3:100694. [PMID: 35858585 PMCID: PMC9381418 DOI: 10.1016/j.xcrm.2022.100694] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/04/2022]
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with an adverse prognosis and significant tumor heterogeneity. Here, we extract quantitative radiomic features from contrast-enhanced magnetic resonance images to construct a breast cancer radiomic dataset (n = 860) and a TNBC radiogenomic dataset (n = 202). We develop and validate radiomic signatures that can fairly differentiate TNBC from other breast cancer subtypes and distinguish molecular subtypes within TNBC. A radiomic feature that captures peritumoral heterogeneity is determined to be a prognostic factor for recurrence-free survival (p = 0.01) and overall survival (p = 0.004) in TNBC. Combined with the established matching TNBC transcriptomic and metabolomic data, we demonstrate that peritumoral heterogeneity is associated with immune suppression and upregulated fatty acid synthesis in tumor samples. Collectively, this multi-omic dataset serves as a useful public resource to promote precise subtyping of TNBC and helps to understand the biological significance of radiomics. A radiomic signature identifies TNBC from other subtypes of breast cancer Radiomic features are predictive of TNBC molecular subtypes A radiomic feature reflecting peritumoral heterogeneity indicates TNBC prognosis Peritumoral heterogeneity correlates with metabolic and immune abnormalities in TNBC
Collapse
|
42
|
Xu A, Chu X, Zhang S, Zheng J, Shi D, Lv S, Li F, Weng X. Prediction Breast Molecular Typing of Invasive Ductal Carcinoma Based on Dynamic Contrast Enhancement Magnetic Resonance Imaging Radiomics Characteristics: A Feasibility Study. Front Oncol 2022; 12:799232. [PMID: 35664741 PMCID: PMC9160981 DOI: 10.3389/fonc.2022.799232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To investigate the feasibility of radiomics in predicting molecular subtype of breast invasive ductal carcinoma (IDC) based on dynamic contrast enhancement magnetic resonance imaging (DCE-MRI). Methods A total of 303 cases with pathologically confirmed IDC from January 2018 to March 2021 were enrolled in this study, including 223 cases from Fudan University Shanghai Cancer Center (training/test set) and 80 cases from Shaoxing Central Hospital (validation set). All the cases were classified as HR+/Luminal, HER2-enriched, and TNBC according to immunohistochemistry. DCE-MRI original images were treated by semi-automated segmentation to initially extract original and wavelet-transformed radiomic features. The extended logistic regression with least absolute shrinkage and selection operator (LASSO) penalty was applied to identify the optimal radiomic features, which were then used to establish predictive models combined with significant clinical risk factors. Receiver operating characteristic curve (ROC), calibration curve, and decision curve analysis were adopted to evaluate the effectiveness and clinical benefit of the models established. Results Of the 223 cases from Fudan University Shanghai Cancer Center, HR+/Luminal cancers were diagnosed in 116 cases (52.02%), HER2-enriched in 71 cases (31.84%), and TNBC in 36 cases (16.14%). Based on the training set, 788 radiomic features were extracted in total and 8 optimal features were further identified, including 2 first-order features, 1 gray-level run length matrix (GLRLM), 4 gray-level co-occurrence matrices (GLCM), and 1 3D shape feature. Three multi-class classification models were constructed by extended logistic regression: clinical model (age, menopause, tumor location, Ki-67, histological grade, and lymph node metastasis), radiomic model, and combined model. The macro-average areas under the ROC curve (macro-AUC) for the three models were 0.71, 0.81, and 0.84 in the training set, 0.73, 0.81, and 0.84 in the test set, and 0.76, 0.82, and 0.83 in the validation set, respectively. Conclusion The DCE-MRI-based radiomic features are significant biomarkers for distinguishing molecular subtypes of breast cancer noninvasively. Notably, the classification performance could be improved with the fusion analysis of multi-modal features.
Collapse
Affiliation(s)
- Aqiao Xu
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| | - Xiufeng Chu
- Department of Surgical, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| | - Shengjian Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jing Zheng
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| | - Dabao Shi
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| | - Shasha Lv
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| | - Feng Li
- Department of Research Collaboration, Research & Development Center (R&D), Beijing Deepwise & League of Doctor of Philosophy (PHD) Technology Co., Ltd, Beijing, China
| | - Xiaobo Weng
- Department of Radiology, The Central Hospital Affiliated to Shaoxing University (Shaoxing Central Hospital), Shaoxing, China
| |
Collapse
|
43
|
Sha Y, Chen J. MRI-based radiomics for the diagnosis of triple-negative breast cancer: a meta-analysis. Clin Radiol 2022; 77:655-663. [DOI: 10.1016/j.crad.2022.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 04/21/2022] [Indexed: 11/03/2022]
|
44
|
Wang G, Shi D, Guo Q, Zhang H, Wang S, Ren K. Radiomics Based on Digital Mammography Helps to Identify Mammographic Masses Suspicious for Cancer. Front Oncol 2022; 12:843436. [PMID: 35433437 PMCID: PMC9012139 DOI: 10.3389/fonc.2022.843436] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 03/04/2022] [Indexed: 12/11/2022] Open
Abstract
Objectives This study aims to build radiomics model of Breast Imaging Reporting and Data System (BI-RADS) category 4 and 5 mammographic masses extracted from digital mammography (DM) for mammographic masses characterization by using a sensitivity threshold similar to that of biopsy. Materials and Methods This retrospective study included 288 female patients (age, 52.41 ± 10.31) who had BI-RADS category 4 or 5 mammographic masses with an indication for biopsy. The patients were divided into two temporal set (training set, 82 malignancies and 110 benign lesions; independent test set, 48 malignancies and 48 benign lesions). A total of 188 radiomics features were extracted from mammographic masses on the combination of craniocaudal (CC) position images and mediolateral oblique (MLO) position images. For the training set, Pearson’s correlation and the least absolute shrinkage and selection operator (LASSO) were used to select non-redundant radiomics features and useful radiomics features, respectively, and support vector machine (SVM) was applied to construct a radiomics model. The receiver operating characteristic curve (ROC) analysis was used to evaluate the classification performance of the radiomics model and to determine a threshold value with a sensitivity higher than 98% to predict the mammographic masses malignancy. For independent test set, identical threshold value was used to validate the classification performance of the radiomics model. The stability of the radiomics model was evaluated by using a fivefold cross-validation method, and two breast radiologists assessed the diagnostic agreement of the radiomics model. Results In the training set, the radiomics model obtained an area under the receiver operating characteristic curve (AUC) of 0.934 [95% confidence intervals (95% CI), 0.898–0.971], a sensitivity of 98.8% (81/82), a threshold of 0.22, and a specificity of 60% (66/110). In the test set, the radiomics model obtained an AUC of 0.901 (95% CI, 0.835–0.961), a sensitivity of 95.8% (46/48), and a specificity of 66.7% (32/48). The radiomics model had relatively stable sensitivities in fivefold cross-validation (training set, 97.39% ± 3.9%; test set, 98.7% ± 4%). Conclusion The radiomics method based on DM may help reduce the temporarily unnecessary invasive biopsies for benign mammographic masses over-classified in BI-RADS category 4 and 5 while providing similar diagnostic performance for malignant mammographic masses as biopsies.
Collapse
Affiliation(s)
| | - Dafa Shi
- Xiang’an Hospital, Xiamen University, Xiamen, China
| | - Qiu Guo
- Xiang’an Hospital, Xiamen University, Xiamen, China
| | - Haoran Zhang
- Xiang’an Hospital, Xiamen University, Xiamen, China
| | - Siyuan Wang
- Xiang’an Hospital, Xiamen University, Xiamen, China
| | - Ke Ren
- Xiang’an Hospital, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, China
- *Correspondence: Ke Ren,
| |
Collapse
|
45
|
Multiparametric 18F-FDG PET/MRI-Based Radiomics for Prediction of Pathological Complete Response to Neoadjuvant Chemotherapy in Breast Cancer. Cancers (Basel) 2022; 14:cancers14071727. [PMID: 35406499 PMCID: PMC8996836 DOI: 10.3390/cancers14071727] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The aim of this study was to assess whether multiparametric 18F-FDG PET/MRI-based radiomics analysis is able to predict pathological complete response in breast cancer patients and hence potentially enhance pretherapeutic patient stratification. METHODS A total of 73 female patients (mean age 49 years; range 27-77 years) with newly diagnosed, therapy-naive breast cancer underwent simultaneous 18F-FDG PET/MRI and were included in this retrospective study. All PET/MRI datasets were imported to dedicated software (ITK-SNAP v. 3.6.0) for lesion annotation using a semi-automated method. Pretreatment biopsy specimens were used to determine tumor histology, tumor and nuclear grades, and immunohistochemical status. Histopathological results from surgical tumor specimens were used as the reference standard to distinguish between complete pathological response (pCR) and noncomplete pathological response. An elastic net was employed to select the most important radiomic features prior to model development. Sensitivity, specificity, positive predictive value, negative predictive value, and accuracy were calculated for each model. RESULTS The best results in terms of AUCs and NPV for predicting complete pathological response in the entire cohort were obtained by the combination of all MR sequences and PET (0.8 and 79.5%, respectively), and no significant differences from the other models were observed. In further subgroup analyses, combining all MR and PET data, the best AUC (0.94) for predicting complete pathologic response was obtained in the HR+/HER2- group. No difference between results with/without the inclusion of PET characteristics was observed in the TN/HER2+ group, each leading to an AUC of 0.92 for all MR and all MR + PET datasets. CONCLUSION 18F-FDG PET/MRI enables comprehensive high-quality radiomics analysis for the prediction of pCR in breast cancer patients, especially in those with HR+/HER2- receptor status.
Collapse
|
46
|
Lepola A, Arponen O, Okuma H, Holli-Helenius K, Junkkari H, Könönen M, Auvinen P, Sudah M, Sutela A, Vanninen R. Association between breast cancer's prognostic factors and 3D textural features of non-contrast-enhanced T1 weighted breast MRI. Br J Radiol 2022; 95:20210702. [PMID: 34826254 PMCID: PMC8822552 DOI: 10.1259/bjr.20210702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVES The aim of this exploratory study was to evaluate whether three-dimensional texture analysis (3D-TA) features of non-contrast-enhanced T1 weighted MRI associate with traditional prognostic factors and disease-free survival (DFS) of breast cancer. METHODS 3D-T1 weighted images from 78 patients with 81 malignant histopathologically verified breast lesions were retrospectively analysed using standard-size volumes of interest. Grey-level co-occurrence matrix (GLCM)-based features were selected for statistical analysis. In statistics the Mann-Whitney U and the Kruskal-Wallis tests, the Cox proportional hazards model and the Kaplan-Meier method were used. RESULTS Tumours with higher histological grade were significantly associated with higher contrast (1 voxel: p = 0.033, 2 voxels: p = 0.036). All the entropy parameters showed significant correlation with tumour grade (p = 0.015-0.050) but there were no statistically significant associations between other TA parameters and tumour grade. The Nottingham Prognostic Index (NPI) was correlated with contrast and sum entropy parameters. A higher sum variance TA parameter was a significant predictor of shorter DFS. CONCLUSION Texture parameters, assessed by 3D-TA from non-enhanced T1 weighted images, indicate tumour heterogeneity but have limited independent prognostic value. However, they are associated with tumour grade, NPI, and DFS. These parameters could be used as an adjunct to contrast-enhanced TA parameters. ADVANCES IN KNOWLEDGE 3D-TA of non-contrast enhanced T1 weighted breast MRI associates with tumour grade, NPI, and DFS. The use of non-contrast 3D-TA parameters in adjunct with contrast-enhanced 3D-TA parameters warrants further research.
Collapse
Affiliation(s)
| | | | | | | | | | - Mervi Könönen
- Department of Clinical Radiology, Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | | | | | | | | |
Collapse
|
47
|
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: 1.0] [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
|
48
|
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: 2.0] [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
|
49
|
Maggio R, Messina F, D’Arrigo B, Maccagno G, Lardo P, Palmisano C, Poggi M, Monti S, Matarazzo I, Laghi A, Pugliese G, Stigliano A. Machine Learning-Based Texture Analysis in the Characterization of Cortisol Secreting vs. Non-Secreting Adrenocortical Incidentalomas in CT Scan. Front Endocrinol (Lausanne) 2022; 13:873189. [PMID: 35784576 PMCID: PMC9248203 DOI: 10.3389/fendo.2022.873189] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
New radioimaging techniques, exploiting the quantitative variables of imaging, permit to identify an hypothetical pathological tissue. We have applied this potential in a series of 72 adrenal incidentalomas (AIs) followed at our center, subdivided in functioning and non-functioning using laboratory findings. Each AI was studied in the preliminary non-contrast phase with a specific software (Mazda), surrounding a region of interest within each lesion. A total of 314 features were extrapolated. Mean and standard deviations of features were obtained and the difference in means between the two groups was statistically analyzed. Receiver Operating Characteristic (ROC) curves were used to identify an optimal cutoff for each variable and a prediction model was constructed via multivariate logistic regression with backward and stepwise selection. A 11-variable prediction model was constructed, and a ROC curve was used to differentiate patients with high probability of functioning AI. Using a threshold value of >-275.147, we obtained a sensitivity of 93.75% and a specificity of 100% in diagnosing functioning AI. On the basis of these results, computed tomography (CT) texture analysis appears a promising tool in the diagnostic definition of AIs.
Collapse
Affiliation(s)
- Roberta Maggio
- Endocrinology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Filippo Messina
- Department of Surgical and Medical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Benedetta D’Arrigo
- Department of Surgical and Medical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Giacomo Maccagno
- Department of Surgical and Medical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Pina Lardo
- Endocrinology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Claudia Palmisano
- Department of Surgical and Medical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Maurizio Poggi
- Endocrinology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Salvatore Monti
- Endocrinology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Iolanda Matarazzo
- Department of Surgical and Medical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Andrea Laghi
- Department of Surgical and Medical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Pugliese
- Endocrinology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Antonio Stigliano
- Endocrinology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
- *Correspondence: Antonio Stigliano,
| |
Collapse
|
50
|
Li X, Guo H, Cong C, Liu H, Zhang C, Luo X, Zhong P, Shi H, Fang J, Wang Y. The Potential Value of Texture Analysis Based on Dynamic Contrast-Enhanced MR Images in the Grading of Breast Phyllode Tumors. Front Oncol 2021; 11:745242. [PMID: 34858821 PMCID: PMC8631520 DOI: 10.3389/fonc.2021.745242] [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: 07/21/2021] [Accepted: 10/18/2021] [Indexed: 12/03/2022] Open
Abstract
Purpose To explore the value of texture analysis (TA) based on dynamic contrast-enhanced MR (DCE-MR) images in the differential diagnosis of benign phyllode tumors (BPTs) and borderline/malignant phyllode tumors (BMPTs). Methods A total of 47 patients with histologically proven phyllode tumors (PTs) from November 2012 to March 2020, including 26 benign BPTs and 21 BMPTs, were enrolled in this retrospective study. The whole-tumor texture features based on DCE-MR images were calculated, and conventional imaging findings were evaluated according to the Breast Imaging Reporting and Data System (BI-RADS). The differences in the texture features and imaging findings between BPTs and BMPTs were compared; the variates with statistical significance were entered into logistic regression analysis. The receiver operating characteristic (ROC) curve was used to assess the diagnostic performance of models from image-based analysis, TA, and the combination of these two approaches. Results Regarding texture features, three features of the histogram, two features of the gray-level co-occurrence matrix (GLCM), and three features of the run-length matrix (RLM) showed significant differences between the two groups (all p < 0.05). Regarding imaging findings, however, only cystic wall morphology showed significant differences between the two groups (p = 0.014). The areas under the ROC curve (AUCs) of image-based analysis, TA, and the combination of these two approaches were 0.687 (95% CI, 0.518–0.825, p = 0.014), 0.886 (95% CI, 0.760–0.960, p < 0.0001), and 0.894 (95% CI, 0.754–0.970, p < 0.0001), respectively. Conclusion TA based on DCE-MR images has potential in differentiating BPTs and BMPTs.
Collapse
Affiliation(s)
- Xiaoguang Li
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Hong Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Chao Cong
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | | | - Chunlai Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiangguo Luo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Peng Zhong
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, China
| | - Hang Shi
- Department of Information, Daping Hospital, Army Medical University, Chongqing, China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Wang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|