51
|
Response Predictivity to Neoadjuvant Therapies in Breast Cancer: A Qualitative Analysis of Background Parenchymal Enhancement in DCE-MRI. J Pers Med 2021; 11:jpm11040256. [PMID: 33915842 PMCID: PMC8065517 DOI: 10.3390/jpm11040256] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Background: For assessing the predictability of oncology neoadjuvant therapy results, the background parenchymal enhancement (BPE) parameter in breast magnetic resonance imaging (MRI) has acquired increased interest. This work aims to qualitatively evaluate the BPE parameter as a potential predictive marker for neoadjuvant therapy. Method: Three radiologists examined, in triple-blind modality, the MRIs of 80 patients performed before the start of chemotherapy, after three months from the start of treatment, and after surgery. They identified the portion of fibroglandular tissue (FGT) and BPE of the contralateral breast to the tumor in the basal control pre-treatment (baseline). Results: We observed a reduction of BPE classes in serial MRI checks performed during neoadjuvant therapy, as compared to baseline pre-treatment conditions, in 61.3% of patients in the intermediate step, and in 86.7% of patients in the final step. BPE reduction was significantly associated with sequential anthracyclines/taxane administration in the first cycle of neoadjuvant therapy compared to anti-HER2 containing therapies. The therapy response was also significantly related to tumor size. There were no associations with menopausal status, fibroglandular tissue (FGT) amount, age, BPE baseline, BPE in intermediate, and in the final MRI step. Conclusions: The measured variability of this parameter during therapy could predict therapy effectiveness in early stages, improving decision-making in the perspective of personalized medicine. Our preliminary results suggest that BPE may represent a predictive factor in response to neoadjuvant therapy in breast cancer, warranting future investigations in conjunction with radiomics.
Collapse
|
52
|
Velichko YS, Mozafarykhamseh A, Trabzonlu TA, Zhang Z, Rademaker AW, Yaghmai V. Association Between the Size and 3D CT-Based Radiomic Features of Breast Cancer Hepatic Metastasis. Acad Radiol 2021; 28:e93-e100. [PMID: 32303447 PMCID: PMC10029938 DOI: 10.1016/j.acra.2020.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 01/05/2023]
Abstract
PURPOSE To evaluate the effect of the anatomic size on 3D radiomic imaging features of the breast cancer hepatic metastases. MATERIALS AND METHODS CT scans of 81 liver metastases from 54 patients with breast cancer were evaluated. Ten most common 3D radiomic features from the histogram and gray level co-occurrence matrix (GLCM) categories were calculated for the hepatic metastases (HM) and compared to normal liver (NL). The effect of size was evaluated by using linear mixed-effects regression models. The effect of size on different radiomic features was analyzed for both liver lesions and background liver. RESULTS Three-dimensional radiomic features from GLCM demonstrate an important size dependence. The texture-feature size dependence was found to be different among feature categories and between the HM and NL, thus demonstrating a discriminatory power for the tissue type. Significant difference in the slope was found for GLCM homogeneity (NL slope = 0.004, slope difference 95% confidence interval [CI] 0.06-0.1, p <0.001), contrast (NL slope = 45, slope difference 95% CI 205-305, p <0.001), correlation (NL slope = 0.04, slope difference 95% CI 0.11-0.21, p <0.001), and dissimilarity (NL slope = 0.7, slope difference 95% CI 3.6-5.4, p <0.001). The GLCM energy (NL slope = 0.002, slope difference 95% CI -0.0005 to -0.0003, p <0.007), and entropy (NL slope = 1.49, slope difference 95% CI 0.07-0.52, p <0.009) exhibited size-dependence for both NL and HM, although demonstrating a difference in the slope between themselves. CONCLUSION Radiomic features of breast cancer hepatic metastasis exhibited significant correlation with tumor size. This finding demonstrates the complex behavior of imaging features and the need to include feature-specific properties into radiomic models.
Collapse
Affiliation(s)
- Yuri S Velichko
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Quantitative Imaging Core Lab, Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | | | - Tugce Agirlar Trabzonlu
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Zhuoli Zhang
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Quantitative Imaging Core Lab, Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Alfred W Rademaker
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Vahid Yaghmai
- Quantitative Imaging Core Lab, Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
53
|
Radiomics MRI for lymph node status prediction in breast cancer patients: the state of art. J Cancer Res Clin Oncol 2021; 147:1587-1597. [PMID: 33758997 DOI: 10.1007/s00432-021-03606-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To create a review of the existing literature on the radiomic approach in predicting the lymph node status of the axilla in breast cancer (BC). MATERIALS AND METHODS Two reviewers conducted the literature search on MEDLINE databases independently. Ten articles on the prediction of sentinel lymph node metastasis in breast cancer with a radiomic approach were selected. The study characteristics and results were reported. The quality of the methodology was evaluated according to the Radiomics Quality Score (RQS). RESULTS All studies were retrospective in design and published between 2017 and 2020. The majority of studies used DCE-MRI sequences and two investigated only pre-contrast images. The sample size was lower than 200 patients for 7 studies. The pre-processing used software, feature extraction and selection methods and classifier development are heterogeneous and a standardization of results is not yet possible. The average RQS score was 11.1 (maximum possible value = 36). The criteria with the lowest scores were the type of study, validation, comparison with a gold standard, potential clinical utility, cost-effective analysis and open science data. CONCLUSION The field of radiomics is a diagnostic approach of relative recent development. The results in predicting axillary lymph node status are encouraging, but there are still weaknesses in the quality of studies that may limit the reproducibility of the results.
Collapse
|
54
|
Fan M, Chen H, You C, Liu L, Gu Y, Peng W, Gao X, Li L. Radiomics of Tumor Heterogeneity in Longitudinal Dynamic Contrast-Enhanced Magnetic Resonance Imaging for Predicting Response to Neoadjuvant Chemotherapy in Breast Cancer. Front Mol Biosci 2021; 8:622219. [PMID: 33869279 PMCID: PMC8044916 DOI: 10.3389/fmolb.2021.622219] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/15/2021] [Indexed: 01/23/2023] Open
Abstract
Breast tumor morphological and vascular characteristics can be changed during neoadjuvant chemotherapy (NACT). The early changes in tumor heterogeneity can be quantitatively modeled by longitudinal dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), which is useful in predicting responses to NACT in breast cancer. In this retrospective analysis, 114 female patients with unilateral unifocal primary breast cancer who received NACT were included in a development (n = 61) dataset and a testing dataset (n = 53). DCE-MRI was performed for each patient before and after treatment (two cycles of NACT) to generate baseline and early follow-up images, respectively. Feature-level changes (delta) of the entire tumor were evaluated by calculating the relative net feature change (deltaRAD) between baseline and follow-up images. The voxel-level change inside the tumor was evaluated, which yielded a Jacobian map by registering the follow-up image to the baseline image. Clinical information and the radiomic features were fused to enhance the predictive performance. The area under the curve (AUC) values were assessed to evaluate the prediction performance. Predictive models using radiomics based on pre- and post-treatment images, Jacobian maps and deltaRAD showed AUC values of 0.568, 0.767, 0.630 and 0.726, respectively. When features from these images were fused, the predictive model generated an AUC value of 0.771. After adding the molecular subtype information in the fused model, the performance was increased to an AUC of 0.809 (sensitivity of 0.826 and specificity of 0.800), which is significantly higher than that of the baseline imaging- and Jacobian map-based predictive models (p = 0.028 and 0.019, respectively). The level of tumor heterogeneity reduction (evaluated by texture feature) is higher in the NACT responders than in the nonresponders. The results suggested that changes in DCE-MRI features that reflect a reduction in tumor heterogeneity following NACT could provide early prediction of breast tumor response. The prediction was improved when the molecular subtype information was combined into the model.
Collapse
Affiliation(s)
- Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China
| | - Hang Chen
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Li Liu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Weijun Peng
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xin Gao
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Lihua Li
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China
| |
Collapse
|
55
|
Tan Y, Mai H, Huang Z, Zhang L, Li C, Wu S, Huang H, Tang W, Liu Y, Jiang K. Additive value of texture analysis based on breast MRI for distinguishing between benign and malignant non-mass enhancement in premenopausal women. BMC Med Imaging 2021; 21:48. [PMID: 33706695 PMCID: PMC7953679 DOI: 10.1186/s12880-021-00571-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/21/2021] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Non-mass enhancement (NME) is a diagnostic dilemma and highly reliant on the experience of the radiologists. Texture analysis (TA) could serve as an objective method to quantify lesion characteristics. However, it remains unclear what role TA plays in a predictive model based on routine MRI characteristics. The purpose of this study was to explore the value of TA in distinguishing between benign and malignant NME in premenopausal women. METHODS Women in whom NME was histologically proven (n = 147) were enrolled (benign: 58; malignant: 89) was retrospective. Then, 102 and 45 patients were classified as the training and validation groups, respectively. Scanning sequences included Fat-suppressed T2-weighted and fat-suppressed contrast-enhanced T1-weighted which were acquired on a 1.5T MRI system. Clinical and routine MR characteristics (CRMC) were evaluated by two radiologists according to the Breast Imaging and Reporting and Data system (2013). Texture features were extracted from all post-contrast sequences in the training group. The combination model was built and then assessed in the validation group. Pearson's chi-square test and Mann-Whitney U test were used to compare categorical variables and continuous variables, respectively. Logistic regression analysis and receiver operating characteristic curve were employed to assess the diagnostic performance of CRMC, TA, and their combination model in NME diagnosis. RESULTS The combination model showed superior diagnostic performance in differentiating between benign and malignant NME compared to that of CRMC or TA alone (AUC, 0.887 vs 0.832 vs 0.74). Moreover, compared to CRMC, the model showed high specificity (72.5% vs 80%). The results obtained in the validation group confirmed the model was promising. CONCLUSIONS With the combined use of TA and CRMC could afford an improved diagnostic performance in differentiating between benign and malignant NME.
Collapse
Affiliation(s)
- Yu Tan
- Department of Radiology, Guangdong Women and Children Hospital, No.521, Xingnan Road, Panyu District, Guangzhou, 511400, China
| | - Hui Mai
- Department of Radiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiqing Huang
- Department of Radiology, Guangdong Women and Children Hospital, No.521, Xingnan Road, Panyu District, Guangzhou, 511400, China
| | - Li Zhang
- Department of Radiology, Guangdong Women and Children Hospital, No.521, Xingnan Road, Panyu District, Guangzhou, 511400, China
| | - Chengwei Li
- Department of Radiology, Guangdong Women and Children Hospital, No.521, Xingnan Road, Panyu District, Guangzhou, 511400, China
| | - Songxin Wu
- Department of Radiology, Guangdong Women and Children Hospital, No.521, Xingnan Road, Panyu District, Guangzhou, 511400, China
| | - Huang Huang
- Department of Radiology, Guangdong Women and Children Hospital, No.521, Xingnan Road, Panyu District, Guangzhou, 511400, China
| | - Wen Tang
- Department of Radiology, Guangdong Women and Children Hospital, No.521, Xingnan Road, Panyu District, Guangzhou, 511400, China
| | - Yongxi Liu
- Department of Radiology, Guangdong Women and Children Hospital, No.521, Xingnan Road, Panyu District, Guangzhou, 511400, China
| | - Kuiming Jiang
- Department of Radiology, Guangdong Women and Children Hospital, No.521, Xingnan Road, Panyu District, Guangzhou, 511400, China.
| |
Collapse
|
56
|
Huang X, Mai J, Huang Y, He L, Chen X, Wu X, Li Y, Yang X, Dong M, Huang J, Zhang F, Liang C, Liu Z. Radiomic Nomogram for Pretreatment Prediction of Pathologic Complete Response to Neoadjuvant Therapy in Breast Cancer: Predictive Value of Staging Contrast-enhanced CT. Clin Breast Cancer 2020; 21:e388-e401. [PMID: 33451965 DOI: 10.1016/j.clbc.2020.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 12/08/2020] [Accepted: 12/13/2020] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The purpose of this study was to predict pathologic complete response (pCR) to neoadjuvant therapy in breast cancer using radiomics based on pretreatment staging contrast-enhanced computed tomography (CECT). PATIENTS AND METHODS A total of 215 patients were retrospectively analyzed. Based on the intratumoral and peritumoral regions of CECT images, radiomic features were extracted and selected, respectively, to develop an intratumoral signature and a peritumoral signature with logistic regression in a training dataset (138 patients from November 2015 to October 2017). We also developed a clinical model with the molecular characterization of the tumor. A radiomic nomogram was further constructed by incorporating the intratumoral and peritumoral signatures with molecular characterization. The performance of the nomogram was validated in terms of discrimination, calibration, and clinical utility in an independent validation dataset (77 patients from November 2017 to December 2018). Stratified analysis was performed to develop a subtype-specific radiomic signature for each subgroup. RESULTS Compared with the clinical model (area under the curve [AUC], 0.756), the radiomic nomogram (AUC, 0.818) achieved better performance for pCR prediction in the validation dataset with continuous net reclassification improvement of 0.787 and good calibration. Decision curve analysis suggested the nomogram was clinically useful. Subtype-specific radiomic signatures showed improved AUCs (luminal subgroup, 0.936; human epidermal growth factor receptor 2-positive subgroup, 0.825; and triple negative subgroup, 0.858) for pCR prediction. CONCLUSION This study has revealed a predictive value of pretreatment staging-CECT and successfully developed and validated a radiomic nomogram for individualized prediction of pCR to neoadjuvant therapy in breast cancer, which could assist clinical decision-making and improve patient outcome.
Collapse
Affiliation(s)
- Xiaomei Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jinhai Mai
- School of Automation Science and Engineering, South China University of Technology, Guangzhou, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lan He
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaomei Wu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yexing Li
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaojun Yang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Mengyi Dong
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jia Huang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fang Zhang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Changhong Liang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Zaiyi Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
57
|
Saint Martin MJ, Orlhac F, Akl P, Khalid F, Nioche C, Buvat I, Malhaire C, Frouin F. A radiomics pipeline dedicated to Breast MRI: validation on a multi-scanner phantom study. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2020; 34:355-366. [PMID: 33180226 DOI: 10.1007/s10334-020-00892-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/27/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Quantitative analysis in MRI is challenging due to variabilities in intensity distributions across patients, acquisitions and scanners and suffers from bias field inhomogeneity. Radiomic studies are impacted by these effects that affect radiomic feature values. This paper describes a dedicated pipeline to increase reproducibility in breast MRI radiomic studies. MATERIALS AND METHODS T1, T2, and T1-DCE MR images of two breast phantoms were acquired using two scanners and three dual breast coils. Images were retrospectively corrected for bias field inhomogeneity and further normalised using Z score or histogram matching. Extracted radiomic features were harmonised between coils by the ComBat method. The whole pipeline was assessed qualitatively and quantitatively using statistical comparisons on two series of radiomic feature values computed in the gel mimicking the normal breast tissue or in dense lesions. RESULTS Intra and inter-acquisition variabilities were strongly reduced by the standardisation pipeline. Harmonisation by ComBat lowered the percentage of radiomic features significantly different between the three coils from 87% after bias field correction and MR normalisation to 3% in the gel, while preserving or improving performance of lesion classification in the phantoms. DISCUSSION A dedicated standardisation pipeline was developed to reduce variabilities in breast MRI, which paves the way for robust multi-scanner radiomic studies but needs to be assessed on patient data.
Collapse
Affiliation(s)
- Marie-Judith Saint Martin
- Inserm, Institut Curie,Université Paris-Saclay, Laboratoire d'Imagerie Translationnelle en Oncologie (LITO), Centre de Recherche de l'Institut Curie, Bât 101B rue Henri Becquerel, 91401, Orsay, France.
| | - Fanny Orlhac
- Inserm, Institut Curie,Université Paris-Saclay, Laboratoire d'Imagerie Translationnelle en Oncologie (LITO), Centre de Recherche de l'Institut Curie, Bât 101B rue Henri Becquerel, 91401, Orsay, France
| | - Pia Akl
- Inserm, Institut Curie,Université Paris-Saclay, Laboratoire d'Imagerie Translationnelle en Oncologie (LITO), Centre de Recherche de l'Institut Curie, Bât 101B rue Henri Becquerel, 91401, Orsay, France
- HCL, Radiologie du Groupement Hospitalier Est, Hôpital Femme Mère Enfant, Unité Fonctionnelle: Imagerie de la Femme, 3 Quai des Célestins, 69002, Lyon, France
- Institut Curie, Service de Radiodiagnostic, 26 rue d'Ulm, 75005, Paris, France
| | - Fahad Khalid
- Inserm, Institut Curie,Université Paris-Saclay, Laboratoire d'Imagerie Translationnelle en Oncologie (LITO), Centre de Recherche de l'Institut Curie, Bât 101B rue Henri Becquerel, 91401, Orsay, France
| | - Christophe Nioche
- Inserm, Institut Curie,Université Paris-Saclay, Laboratoire d'Imagerie Translationnelle en Oncologie (LITO), Centre de Recherche de l'Institut Curie, Bât 101B rue Henri Becquerel, 91401, Orsay, France
| | - Irène Buvat
- Inserm, Institut Curie,Université Paris-Saclay, Laboratoire d'Imagerie Translationnelle en Oncologie (LITO), Centre de Recherche de l'Institut Curie, Bât 101B rue Henri Becquerel, 91401, Orsay, France
| | - Caroline Malhaire
- Inserm, Institut Curie,Université Paris-Saclay, Laboratoire d'Imagerie Translationnelle en Oncologie (LITO), Centre de Recherche de l'Institut Curie, Bât 101B rue Henri Becquerel, 91401, Orsay, France
- Institut Curie, Service de Radiodiagnostic, 26 rue d'Ulm, 75005, Paris, France
| | - Frédérique Frouin
- Inserm, Institut Curie,Université Paris-Saclay, Laboratoire d'Imagerie Translationnelle en Oncologie (LITO), Centre de Recherche de l'Institut Curie, Bât 101B rue Henri Becquerel, 91401, Orsay, France
| |
Collapse
|
58
|
Staal FCR, van der Reijd DJ, Taghavi M, Lambregts DMJ, Beets-Tan RGH, Maas M. Radiomics for the Prediction of Treatment Outcome and Survival in Patients With Colorectal Cancer: A Systematic Review. Clin Colorectal Cancer 2020; 20:52-71. [PMID: 33349519 DOI: 10.1016/j.clcc.2020.11.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 09/03/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023]
Abstract
Prediction of outcome in patients with colorectal cancer (CRC) is challenging as a result of lack of a robust biomarker and heterogeneity between and within tumors. The aim of this review was to assess the current possibilities and limitations of radiomics (on computed tomography [CT], magnetic resonance imaging [MRI], and positron emission tomography [PET]) for the prediction of treatment outcome and long-term outcome in CRC. Medline/PubMed was searched up to August 2020 for studies that used radiomics for the prediction of response to treatment and survival in patients with CRC (based on pretreatment imaging). The Quality Assessment of Diagnostic Accuracy Studies (QUADAS) tool and Radiomics Quality Score (RQS) were used for quality assessment. A total of 76 studies met the inclusion criteria and were included for further analysis. Radiomics analyses were performed on MRI in 41 studies, on CT in 30 studies, and on 18F-FDG-PET/CT in 10 studies. Heterogeneous results were reported regarding radiomics methods and included features. High-quality studies (n = 13), consisting mainly of MRI-based radiomics to predict response in rectal cancer, were able to predict response with good performance. Radiomics literature in CRC is highly heterogeneous, but it nonetheless holds promise for the prediction of outcome. The most evidence is available for MRI-based radiomics in rectal cancer. Future radiomics research in CRC should focus on independent validation of existing models rather than on developing new models.
Collapse
Affiliation(s)
- Femke C R Staal
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Denise J van der Reijd
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marjaneh Taghavi
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Doenja M J Lambregts
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Regina G H Beets-Tan
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands; Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Monique Maas
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
59
|
Arefan D, Chai R, Sun M, Zuley ML, Wu S. Machine learning prediction of axillary lymph node metastasis in breast cancer: 2D versus 3D radiomic features. Med Phys 2020; 47:6334-6342. [PMID: 33058224 DOI: 10.1002/mp.14538] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 08/07/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The purpose of this study was to distinguish axillary lymph node (ALN) status using preoperative breast DCE-MRI radiomics and compare the effects of two-dimensional (2D) and three-dimensional (3D) analysis. METHODS A retrospective study including 154 breast cancer patients all confirmed by pathology; 80 with ALN metastasis and 74 without. All MRI scans were achieved at a 3.0 Tesla scanner with 7 post-contrast MR phases sequentially acquired with a temporal resolution of 60 s. MRI radiomic features were extracted separately from a 2D single slice (i.e., the representative slice) and the 3D tumor volume. Several machine learning classifiers were built and compared using 2D or 3D analysis to distinguish positive vs negative ALN status. We performed independent test and 10-fold cross validation with multiple repetitions, and used bootstrap test, least absolute shrinkage selection operator, and receiver operating characteristic (ROC) curve analysis as statistical tests. RESULTS The highest area under the ROC curve (AUC) was 0.81 (95% confidence intervals [CI]: 0.80-0.83) and 0.82 (95% CI: 0.81-0.82) for 2D and 3D analysis, respectively; the corresponding accuracy was 79% and 80%. The linear discriminant analysis (LDA) classifier achieved the highest classification performance. None of the AUC differences between 2D and 3D analysis was statistically significant for the several tested machine learning classifiers (all P> 0.05). CONCLUSIONS Radiomic features from segmented tumor region in breast MRI were associated with ALN status. The separate radiomic analysis on 3D tumor volume showed a similar effect to the 2D analysis on the single representative slice in the tested machine learning classifiers.
Collapse
Affiliation(s)
- Dooman Arefan
- Department of Radiology, University of Pittsburgh, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
| | - Ruimei Chai
- Department of Radiology, First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Min Sun
- UPMC Hillman Cancer Center at St. Margaret, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15215, USA
| | - Margarita L Zuley
- Department of Radiology, University of Pittsburgh, School of Medicine, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA.,Magee-Womens Hospital of University of Pittsburgh Medical Center, 300 Halket St, Pittsburgh, PA, 15213, USA
| | - Shandong Wu
- Departments of Radiology of Biomedical Informatics of Bioengineering, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
| |
Collapse
|
60
|
Li Q, Xiao Q, Li J, Duan S, Wang H, Gu Y. MRI-Based Radiomic Signature as a Prognostic Biomarker for HER2-Positive Invasive Breast Cancer Treated with NAC. Cancer Manag Res 2020; 12:10603-10613. [PMID: 33149669 PMCID: PMC7602910 DOI: 10.2147/cmar.s271876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/19/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose To identify MRI-based radiomics signature (Rad-score) as a biomarker of risk stratification for disease-free survival (DFS) in patients with HER2-positive invasive breast cancer treated with trastuzumab-based neoadjuvant chemotherapy (NAC) and establish a radiomics-clinicoradiologic-based nomogram that combines Rad-score, MRI findings, and clinicopathological variables for DFS estimation. Patients and Methods A total of 127 patients were divided into a training set and testing set according to the ratio of 7:3. Radiomic features were extracted from multiphase CE-MRI (CEm). Rad-score was calculated using the LASSO (least absolute shrinkage and selection operator) regression analysis. The cutoff point of Rad-score to divide the patients into high- and low-risk groups was determined by receiver operating characteristic curve analysis. A Kaplan–Meier survival curves and the Log rank test were used to investigate the association of the Rad-score with DFS. Univariate and multivariate Cox proportional hazards model were used to determine the association of Rad-score, MRI features, and clinicopathological variables with DFS. A radiomics-clinicoradiologic-based nomogram combining the Rad-score, MRI features, and clinicopathological findings was plotted to validate the radiomic signatures for DFS estimation. Results The Rad-score stratified patients into high- and low-risk groups for DFS in the training set (P<0.0001) and was validated in the testing set (P=0.002). The radiomics-clinicoradiologic-based nomogram estimated DFS (training set: C-index=0.974, 95% confidence interval (CI)=0.954–0.994; testing set: C-index=0.917, 95% CI=0.842–0.991) better than the clinicoradiologic-based nomogram (training set: C-index=0.855, 95% CI=0.739–0.971; testing set: C-index=0.831, 95% CI=0.643–0.999). Conclusion The Rad-score is an independent biomarker for the estimation of DFS in invasive HER2-positive breast cancer with NAC and the radiomics-clinicoradiologic-based nomogram improved individualized DFS estimation.
Collapse
Affiliation(s)
- Qin Li
- Shanghai Institute of Medical Imaging, Shanghai, China.,Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qin Xiao
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jianwei Li
- Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | | | - He Wang
- Shanghai Center for Mathematical Sciences, Fudan University, Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
61
|
Radiogenomic signatures reveal multiscale intratumour heterogeneity associated with biological functions and survival in breast cancer. Nat Commun 2020; 11:4861. [PMID: 32978398 PMCID: PMC7519071 DOI: 10.1038/s41467-020-18703-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 09/08/2020] [Indexed: 12/24/2022] Open
Abstract
Advanced tumours are often heterogeneous, consisting of subclones with various genetic alterations and functional roles. The precise molecular features that characterize the contributions of multiscale intratumour heterogeneity to malignant progression, metastasis, and poor survival are largely unknown. Here, we address these challenges in breast cancer by defining the landscape of heterogeneous tumour subclones and their biological functions using radiogenomic signatures. Molecular heterogeneity is identified by a fully unsupervised deconvolution of gene expression data. Relative prevalence of two subclones associated with cell cycle and primary immunodeficiency pathways identifies patients with significantly different survival outcomes. Radiogenomic signatures of imaging scale heterogeneity are extracted and used to classify patients into groups with distinct subclone compositions. Prognostic value is confirmed by survival analysis accounting for clinical variables. These findings provide insight into how a radiogenomic analysis can identify the biological activities of specific subclones that predict prognosis in a noninvasive and clinically relevant manner. Tumours are made up of heterogeneous subclones. Here, the authors show using breast cancer imaging and gene expression datasets that these subclones can be inferred by the deconvolution of gene expression data, mapped to MRI derived radiogenomic signatures and used to estimate prognosis.
Collapse
|
62
|
Tan H, Gan F, Wu Y, Zhou J, Tian J, Lin Y, Wang M. Preoperative Prediction of Axillary Lymph Node Metastasis in Breast Carcinoma Using Radiomics Features Based on the Fat-Suppressed T2 Sequence. Acad Radiol 2020; 27:1217-1225. [PMID: 31879160 DOI: 10.1016/j.acra.2019.11.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/01/2019] [Accepted: 11/03/2019] [Indexed: 12/13/2022]
Abstract
RATIONALE AND OBJECTIVES To investigate the value of radiomics method based on the fat-suppressed T2 sequence for preoperative predicting axillary lymph node (ALN) metastasis in breast carcinoma. MATERIALS AND METHODS The data of 329 invasive breast cancer patients were divided into the primary cohort (n = 269) and validation cohort (n = 60). Radiomics features were extracted from the fat-suppressed T2-weighted images on breast MRI, and ALN metastasis-related radiomics feature selection was performed using Mann-Whitney U-test and support vector machines with recursive feature elimination; then a radiomics signature was constructed by linear support vector machine. The predictive models were constructed using a linear regression model based on the clinicopathologic factors and radiomics signature, and nomogram was used for a visual prediction of the combined model. The predictive performances are evaluated with the sensitivity, specificity, accuracy, and area under the receiver operating characteristic curve. RESULTS A total of 647 radiomics features were extracted from each patient. About 23 ALN metastasis-related radiomics features were selected to construct the radiomics signature, including 17 texture features, 5 first-order statistical features, and one shape feature; patient age, tumor size, HER2 status, and vascular cancer thrombus accompanied or not were selected to construct the cilinicopathologic feature model. The sensitivity, specificity, accuracy, and are under the curve value of radiomics signature, clinicopathologic feature model, and the nomogram were 65.22%, 81.08%, 75.00%, and 0.819 (95% confidence interval [CI]: 0.776-0.861), 30.44%, 81.08%, 61.67%, and 0.605 (95% CI: 0.571-0.624) and 60.87%, 89.19%, 78.33%, and 0.810 (95% CI: 0.761-0.855), respectively. CONCLUSION Radiomics methods based on the fat-suppressed T2 sequence and the nomogram are helpful for preoperative accurate predicting ALN metastasis.
Collapse
Affiliation(s)
- Hongna Tan
- Department of Radiology, Henan Provincial People's Hospital & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province & People's Hospital of Zhengzhou University, 7 Road, Weiwu Road, Jinshui District, Zhengzhou 450003, Henan, China
| | - Fuwen Gan
- Collaborative Innovation Center for Internet Healthcare & School of Information Engineering, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yaping Wu
- Department of Radiology, Henan Provincial People's Hospital & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province & People's Hospital of Zhengzhou University, 7 Road, Weiwu Road, Jinshui District, Zhengzhou 450003, Henan, China
| | - Jing Zhou
- Department of Radiology, Henan Provincial People's Hospital & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province & People's Hospital of Zhengzhou University, 7 Road, Weiwu Road, Jinshui District, Zhengzhou 450003, Henan, China
| | - Jie Tian
- Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yusong Lin
- Collaborative Innovation Center for Internet Healthcare & School of Information Engineering, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital & Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province & People's Hospital of Zhengzhou University, 7 Road, Weiwu Road, Jinshui District, Zhengzhou 450003, Henan, China.
| |
Collapse
|
63
|
Fan M, Liu Z, Xu M, Wang S, Zeng T, Gao X, Li L. Generative adversarial network-based super-resolution of diffusion-weighted imaging: Application to tumour radiomics in breast cancer. NMR IN BIOMEDICINE 2020; 33:e4345. [PMID: 32521567 DOI: 10.1002/nbm.4345] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/19/2020] [Accepted: 05/14/2020] [Indexed: 06/11/2023]
Abstract
Diffusion-weighted imaging (DWI) is increasingly used to guide the clinical management of patients with breast tumours. However, accurate tumour characterization with DWI and the corresponding apparent diffusion coefficient (ADC) maps are challenging due to their limited resolution. This study aimed to produce super-resolution (SR) ADC images and to assess the clinical utility of these SR images by performing a radiomic analysis for predicting the histologic grade and Ki-67 expression status of breast cancer. To this end, 322 samples of dynamic enhanced magnetic resonance imaging (DCE-MRI) and the corresponding DWI data were collected. A SR generative adversarial (SRGAN) and an enhanced deep SR (EDSR) network along with the bicubic interpolation were utilized to generate SR-ADC images from which radiomic features were extracted. The dataset was randomly separated into a development dataset (n = 222) to establish a deep SR model using DCE-MRI and a validation dataset (n = 100) to improve the resolution of ADC images. This random separation of datasets was performed 10 times, and the results were averaged. The EDSR method was significantly better than the SRGAN and bicubic methods in terms of objective quality criteria. Univariate and multivariate predictive models of radiomic features were established to determine the area under the receiver operating characteristic curve (AUC). Individual features from the tumour SR-ADC images showed a higher performance with the EDSR and SRGAN methods than with the bicubic method and the original images. Multivariate analysis of the collective radiomics showed that the EDSR- and SRGAN-based SR-ADC images performed better than the bicubic method and original images in predicting either Ki-67 expression levels (AUCs of 0.818 and 0.801, respectively) or the tumour grade (AUCs of 0.826 and 0.828, respectively). This work demonstrates that in addition to improving the resolution of ADC images, deep SR networks can also improve tumour image-based diagnosis in breast cancer.
Collapse
Affiliation(s)
- Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China
| | - Zuhui Liu
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China
| | - Maosheng Xu
- Department of Radiology, First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, Hangzhou, China
| | - Shiwei Wang
- Department of Radiology, First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, Hangzhou, China
| | - Tieyong Zeng
- Department of Mathematics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Xin Gao
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Lihua Li
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China
| |
Collapse
|
64
|
Preliminary study on discriminating HER2 2+ amplification status of breast cancers based on texture features semi-automatically derived from pre-, post-contrast, and subtraction images of DCE-MRI. PLoS One 2020; 15:e0234800. [PMID: 32555662 PMCID: PMC7299320 DOI: 10.1371/journal.pone.0234800] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 06/02/2020] [Indexed: 01/10/2023] Open
Abstract
Objective To investigate whether texture features extracted from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) are associated with human epidermal growth factor receptor type 2 (HER2) 2+ status of breast cancer. Materials and methods 92 MRI cases including 52 HER2 2+ positive and 40 negative patients confirmed by fluorescence in situ hybridization were retrospectively selected. The lesion area was semi-automatically delineated, and a total of 488 texture features were respectively extracted from precontrast, postcontrast, and subtraction images. The Student’s t-test or Mann-Whitney U test was performed to identify statistically significant features between different HER2 2+ amplification groups. Least absolute shrinkage and selection operator (LASSO) was used to search for the optimal feature subsets. Three machine learning classifiers, logistic regression analysis (LRA), quadratic discriminant analysis (QDA), and support vector machine (SVM), were used with a leave-one-out cross validation method to establish the classification models of HER2 2+ status. Classification performance was evaluated by receiver operating characteristic (ROC) analysis. Results Based on the texture analysis with SVM model, the areas under the ROC curve (AUCs) were 0.890 for subtraction images, 0.736 for postcontrast images, and 0.672 for precontrast images, respectively. For LRA model, the AUCs were 0.884, 0.733, and 0.623, respectively. For QDA model, the AUCs were 0.831, 0.726, and 0.568, respectively. LRA and the SVM model with subtraction images reached significantly better performance than the QDA model (P = 0.0227 and P = 0.0088, respectively). Conclusion Texture features of breast cancer extracted from DCE-MRI are associated with HER2 2+ status. Additional studies are necessary to confirm the present preliminary findings.
Collapse
|
65
|
El Adoui M, Drisis S, Benjelloun M. Multi-input deep learning architecture for predicting breast tumor response to chemotherapy using quantitative MR images. Int J Comput Assist Radiol Surg 2020; 15:1491-1500. [DOI: 10.1007/s11548-020-02209-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022]
|
66
|
Sutton EJ, Onishi N, Fehr DA, Dashevsky BZ, Sadinski M, Pinker K, Martinez DF, Brogi E, Braunstein L, Razavi P, El-Tamer M, Sacchini V, Deasy JO, Morris EA, Veeraraghavan H. A machine learning model that classifies breast cancer pathologic complete response on MRI post-neoadjuvant chemotherapy. Breast Cancer Res 2020; 22:57. [PMID: 32466777 PMCID: PMC7254668 DOI: 10.1186/s13058-020-01291-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 05/10/2020] [Indexed: 12/13/2022] Open
Abstract
Background For breast cancer patients undergoing neoadjuvant chemotherapy (NAC), pathologic complete response (pCR; no invasive or in situ) cannot be assessed non-invasively so all patients undergo surgery. The aim of our study was to develop and validate a radiomics classifier that classifies breast cancer pCR post-NAC on MRI prior to surgery. Methods This retrospective study included women treated with NAC for breast cancer from 2014 to 2016 with (1) pre- and post-NAC breast MRI and (2) post-NAC surgical pathology report assessing response. Automated radiomics analysis of pre- and post-NAC breast MRI involved image segmentation, radiomics feature extraction, feature pre-filtering, and classifier building through recursive feature elimination random forest (RFE-RF) machine learning. The RFE-RF classifier was trained with nested five-fold cross-validation using (a) radiomics only (model 1) and (b) radiomics and molecular subtype (model 2). Class imbalance was addressed using the synthetic minority oversampling technique. Results Two hundred seventy-three women with 278 invasive breast cancers were included; the training set consisted of 222 cancers (61 pCR, 161 no-pCR; mean age 51.8 years, SD 11.8), and the independent test set consisted of 56 cancers (13 pCR, 43 no-pCR; mean age 51.3 years, SD 11.8). There was no significant difference in pCR or molecular subtype between the training and test sets. Model 1 achieved a cross-validation AUROC of 0.72 (95% CI 0.64, 0.79) and a similarly accurate (P = 0.1) AUROC of 0.83 (95% CI 0.71, 0.94) in both the training and test sets. Model 2 achieved a cross-validation AUROC of 0.80 (95% CI 0.72, 0.87) and a similar (P = 0.9) AUROC of 0.78 (95% CI 0.62, 0.94) in both the training and test sets. Conclusions This study validated a radiomics classifier combining radiomics with molecular subtypes that accurately classifies pCR on MRI post-NAC.
Collapse
Affiliation(s)
- Elizabeth J Sutton
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Natsuko Onishi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Duc A Fehr
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brittany Z Dashevsky
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meredith Sadinski
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katja Pinker
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Danny F Martinez
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Edi Brogi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lior Braunstein
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mahmoud El-Tamer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Virgilio Sacchini
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth A Morris
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Harini Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
67
|
Conti A, Duggento A, Indovina I, Guerrisi M, Toschi N. Radiomics in breast cancer classification and prediction. Semin Cancer Biol 2020; 72:238-250. [PMID: 32371013 DOI: 10.1016/j.semcancer.2020.04.002] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022]
Abstract
Breast Cancer (BC) is the common form of cancer in women. Its diagnosis and screening are usually performed through different imaging modalities such as mammography, magnetic resonance imaging and ultrasound. However, mammography and ultrasound-imaging techniques have limited sensitivity and specificity both in identifying lesions and in differentiating malign from benign lesions, especially in presence of dense breast parenchyma. Due to the higher resolution of magnetic resonance images, MRI represents the method with the higher specificity and sensitivity among all the available tools, in both lesions' identification and diagnosis. However, especially for diagnosis, even MRI has limitations that are only partially solved if combined with mammography. Unfortunately, due to the limits of all these imaging tools, in order to have a certain diagnosis, patients often receive painful and costly bioptics procedures. In this context, several computational approaches have been developed to increase sensitivity, while maintaining the same specificity, in BC diagnosis and screening. Amongst these, radiomics has been increasingly gaining ground in oncology to improve cancer diagnosis, prognosis and treatment. Radiomics derives multiple quantitative features from single or multiple medical imaging modalities, highlighting image traits which are not visible to the naked eye and hence significantly augmenting the discriminatory and predictive potential of medical imaging. This review article aims to summarize the state of the art in radiomics-based BC research. The dominating evidence extracted from the literature points towards a high potential of radiomics in disentangling malignant from benign breast lesions, classifying BC types and grades and also in predicting treatment response and recurrence risk. In the era of personalized medicine, radiomics has the potential to improve diagnosis, prognosis, prediction, monitoring, image-based intervention, and assessment of therapeutic response in BC.
Collapse
Affiliation(s)
- Allegra Conti
- Laboratory of Neuromotor Physiology, IRCCS Santa Lucia Foundation, Via Ardeatina, 306, 00179, Rome, Italy; Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| | - Andrea Duggento
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| | - Iole Indovina
- Laboratory of Neuromotor Physiology, IRCCS Santa Lucia Foundation, Via Ardeatina, 306, 00179, Rome, Italy; Department of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Maria Guerrisi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Boston, MA, United States.
| |
Collapse
|
68
|
Feng B, Chen X, Chen Y, Liu K, Li K, Liu X, Yao N, Li Z, Li R, Zhang C, Ji J, Long W. Radiomics nomogram for preoperative differentiation of lung tuberculoma from adenocarcinoma in solitary pulmonary solid nodule. Eur J Radiol 2020; 128:109022. [PMID: 32371184 DOI: 10.1016/j.ejrad.2020.109022] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/05/2020] [Accepted: 04/13/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE To investigate the preoperative differential diagnostic performance of a radiomics nomogram in tuberculous granuloma (TBG) and lung adenocarcinoma (LAC) appearing as solitary pulmonary solid nodules (SPSNs). METHOD We retrospectively recruited 426 patients with SPSNs from two centers and assigned them to training (n = 123), internal validation (n = 121), and external validation cohorts (n = 182). A model of deep learning (DL) was built for tumor segmentation from routine computed tomography (CT) images and extraction of 3D radiomics features. We used the least absolute shrinkage and selection operator (LASSO) logistic regression to build a radiomics signature. A clinical model was developed with clinical factors, including age, gender, and CT-based subjective findings (eg, lesion size, lesion location, lesion margin, lobulated sharp, and spiculation sign). We constructed individualized radiomics nomograms incorporating the radiomics signature and clinical factors to validate the diagnostic ability. RESULTS Three factors - radiomics signature, age, and spiculation sign - were found to be independent predictors and were used to build the radiomics nomogram, which showed better diagnostic accuracy than any single model (all net reclassification improvement p < 0.05). The area under curve yielded was 0.9660 (95% confidence interval [CI], 0.9390-0.9931), 0.9342 (95% CI, 0.8944-0.9739), and 0.9064 (95% CI, 0.8639-0.9490) for the training, internal validation, and external validation cohorts, respectively. Decision curve analysis (DCA) and stratification analysis showed the nomogram has potential for generalizability. CONCLUSION The radiomics nomogram we developed can preoperatively distinguish between LAC and TBG in patient with a SPSN.
Collapse
Affiliation(s)
- Bao Feng
- The Department of Radiology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China; School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi Province, China
| | - Xiangmeng Chen
- The Department of Radiology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China
| | - Yehang Chen
- School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi Province, China
| | - Kunfeng Liu
- The Department of Radiology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Kunwei Li
- The Department of Radiology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Xueguo Liu
- The Department of Radiology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Nan Yao
- The Department of Radiology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China
| | - Zhi Li
- School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi Province, China
| | - Ronggang Li
- The Department of Pathology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China
| | - Chaotong Zhang
- The Department of Radiology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China
| | - Jianbo Ji
- School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi Province, China
| | - Wansheng Long
- The Department of Radiology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China.
| |
Collapse
|
69
|
Teixeira SRC, de Camargo Júnior HSA, Cabello C. Background parenchymal enhancement: behavior during neoadjuvant chemotherapy for breast cancer and relationship with a pathological complete response. Radiol Bras 2020; 53:95-104. [PMID: 32336824 PMCID: PMC7170574 DOI: 10.1590/0100-3984.2019.0057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Objective To evaluate background parenchymal enhancement (BPE) and its characteristics, as well as its behavior before and after neoadjuvant chemotherapy (NAC), in both breasts of patients with unilateral breast cancer. Materials and Methods This was a retrospective, cross-sectional observational study involving a consecutive sample of women with invasive breast cancer who underwent breast magnetic resonance imaging (MRI) between July 2007 and July 2017, as well as undergoing dynamic contrast-enhanced MRI before and after NAC. In both breasts, we evaluated the BPE in accordance with the Breast Imaging Reporting and Data System. We applied logistic regression analysis, and values of p < 0.05 were considered significant. Results We evaluated 150 women. The mean age was 45.2 years (range, 20-74 years). The variables correlating independently with a high pre-NAC BPE, in the affected and contralateral breasts, were being under 50 years of age (odds ratio [OR] = 6.55; 95% confidence interval [95% CI]: 2.32-18.46, for both breasts) and a post-NAC BPE reduction (OR = 17.75; 95% CI: 4.94-63.73 and OR = 18.47; 95% CI: 5.19-66.49, respectively). Conclusion Patients with invasive unilateral breast cancer who have a high pre-NAC BPE in both breasts tend to be under 50 years of age and to show a post-NAC reduction in BPE.
Collapse
Affiliation(s)
- Sandra Regina Campos Teixeira
- Centro de Atenção Integral à Saúde da Mulher da Universidade Estadual de Campinas (Caism-Unicamp), Campinas, SP, Brazil.,CDE - Diagnóstico por Imagem, Campinas, SP, Brazil
| | | | - Cesar Cabello
- Centro de Atenção Integral à Saúde da Mulher da Universidade Estadual de Campinas (Caism-Unicamp), Campinas, SP, Brazil
| |
Collapse
|
70
|
Pesapane F, Suter MB, Rotili A, Penco S, Nigro O, Cremonesi M, Bellomi M, Jereczek-Fossa BA, Pinotti G, Cassano E. Will traditional biopsy be substituted by radiomics and liquid biopsy for breast cancer diagnosis and characterisation? Med Oncol 2020; 37:29. [PMID: 32180032 DOI: 10.1007/s12032-020-01353-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/26/2020] [Indexed: 02/06/2023]
Abstract
The diagnosis of breast cancer currently relies on radiological and clinical evaluation, confirmed by histopathological examination. However, such approach has some limitations as the suboptimal sensitivity, the long turnaround time for recall tests, the invasiveness of the procedure and the risk that some features of target lesions may remain undetected, making re-biopsy a necessity. Recent technological advances in the field of artificial intelligence hold promise in addressing such medical challenges not only in cancer diagnosis, but also in treatment assessment, and monitoring of disease progression. In the perspective of a truly personalised medicine, based on the early diagnosis and individually tailored treatments, two new technologies, namely radiomics and liquid biopsy, are rising as means to obtain information from diagnosis to molecular profiling and response assessment, without the need of a biopsied tissue sample. Radiomics works through the extraction of quantitative peculiar features of cancer from radiological data, while liquid biopsy gets the whole of the malignancy's biology from something as easy as a blood sample. Both techniques hopefully will identify diagnostic and prognostic information of breast cancer potentially reducing the need for invasive (and often difficult to perform) biopsies and favouring an approach that is as personalised as possible for each patient. Nevertheless, such techniques will not substitute tissue biopsy in the near future, and even in further times they will require the aid of other parameters to be correctly interpreted and acted upon.
Collapse
Affiliation(s)
- Filippo Pesapane
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy.
| | | | - Anna Rotili
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Silvia Penco
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Olga Nigro
- Medical Oncology, ASST Sette Laghi, Viale Borri 57, 21100, Varese, VA, Italy
| | - Marta Cremonesi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Massimo Bellomi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Radiology, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Graziella Pinotti
- Medical Oncology, ASST Sette Laghi, Viale Borri 57, 21100, Varese, VA, Italy
| | - Enrico Cassano
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| |
Collapse
|
71
|
Jiang Z, Yin J. Performance evaluation of texture analysis based on kinetic parametric maps from breast DCE-MRI in classifying benign from malignant lesions. J Surg Oncol 2020; 121:1181-1190. [PMID: 32167588 DOI: 10.1002/jso.25901] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/02/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND OBJECTIVES To investigate the performance of texture analysis based on enhancement kinetic parametric maps derived from breast dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) in discriminating benign from malignant tumors. METHODS A total of 192 cases confirmed by histopathology were retrospectively selected from our Picture Archiving and Communication System, including 93 benign and 99 malignant tumors. Lesion areas were delineated semi-automatically, and six kinetic parametric maps reflecting the perfusion information were generated, namely the maximum slope of increase (MSI), slope of signal intensity (SIslope ), initial percentage of peak enhancement (Einitial ), percentage of peak enhancement (Epeak ), early signal enhancement ratio (ESER), and second enhancement percentage (SEP) maps. A total of 286 texture features were extracted from those quantitative parametric maps. The Student t test or Mann-Whitney U test was used to select features that were statistically significantly different between the benign and malignant groups. A support vector machine was employed with a leave-one-out cross-validation method to establish the classification model. Classification performance was evaluated according to the receiver operating characteristic (ROC) theory. RESULTS The areas under ROC curves (AUCs) indicating the diagnostic performance were 0.925 for MSI, 0.854 for SIslope , 0.756 for Einitial , 0.923 for Epeak , 0.871 for ESER and 0.881 for SEP. Significant differences in AUCs were found between Einitial vs MSI, Einitial vs Epeak and Einitial vs SEP (P < .05). There were no significant differences in other pairwise comparisons. CONCLUSION Texture analysis of the kinetic parametric maps derived from breast DCE-MRI can contribute to the discrimination between malignant and benign lesions. It can be considered as a supplementary tool for breast diagnosis.
Collapse
Affiliation(s)
- Zejun Jiang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China.,Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiandong Yin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
72
|
Zhang Q, Peng Y, Liu W, Bai J, Zheng J, Yang X, Zhou L. Radiomics Based on Multimodal MRI for the Differential Diagnosis of Benign and Malignant Breast Lesions. J Magn Reson Imaging 2020; 52:596-607. [PMID: 32061014 DOI: 10.1002/jmri.27098] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND MRI-based radiomics has been used to diagnose breast lesions; however, little research combining quantitative pharmacokinetic parameters of dynamic contrast-enhanced MRI (DCE-MRI) and diffusion kurtosis imaging (DKI) exists. PURPOSE To develop and validate a multimodal MRI-based radiomics model for the differential diagnosis of benign and malignant breast lesions and analyze the discriminative abilities of different MR sequences. STUDY TYPE Retrospective. POPULATION In all, 207 female patients with 207 histopathology-confirmed breast lesions (95 benign and 112 malignant) were included in the study. Then 159 patients were assigned to the training group, and 48 patients comprised the validation group. FIELD STRENGTH/SEQUENCE T2 -weighted (T2 W), T1 -weighted (T1 W), diffusion-weighted MR imaging (b-values = 0, 500, 800, and 2000 seconds/mm2 ) and quantitative DCE-MRI were performed on a 3.0T MR scanner. ASSESSMENT Radiomics features were extracted from T2 WI, T1 WI, DKI, apparent diffusion coefficient (ADC) maps, and DCE pharmacokinetic parameter maps in the training set. Models based on each sequence or combinations of sequences were built using a support vector machine (SVM) classifier and used to differentiate benign and malignant breast lesions in the validation set. STATISTICAL TESTS Optimal feature selection was performed by Spearman's rank correlation coefficients and the least absolute shrinkage and selection operator algorithm (LASSO). Receiver operating characteristic (ROC) curves were used to assess the diagnostic performance of the radiomics models in the validation set. RESULTS The area under the ROC curve (AUC) of the optimal radiomics model, including T2 WI, DKI, and quantitative DCE-MRI parameter maps was 0.921, with an accuracy of 0.833. The AUCs of the models based on T1 WI, T2 WI, ADC map, DKI, and DCE pharmacokinetic parameter maps were 0.730, 0.791, 0.770, 0.788, and 0.836, respectively. DATA CONCLUSION The model based on radiomics features from T2 WI, DKI, and quantitative DCE pharmacokinetic parameter maps has a high discriminatory ability for benign and malignant breast lesions. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY STAGE: 2 J. Magn. Reson. Imaging 2020;52:596-607.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunsong Peng
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Wei Liu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiayuan Bai
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Zheng
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Xiaodong Yang
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Lijuan Zhou
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
73
|
Zhou J, Lu J, Gao C, Zeng J, Zhou C, Lai X, Cai W, Xu M. Predicting the response to neoadjuvant chemotherapy for breast cancer: wavelet transforming radiomics in MRI. BMC Cancer 2020; 20:100. [PMID: 32024483 PMCID: PMC7003343 DOI: 10.1186/s12885-020-6523-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023] Open
Abstract
Background The purpose of this study was to investigate the value of wavelet-transformed radiomic MRI in predicting the pathological complete response (pCR) to neoadjuvant chemotherapy (NAC) for patients with locally advanced breast cancer (LABC). Methods Fifty-five female patients with LABC who underwent contrast-enhanced MRI (CE-MRI) examination prior to NAC were collected for the retrospective study. According to the pathological assessment after NAC, patient responses to NAC were categorized into pCR and non-pCR. Three groups of radiomic textures were calculated in the segmented lesions, including (1) volumetric textures, (2) peripheral textures, and (3) wavelet-transformed textures. Six models for the prediction of pCR were Model I: group (1), Model II: group (1) + (2), Model III: group (3), Model IV: group (1) + (3), Model V: group (2) + (3), and Model VI: group (1) + (2) + (3). The performance of predicting models was compared using the area under the receiver operating characteristic (ROC) curves (AUC). Results The AUCs of the six models for the prediction of pCR were 0.816 ± 0.033 (Model I), 0.823 ± 0.020 (Model II), 0.888 ± 0.025 (Model III), 0.876 ± 0.015 (Model IV), 0.885 ± 0.030 (Model V), and 0.874 ± 0.019 (Model VI). The performance of four models with wavelet-transformed textures (Models III, IV, V, and VI) was significantly better than those without wavelet-transformed textures (Model I and II). In addition, the inclusion of volumetric textures or peripheral textures or both did not result in any improvements in performance. Conclusions Wavelet-transformed textures outperformed volumetric and/or peripheral textures in the radiomic MRI prediction of pCR to NAC for patients with LABC, which can potentially serve as a surrogate biomarker for the prediction of the response of LABC to NAC.
Collapse
Affiliation(s)
- Jiali Zhou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Shangcheng District, Hangzhou, 310006, People's Republic of China.,The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China.,Ningbo First Hospital, Ningbo, China
| | - Jinghui Lu
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 25 New Chardon St., 400C, Boston, MA, 02114, USA
| | - Chen Gao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Shangcheng District, Hangzhou, 310006, People's Republic of China.,The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingjing Zeng
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Changyu Zhou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Shangcheng District, Hangzhou, 310006, People's Republic of China.,The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaobo Lai
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Shangcheng District, Hangzhou, 310006, People's Republic of China.,The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenli Cai
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 25 New Chardon St., 400C, Boston, MA, 02114, USA.
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Shangcheng District, Hangzhou, 310006, People's Republic of China. .,The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
74
|
Ye DM, Wang HT, Yu T. The Application of Radiomics in Breast MRI: A Review. Technol Cancer Res Treat 2020; 19:1533033820916191. [PMID: 32347167 PMCID: PMC7225803 DOI: 10.1177/1533033820916191] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/21/2020] [Accepted: 02/27/2020] [Indexed: 12/13/2022] Open
Abstract
Breast cancer has been a worldwide burden of women's health. Although concerns have been raised for early diagnosis and timely treatment, the efforts are still needed for precision medicine and individualized treatment. Radiomics is a new technology with immense potential to obtain mineable data to provide rich information about the diagnosis and prognosis of breast cancer. In our study, we introduced the workflow and application of radiomics as well as its outlook and challenges based on published studies. Radiomics has the potential ability to differentiate between malignant and benign breast lesions, predict axillary lymph node status, molecular subtypes of breast cancer, tumor response to chemotherapy, and survival outcomes. Our study aimed to help clinicians and radiologists to know the basic information of radiomics and encourage cooperation with scientists to mine data for better application in clinical practice.
Collapse
Affiliation(s)
- Dong-Man Ye
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People’s Republic of China
| | - Hao-Tian Wang
- Dalian Medical University, The First Clinical College, Dalian, Liaoning Province, People’s Republic of China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, People’s Republic of China
| |
Collapse
|
75
|
Sollini M, Antunovic L, Chiti A, Kirienko M. Towards clinical application of image mining: a systematic review on artificial intelligence and radiomics. Eur J Nucl Med Mol Imaging 2019; 46:2656-2672. [PMID: 31214791 PMCID: PMC6879445 DOI: 10.1007/s00259-019-04372-x] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 05/23/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE The aim of this systematic review was to analyse literature on artificial intelligence (AI) and radiomics, including all medical imaging modalities, for oncological and non-oncological applications, in order to assess how far the image mining research stands from routine medical application. To do this, we applied a trial phases classification inspired from the drug development process. METHODS Among the articles we considered for inclusion from PubMed were multimodality AI and radiomics investigations, with a validation analysis aimed at relevant clinical objectives. Quality assessment of selected papers was performed according to the QUADAS-2 criteria. We developed the phases classification criteria for image mining studies. RESULTS Overall 34,626 articles were retrieved, 300 were selected applying the inclusion/exclusion criteria, and 171 high-quality papers (QUADAS-2 ≥ 7) were identified and analysed. In 27/171 (16%), 141/171 (82%), and 3/171 (2%) studies the development of an AI-based algorithm, radiomics model, and a combined radiomics/AI approach, respectively, was described. A total of 26/27(96%) and 1/27 (4%) AI studies were classified as phase II and III, respectively. Consequently, 13/141 (9%), 10/141 (7%), 111/141 (79%), and 7/141 (5%) radiomics studies were classified as phase 0, I, II, and III, respectively. All three radiomics/AI studies were categorised as phase II trials. CONCLUSIONS The results of the studies are promising but still not mature enough for image mining tools to be implemented in the clinical setting and be widely used. The transfer learning from the well-known drug development process, with some specific adaptations to the image mining discipline could represent the most effective way for radiomics and AI algorithms to become the standard of care tools.
Collapse
Affiliation(s)
- Martina Sollini
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
| | - Lidija Antunovic
- Nuclear Medicine, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Arturo Chiti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
- Nuclear Medicine, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Margarita Kirienko
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy.
| |
Collapse
|
76
|
Fan M, Yuan W, Zhao W, Xu M, Wang S, Gao X, Li L. Joint Prediction of Breast Cancer Histological Grade and Ki-67 Expression Level Based on DCE-MRI and DWI Radiomics. IEEE J Biomed Health Inform 2019; 24:1632-1642. [PMID: 31794406 DOI: 10.1109/jbhi.2019.2956351] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Histologic grade and Ki-67 proliferation status are important clinical indictors for breast cancer prognosis and treatment. The purpose of this study is to improve prediction accuracy of these clinical indicators based on tumor radiomic analysis. METHODS We jointly predicted Ki-67 and tumor grade with a multitask learning framework by separately utilizing radiomics from tumor MRI series. Additionally, we showed how multitask learning models (MTLs) could be extended to combined radiomics from the MRI series for a better prediction based on the assumption that features from different sources of images share common patterns while providing complementary information. Tumor radiomic analysis was performed with morphological, statistical and textural features extracted on the DWI and dynamic contrast-enhanced MRI (DCE-MRI) series of the precontrast and subtraction images, respectively. RESULTS Joint prediction of Ki-67 status and tumor grade on MR images using the MTL achieved performance improvements over that of single-task-based predictive models. Similarly, for the prediction tasks of Ki-67 and tumor grade, the MTL for combined precontrast and apparent diffusion coefficient (ADC) images achieved AUCs of 0.811 and 0.816, which were significantly better than that of the single-task- based model with p values of 0.005 and 0.017, respectively. CONCLUSION Mapping MRI radiomics to two related clinical indicators improves prediction performance for both Ki-67 expression level and tumor grade. SIGNIFICANCE Joint prediction of indicators by multitask learning that combines correlations of MRI radiomics is important for optimal tumor therapy and treatment because clinical decisions are made by integrating multiple clinical indicators.
Collapse
|
77
|
Eun NL, Kang D, Son EJ, Park JS, Youk JH, Kim JA, Gweon HM. Texture Analysis with 3.0-T MRI for Association of Response to Neoadjuvant Chemotherapy in Breast Cancer. Radiology 2019; 294:31-41. [PMID: 31769740 DOI: 10.1148/radiol.2019182718] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Previous studies have suggested that texture analysis is a promising tool in the diagnosis, characterization, and assessment of treatment response in various cancer types. Therefore, application of texture analysis may be helpful for early prediction of pathologic response in breast cancer. Purpose To investigate whether texture analysis of features from MRI is associated with pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC) in breast cancer. Materials and Methods This retrospective study included 136 women (mean age, 47.9 years; range, 31-70 years) who underwent NAC and subsequent surgery for breast cancer between January 2012 and August 2017. Patients were monitored with 3.0-T MRI before (pretreatment) and after (midtreatment) three or four cycles of NAC. Texture analysis was performed at pre- and midtreatment T2-weighted MRI, contrast material-enhanced T1-weighted MRI, diffusion-weighted MRI, and apparent diffusion coefficient (ADC) mapping by using commercial software. A random forest method was applied to build a predictive model for classifying those with pCR with use of texture parameters. Diagnostic performance for predicting pCR was assessed and compared with that of six other machine learning classifiers (adaptive boosting, decision tree, k-nearest neighbor, linear support vector machine, naive Bayes, and linear discriminant analysis) by using the Wald test and DeLong method. Results Forty of the 136 patients (29%) achieved pCR after NAC. In the prediction of pCR, the random forest classifier showed the lowest diagnostic performance with pretreatment ADC (area under the receiver operating characteristic curve [AUC], 0.53; 95% confidence interval: 0.44, 0.61) and the highest diagnostic performance with midtreatment contrast-enhanced T1-weighted MRI (AUC, 0.82; 95% confidence interval: 0.74, 0.88) among pre- and midtreatment T2-weighted MRI, contrast-enhanced T1-weighted MRI, diffusion-weighted MRI, and ADC mapping. Conclusion Texture parameters using a random forest method of contrast-enhanced T1-weighted MRI at midtreatment of neoadjuvant chemotherapy were valuable and associated with pathologic complete response in breast cancer. © RSNA, 2019 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Na Lae Eun
- From the Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, 06273 Seoul, Republic of Korea (N.L.E., E.J.S., J.H.Y., J.A.K., H.M.G.); Department of Radiology, Hanyang University, College of Medicine, Seoul, Republic of Korea (N.L.E., J.S.P.); and Department of Healthcare Information Technology, Inje University, Gimhae, Republic of Korea (D.K.)
| | - Daesung Kang
- From the Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, 06273 Seoul, Republic of Korea (N.L.E., E.J.S., J.H.Y., J.A.K., H.M.G.); Department of Radiology, Hanyang University, College of Medicine, Seoul, Republic of Korea (N.L.E., J.S.P.); and Department of Healthcare Information Technology, Inje University, Gimhae, Republic of Korea (D.K.)
| | - Eun Ju Son
- From the Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, 06273 Seoul, Republic of Korea (N.L.E., E.J.S., J.H.Y., J.A.K., H.M.G.); Department of Radiology, Hanyang University, College of Medicine, Seoul, Republic of Korea (N.L.E., J.S.P.); and Department of Healthcare Information Technology, Inje University, Gimhae, Republic of Korea (D.K.)
| | - Jeong Seon Park
- From the Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, 06273 Seoul, Republic of Korea (N.L.E., E.J.S., J.H.Y., J.A.K., H.M.G.); Department of Radiology, Hanyang University, College of Medicine, Seoul, Republic of Korea (N.L.E., J.S.P.); and Department of Healthcare Information Technology, Inje University, Gimhae, Republic of Korea (D.K.)
| | - Ji Hyun Youk
- From the Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, 06273 Seoul, Republic of Korea (N.L.E., E.J.S., J.H.Y., J.A.K., H.M.G.); Department of Radiology, Hanyang University, College of Medicine, Seoul, Republic of Korea (N.L.E., J.S.P.); and Department of Healthcare Information Technology, Inje University, Gimhae, Republic of Korea (D.K.)
| | - Jeong-Ah Kim
- From the Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, 06273 Seoul, Republic of Korea (N.L.E., E.J.S., J.H.Y., J.A.K., H.M.G.); Department of Radiology, Hanyang University, College of Medicine, Seoul, Republic of Korea (N.L.E., J.S.P.); and Department of Healthcare Information Technology, Inje University, Gimhae, Republic of Korea (D.K.)
| | - Hye Mi Gweon
- From the Department of Radiology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, 06273 Seoul, Republic of Korea (N.L.E., E.J.S., J.H.Y., J.A.K., H.M.G.); Department of Radiology, Hanyang University, College of Medicine, Seoul, Republic of Korea (N.L.E., J.S.P.); and Department of Healthcare Information Technology, Inje University, Gimhae, Republic of Korea (D.K.)
| |
Collapse
|
78
|
Exploring breast cancer response prediction to neoadjuvant systemic therapy using MRI-based radiomics: A systematic review. Eur J Radiol 2019; 121:108736. [PMID: 31734639 DOI: 10.1016/j.ejrad.2019.108736] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/26/2019] [Accepted: 10/31/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE MRI-based tumor response prediction to neoadjuvant systemic therapy (NST) in breast cancer patients is increasingly being studied using radiomics with outcomes that appear to be promising. The aim of this study is to systematically review the current literature and reflect on its quality. METHODS PubMed and EMBASE databases were systematically searched for studies investigating MRI-based radiomics for tumor response prediction. Abstracts were screened by two reviewers independently. The quality of the radiomics workflow of eligible studies was assessed using the Radiomics Quality Score (RQS). An overview of the methodologies used in steps of the radiomics workflow and current results are presented. RESULTS Sixteen studies were included with cohort sizes ranging from 35 to 414 patients. The RQS scores varied from 0 % to 41.2 %. Methodologies in the radiomics workflow varied greatly, especially region of interest segmentation, features selection, and model development with heterogeneous outcomes as a result. Seven studies applied univariate analysis and nine studies applied multivariate analysis. Most studies performed their analysis on the pretreatment dynamic contrast-enhanced T1-weighted sequence. Entropy was the best performing individual feature with AUC values ranging from 0.83 to 0.85. The best performing multivariate prediction model, based on logistic regression analysis, scored a validation AUC of 0.94. CONCLUSION This systematic review revealed large methodological heterogeneity for each step of the MRI-based radiomics workflow, consequently, the (overall promising) results are difficult to compare. Consensus for standardization of MRI-based radiomics workflow for tumor response prediction to NST in breast cancer patients is needed to further improve research.
Collapse
|
79
|
Liu B, Cheng J, Guo DJ, He XJ, Luo YD, Zeng Y, Li CM. Prediction of prostate cancer aggressiveness with a combination of radiomics and machine learning-based analysis of dynamic contrast-enhanced MRI. Clin Radiol 2019; 74:896.e1-896.e8. [PMID: 31495546 DOI: 10.1016/j.crad.2019.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022]
Abstract
AIM To investigate whether the combination of radiomics and automatic machine learning-based classification of original images from multiphase dynamic contrast-enhanced (DCE)-magnetic resonance imaging (MRI) can predict prostate cancer (PCa) aggressiveness before biopsy. MATERIALS AND METHODS Forty consecutive biopsy-confirmed PCa patients were included. Biopsy was performed within 4 weeks after the DCE-MRI examinations. According to the time-signal-intensity curve, lesion segmentation was performed on the first and on the strongest phase of the enhancement on the original DCE-MRI images, and 1,029 quantitative radiomics features were calculated automatically from each lesion, wherein there were three datasets available (Dataset-F, Dataset-S and Dataset-FS). The variance threshold method, select k-best method and least absolute shrinkage and selection operator (LASSO) algorithm were used to reduce the feature dimensions. Five machine learning approaches leveraging cross-validation were employed, and the clinical value of each model was evaluated by area under the receiver operating characteristic curve (AUC). Correlation analysis was performed between the features of the machine learning model that achieved the best classification performance and the Gleason score (GS) of the PCa lesion. RESULTS Eight, four, and 16 features were selected as optimal subsets in Dataset-F, -S and -FS, respectively. Among all three datasets, logistic regression (LR)-based analysis with Dataset-FS had the highest predication efficacy (AUC=0.93). Ten features in Dataset-FS showed significantly positively correlation with GS. The model performance of Dataset-F was generally better than that in Dataset-S. CONCLUSIONS A combination of radiomics and machine learning-analysis based analysis of the union of the first and strongest phases of original DCE-MRI images can predict PCa aggressiveness non-invasively, accurately, and automatically.
Collapse
Affiliation(s)
- B Liu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Yuzhong District, Chongqing, 400000, China
| | - J Cheng
- Basic Medical College of Chongqing Medical University, No. 1 Medical School Road, Yuzhong District, Chongqing, 400042, China
| | - D J Guo
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Yuzhong District, Chongqing, 400000, China
| | - X J He
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Yuzhong District, Chongqing, 400000, China
| | - Y D Luo
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Yuzhong District, Chongqing, 400000, China
| | - Y Zeng
- Department of Radiology, The Third Affiliated Hospital of Chongqing Medical University, No. 1 Shuanghu Branch Road, Yubei District, Chongqing, 401120, China.
| | - C M Li
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Yuzhong District, Chongqing, 400000, China.
| |
Collapse
|
80
|
Molecular Subtypes Recognition of Breast Cancer in Dynamic Contrast-Enhanced Breast Magnetic Resonance Imaging Phenotypes from Radiomics Data. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2019; 2019:6978650. [PMID: 31827586 PMCID: PMC6885255 DOI: 10.1155/2019/6978650] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/10/2019] [Indexed: 12/28/2022]
Abstract
Background and Objective Breast cancer is a major cause of mortality among women if not treated in early stages. Recognizing molecular markers from DCE-MRI directly to distinguish the four molecular subtypes without invasive biopsy is helpful for guiding treatment plans for breast cancer, which provides a fast way to consequential treatment plan decision in early time and best opportunity for patients. Methods This study presents an approach of molecular subtypes recognition from breast cancer image phenotypes by radiomics. An improved region growth algorithm with dynamic threshold without user interaction is proposed for cancer lesion segmentation, which gives the precise border of lesion other than area with background. The lesions are extracted automatically based on radiologists' annotation which guarantees the lesion is segmented correctly. Various features are extracted on lesions data including texture, morphology, dynamic kinetics, and statistics features carried out on a large patient cohort, which are used to validate the relationship between image phenotypes and the molecular subtypes. A new algorithm of multimodel-based recursive feature elimination is applied on the radiomics data generated by the feature extraction process. This method obtains the feature subset with stable performance for different classification models, and the gradient boosting decision tree model gets the best results of both classification performance and imbalance performance on molecular subtypes. Result From the experimental results, 69 optimal features from 143 original features are found by the multimodel-based recursive feature elimination algorithms and the gradient boosting decision tree classifier obtains a good performance with accuracy 0.87, precise 0.88, recall 0.87, and F1-score 0.87. The dataset with 637 patients in this paper has serious imbalance problem on different molecular subtypes, and the the robust features that are generated by multimodel-based recursive feature eliminiation algorithm make the gradient boosting decision tree classifier have good behaviors. The recognition precision for the four molecular subtypes of luminal A, luminal B, HER-2, and basal-like are 0.91, 0.89, 0.83, and 0.87, respectively. Conclusions The improved lesion segmentation method gives more precise lesion edge, which not only saves the time of automatic extraction of lesion region of interest without threshold setting for each case, but also prevents the segmentation error by manual and prejudice from different radiologists. The feature selection algorithm of multimodel-based recursive feature elimination has the ability to find robust and optimal features that distinguish the four molecular subtypes from image phenotypes. The gradient boosting decision tree classifier rather plays a main role in recognition than other models used in this paper.
Collapse
|
81
|
Fan M, Liu Z, Xie S, Xu M, Wang S, Gao X, Li L. Integration of dynamic contrast-enhanced magnetic resonance imaging and T2-weighted imaging radiomic features by a canonical correlation analysis-based feature fusion method to predict histological grade in ductal breast carcinoma. Phys Med Biol 2019; 64:215001. [PMID: 31470420 DOI: 10.1088/1361-6560/ab3fd3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tumour histological grade has prognostic implications in breast cancer. Tumour features in dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and T2-weighted (T2W) imaging can provide related and complementary information in the analysis of breast lesions to improve MRI-based histological status prediction in breast cancer. A dataset of 167 patients with invasive ductal carcinoma (IDC) was assembled, consisting of 72 low/intermediate-grade and 95 high-grade cases with preoperative DCE-MRI and T2W images. The data cohort was separated into development (n = 111) and validation (n = 56) cohorts. Each tumour was segmented in the precontrast and the intermediate and last postcontrast DCE-MR images and was mapped to the tumour in the T2W images. Radiomic features, including texture, morphology, and histogram distribution features in the tumour image, were extracted for those image series. Features from the DCE-MR and T2W images were fused by a canonical correlation analysis (CCA)-based method. The support vector machine (SVM) classifiers were trained and tested on the development and validation cohorts, respectively. SVM-based recursive feature elimination (SVM-RFE) was adopted to identify the optimal features for prediction. The areas under the ROC curves (AUCs) for the T2W images and the DCE-MRI series of precontrast, intermediate and last postcontrast images were 0.750 ± 0.047, 0.749 ± 0.047, and 0.788 ± 0.045, respectively, for the development cohort and 0.715 ± 0.068, 0.704 ± 0.073, and 0.744 ± 0.067, respectively, for the validation cohort. After the CCA-based fusion of features from the DCE-MRI series and T2W images, the AUCs increased to 0.751 ± 0.065, 0.803 ± 0.0600 and 794 ± 0.060 in the validation cohort. Moreover, the method of fusing features between DCE-MRI and T2W images using CCA achieved better performance than the concatenation-based feature fusion or classifier fusion methods. Our results demonstrated that anatomical and functional MR images yield complementary information, and feature fusion of radiomic features by matrix transformation to optimize their correlations produced a classifier with improved performance for predicting the histological grade of IDC.
Collapse
Affiliation(s)
- Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
82
|
Impact of Machine Learning With Multiparametric Magnetic Resonance Imaging of the Breast for Early Prediction of Response to Neoadjuvant Chemotherapy and Survival Outcomes in Breast Cancer Patients. Invest Radiol 2019; 54:110-117. [PMID: 30358693 DOI: 10.1097/rli.0000000000000518] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE The aim of this study was to assess the potential of machine learning with multiparametric magnetic resonance imaging (mpMRI) for the early prediction of pathological complete response (pCR) to neoadjuvant chemotherapy (NAC) and of survival outcomes in breast cancer patients. MATERIALS AND METHODS This institutional review board-approved prospective study included 38 women (median age, 46.5 years; range, 25-70 years) with breast cancer who were scheduled for NAC and underwent mpMRI of the breast at 3 T with dynamic contrast-enhanced (DCE), diffusion-weighted imaging (DWI), and T2-weighted imaging before and after 2 cycles of NAC. For each lesion, 23 features were extracted: qualitative T2-weighted and DCE-MRI features according to BI-RADS (Breast Imaging Reporting and Data System), quantitative pharmacokinetic DCE features (mean plasma flow, volume distribution, mean transit time), and DWI apparent diffusion coefficient (ADC) values. To apply machine learning to mpMRI, 8 classifiers including linear support vector machine, linear discriminant analysis, logistic regression, random forests, stochastic gradient descent, decision tree, adaptive boosting, and extreme gradient boosting (XGBoost) were used to rank the features. Histopathologic residual cancer burden (RCB) class (with RCB 0 being a pCR), recurrence-free survival (RFS), and disease-specific survival (DSS) were used as the standards of reference. Classification accuracy with area under the receiving operating characteristic curve (AUC) was assessed using all the extracted qualitative and quantitative features for pCR as defined by RCB class, RFS, and DSS using recursive feature elimination. To overcome overfitting, 4-fold cross-validation was used. RESULTS Machine learning with mpMRI achieved stable performance as shown by mean classification accuracies for the prediction of RCB class (AUC, 0.86) and DSS (AUC, 0.92) based on XGBoost and the prediction of RFS (AUC, 0.83) with logistic regression. The XGBoost classifier achieved the most stable performance with high accuracies compared with other classifiers. The most relevant features for the prediction of RCB class were as follows: changes in lesion size, complete pattern of shrinkage, and mean transit time on DCE-MRI; minimum ADC on DWI; and peritumoral edema on T2-weighted imaging. The most relevant features for prediction of RFS were as follows: volume distribution, mean plasma flow, and mean transit time; DCE-MRI lesion size; minimum, maximum, and mean ADC with DWI. The most relevant features for prediction of DSS were as follows: lesion size, volume distribution, and mean plasma flow on DCE-MRI, and maximum ADC with DWI. CONCLUSIONS Machine learning with mpMRI of the breast enables early prediction of pCR to NAC as well as survival outcomes in breast cancer patients with high accuracy and thus may provide valuable predictive information to guide treatment decisions.
Collapse
|
83
|
Reig B, Heacock L, Geras KJ, Moy L. Machine learning in breast MRI. J Magn Reson Imaging 2019; 52:998-1018. [PMID: 31276247 DOI: 10.1002/jmri.26852] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 12/13/2022] Open
Abstract
Machine-learning techniques have led to remarkable advances in data extraction and analysis of medical imaging. Applications of machine learning to breast MRI continue to expand rapidly as increasingly accurate 3D breast and lesion segmentation allows the combination of radiologist-level interpretation (eg, BI-RADS lexicon), data from advanced multiparametric imaging techniques, and patient-level data such as genetic risk markers. Advances in breast MRI feature extraction have led to rapid dataset analysis, which offers promise in large pooled multiinstitutional data analysis. The object of this review is to provide an overview of machine-learning and deep-learning techniques for breast MRI, including supervised and unsupervised methods, anatomic breast segmentation, and lesion segmentation. Finally, it explores the role of machine learning, current limitations, and future applications to texture analysis, radiomics, and radiogenomics. Level of Evidence: 3 Technical Efficacy Stage: 2 J. Magn. Reson. Imaging 2019. J. Magn. Reson. Imaging 2020;52:998-1018.
Collapse
Affiliation(s)
- Beatriu Reig
- The Department of Radiology, New York University School of Medicine, New York, New York, USA
| | - Laura Heacock
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York, USA
| | - Krzysztof J Geras
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York, USA
| | - Linda Moy
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, New York, USA.,Center for Advanced Imaging Innovation and Research (CAI2 R), New York University School of Medicine, New York, New York, USA
| |
Collapse
|
84
|
Han L, Zhu Y, Liu Z, Yu T, He C, Jiang W, Kan Y, Dong D, Tian J, Luo Y. Radiomic nomogram for prediction of axillary lymph node metastasis in breast cancer. Eur Radiol 2019; 29:3820-3829. [PMID: 30701328 DOI: 10.1007/s00330-018-5981-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/17/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To develop a radiomic nomogram for preoperative prediction of axillary lymph node (LN) metastasis in breast cancer patients. METHODS Preoperative magnetic resonance imaging data from 411 breast cancer patients was studied. Patients were assigned to either a training cohort (n = 279) or a validation cohort (n = 132). Eight hundred eight radiomic features were extracted from the first phase of T1-DCE images. A support vector machine was used to develop a radiomic signature, and logistic regression was used to develop a nomogram. RESULTS The radiomic signature based on 12 LN status-related features was constructed to predict LN metastasis, its prediction ability was moderate, with an area under the curve (AUC) of 0.76 and 0.78 in training and validation cohorts, respectively. Based on a radiomic signature and clinical features, a nomogram was developed and showed excellent predictive ability for LN metastasis (AUC 0.84 and 0.87 in training and validation sets, respectively). Another radiomic signature was constructed to distinguish the number of metastatic LNs (less than 2 positive nodes/more than 2 positive nodes), which also showed moderate performance (AUC 0.79). CONCLUSIONS We developed a nomogram and a radiomic signature that can be used to identify LN metastasis and distinguish the number of metastatic LNs (less than 2 positive nodes/more than 2 positive nodes). Both nomogram and radiomic signature can be used as tools to assist clinicians in assessing LN metastasis in breast cancer patients. KEY POINTS • ALNM is an important factor affecting breast cancer patients' treatment and prognosis. • Traditional imaging examinations have limited value for evaluating axillary LNs status. • We developed a radiomic nomogram based on MR imagings to predict LN metastasis.
Collapse
Affiliation(s)
- Lu Han
- Cancer Hospital of China Medical University, Shenyang, 110042, China
- Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Yongbei Zhu
- CAS Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Institute of Automation, Beijing, 100190, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Institute of Automation, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tao Yu
- Cancer Hospital of China Medical University, Shenyang, 110042, China
- Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Cuiju He
- Cancer Hospital of China Medical University, Shenyang, 110042, China
- Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Wenyan Jiang
- Cancer Hospital of China Medical University, Shenyang, 110042, China
- Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Yangyang Kan
- Cancer Hospital of China Medical University, Shenyang, 110042, China
- Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Institute of Automation, Beijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Institute of Automation, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100191, China
| | - Yahong Luo
- Cancer Hospital of China Medical University, Shenyang, 110042, China.
- Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| |
Collapse
|
85
|
Abstract
PURPOSE OF REVIEW To briefly review the radiomics concept, its applications, and challenges in oncology in the era of precision medicine. RECENT FINDINGS Over the last 5 years, more than 500 studies have evaluated the role of radiomics to predict tumor diagnosis, genetic pattern, tumor response to therapy, and survival in multiple cancers. This new post-processing method is aimed at extracting multiple quantitative features from the image and converting them into mineable data. Radiomics models developed have shown promising results and may play a role in the near future in the daily patient management especially to assess tumor heterogeneity acting as a whole tumor virtual biopsy. For now, radiomics is limited by its lack of standardization; future challenges will be to provide robust and reproducible metrics extracted from large multicenter databases.
Collapse
|
86
|
Liao GJ, Henze Bancroft LC, Strigel RM, Chitalia RD, Kontos D, Moy L, Partridge SC, Rahbar H. Background parenchymal enhancement on breast MRI: A comprehensive review. J Magn Reson Imaging 2019; 51:43-61. [PMID: 31004391 DOI: 10.1002/jmri.26762] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/22/2022] Open
Abstract
The degree of normal fibroglandular tissue that enhances on breast MRI, known as background parenchymal enhancement (BPE), was initially described as an incidental finding that could affect interpretation performance. While BPE is now established to be a physiologic phenomenon that is affected by both endogenous and exogenous hormone levels, evidence supporting the notion that BPE frequently masks breast cancers is limited. However, compelling data have emerged to suggest BPE is an independent marker of breast cancer risk and breast cancer treatment outcomes. Specifically, multiple studies have shown that elevated BPE levels, measured qualitatively or quantitatively, are associated with a greater risk of developing breast cancer. Evidence also suggests that BPE could be a predictor of neoadjuvant breast cancer treatment response and overall breast cancer treatment outcomes. These discoveries come at a time when breast cancer screening and treatment have moved toward an increased emphasis on targeted and individualized approaches, of which the identification of imaging features that can predict cancer diagnosis and treatment response is an increasingly recognized component. Historically, researchers have primarily studied quantitative tumor imaging features in pursuit of clinically useful biomarkers. However, the need to segment less well-defined areas of normal tissue for quantitative BPE measurements presents its own unique challenges. Furthermore, there is no consensus on the optimal timing on dynamic contrast-enhanced MRI for BPE quantitation. This article comprehensively reviews BPE with a particular focus on its potential to increase precision approaches to breast cancer risk assessment, diagnosis, and treatment. It also describes areas of needed future research, such as the applicability of BPE to women at average risk, the biological underpinnings of BPE, and the standardization of BPE characterization. Level of Evidence: 3 Technical Efficacy Stage: 5 J. Magn. Reson. Imaging 2020;51:43-61.
Collapse
Affiliation(s)
- Geraldine J Liao
- Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA.,Department of Radiology, Virginia Mason Medical Center, Seattle, Washington, USA
| | | | - Roberta M Strigel
- Department of Radiology, University of Wisconsin, Madison, Wisconsin, USA.,Department of Medical Physics, University of Wisconsin, Madison, Wisconsin, USA.,Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Rhea D Chitalia
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Despina Kontos
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Linda Moy
- Department of Radiology, New York University School of Medicine, New York, New York, USA
| | - Savannah C Partridge
- Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Habib Rahbar
- Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
87
|
Saeedi E, Dezhkam A, Beigi J, Rastegar S, Yousefi Z, Mehdipour LA, Abdollahi H, Tanha K. Radiomic Feature Robustness and Reproducibility in Quantitative Bone Radiography: A Study on Radiologic Parameter Changes. J Clin Densitom 2019; 22:203-213. [PMID: 30078528 DOI: 10.1016/j.jocd.2018.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/16/2018] [Accepted: 06/20/2018] [Indexed: 10/28/2022]
Abstract
The purpose of this study was to investigate the robustness of different radiography radiomic features over different radiologic parameters including kV, mAs, filtration, tube angles, and source skin distance (SSD). A tibia bone phantom was prepared and all imaging studies was conducted on this phantom. Different radiologic parameters including kV, mAs, filtration, tube angles, and SSD were studied. A region of interest was drawn on the images and many features from different feature sets including histogram, gradient, run-length matrix, co-occurrence matrix, autoregressive model, and wavelet derived parameters were extracted. All radiomic features were categorized based on coefficient of variation (COV). Bland-Altman analysis also was used to evaluate the mean, standard deviation, and upper/lower reproducibility limits for radiomic features in response to variation in each testing parameters. Results on COV in all features showed that 22%, 34%, and 45% of features were most robust (COV ≤ 5%) against kV, mAs, and SSD respectively and there was no robust features against filtration and tube angle. Also, all features (100%) and 76% of which showed large variations (COV > 20%) against filtrations and tube angle respectively. Autoregressive model feature set has no robust features against all radiologic parameters. Features including sum-average, sum-entropy, correlation, mean, and percentile (50, 90, and 99) belong to co-occurrence matrix and histogram feature sets were found as most robust features. Bland-Altman analysis showed the high reproducibity of some feature sets against radiologic parameter changes. The results presented here indicated that radiologic parameters have great impacts on radiomic feature values and caution should be taken into account when work with these features. In quantitative bone studies, robust features with low COV can be selected for clinical or research applications. Reproducible features also can be obtained using Bland-Altman analysis.
Collapse
Affiliation(s)
- Ehsan Saeedi
- Student Research Committee, Paramedical Faculty, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Radiology Technology, Paramedical Faculty, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Dezhkam
- Student Research Committee, Paramedical Faculty, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Radiology Technology, Paramedical Faculty, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Jalal Beigi
- Student Research Committee, Paramedical Faculty, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Radiology Technology, Paramedical Faculty, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Sajjad Rastegar
- Student Research Committee, Paramedical Faculty, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Radiology Technology, Paramedical Faculty, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Yousefi
- Student Research Committee, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran; Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran
| | - Lotf Ali Mehdipour
- Department of Radiology Technology, Paramedical Faculty, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Hamid Abdollahi
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Kiarash Tanha
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
88
|
Liu Z, Li Z, Qu J, Zhang R, Zhou X, Li L, Sun K, Tang Z, Jiang H, Li H, Xiong Q, Ding Y, Zhao X, Wang K, Liu Z, Tian J. Radiomics of Multiparametric MRI for Pretreatment Prediction of Pathologic Complete Response to Neoadjuvant Chemotherapy in Breast Cancer: A Multicenter Study. Clin Cancer Res 2019; 25:3538-3547. [PMID: 30842125 DOI: 10.1158/1078-0432.ccr-18-3190] [Citation(s) in RCA: 300] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/16/2018] [Accepted: 03/05/2019] [Indexed: 01/06/2023]
Abstract
PURPOSE We evaluated the performance of the newly proposed radiomics of multiparametric MRI (RMM), developed and validated based on a multicenter dataset adopting a radiomic strategy, for pretreatment prediction of pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC) in breast cancer. EXPERIMENTAL DESIGN A total of 586 potentially eligible patients were retrospectively enrolled from four hospitals (primary cohort and external validation cohort 1-3). Quantitative imaging features were extracted from T2-weighted imaging, diffusion-weighted imaging, and contrast-enhanced T1-weighted imaging before NAC for each patient. With features selected using a coarse to fine feature selection strategy, four radiomic signatures were constructed based on each of the three MRI sequences and their combination. RMM was developed based on the best radiomic signature incorporating with independent clinicopathologic risk factors. The performance of RMM was assessed with respect to its discrimination and clinical usefulness, and compared with that of clinical information-based prediction model. RESULTS Radiomic signature combining multiparametric MRI achieved an AUC of 0.79 (the highest among the four radiomic signatures). The signature further achieved good performances in hormone receptor-positive and HER2-negative group and triple-negative group. RMM yielded an AUC of 0.86, which was significantly higher than that of clinical model in two of the three external validation cohorts. CONCLUSIONS The study suggested a possibility that RMM provided a potential tool to develop a model for predicting pCR to NAC in breast cancer.
Collapse
Affiliation(s)
- Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhuolin Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, China
| | - Jinrong Qu
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Renzhi Zhang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuezhi Zhou
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China.,Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, China
| | - Longfei Li
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China.,Collaborative Innovation Center for Internet Healthcare, Zhengzhou University, Zhengzhou, Henan, China
| | - Kai Sun
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China.,Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, China
| | - Zhenchao Tang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China
| | - Hui Jiang
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Hailiang Li
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Qianqian Xiong
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yingying Ding
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, China
| | - Xinming Zhao
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, China. .,University of Chinese Academy of Sciences, Beijing, China.,Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China
| |
Collapse
|
89
|
Sadick M, Richers J, Tuschy B, Schad LR, Schoenberg SO, Zöllner FG. Feasibility of quantitative MR-perfusion imaging to monitor treatment response after uterine artery embolization (UAE) in symptomatic uterus fibroids. Magn Reson Imaging 2019; 59:31-38. [PMID: 30807812 DOI: 10.1016/j.mri.2019.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 01/06/2023]
Abstract
INTRODUCTION In 25% of women, symptomatic uterus myomas are diagnosed with clinical and functional impairment ranging from abdominal and pelvic pain to dys- and hypermenorrhea, dyspareunia, pollakiuria and infertility. Women undergoing a treatment increasingly prefer nowadays minimal invasive, uterus preserving therapies like uterine artery embolization (UAE) over surgical hysterectomy, nowadays. To emphasize the efficacy of UAE as a uterus preserving treatment with targeted therapy of myomas only, analysis of tissue perfusion pre and post embolization is required. The purpose of this study was to assess treatment response in UAE in females with symptomatic uterus myomas by quantitative magnetic resonance perfusion imaging. METHODS Seven females scheduled for uterus myoma embolization underwent three MRI examinations (pre, post, follow-up) including morphological and dynamic contrast enhanced perfusion imaging at 3 T. To measure tumor volume, regions-of-interest covering the tumor and the uterus were drawn by two readers in consensus. Blood flow, blood volume, and mean transit time were calculated by a pixel-by-pixel deconvolution approach. Kruskal-Wallis/Friedman test was employed to test whether the group medians differ significantly with correction for multiple comparisons using Bonferroni method. RESULTS Change of volume could be observed in all patients after embolization but was significantly different only between pre/post and follow-up time point. Measured differences in all perfusion parameters were significant between pre-intervention and post-intervention/follow-up in the myomas, no significant differences could be detected for the uterus tissue. CONCLUSIONS Our results demonstrate devascularization of symptomatic myomas which correlates with cessation of hypermenorrhea in all treated patients without affecting healthy uterus tissue. Supplementing UAE with perfusion imaging to monitor early treatment response is feasible and might provide valuable information for the follow-up of patients and contribute to providing confidence for the patients in treatment success.
Collapse
Affiliation(s)
- Maliha Sadick
- Institute of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Jakob Richers
- Institute of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Benjamin Tuschy
- Department of Gynaecology and Obstetrics, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Lothar R Schad
- Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Stefan O Schoenberg
- Institute of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Frank G Zöllner
- Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany.
| |
Collapse
|
90
|
Liu Z, Feng B, Li C, Chen Y, Chen Q, Li X, Guan J, Chen X, Cui E, Li R, Li Z, Long W. Preoperative prediction of lymphovascular invasion in invasive breast cancer with dynamic contrast-enhanced-MRI-based radiomics. J Magn Reson Imaging 2019; 50:847-857. [PMID: 30773770 DOI: 10.1002/jmri.26688] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/02/2019] [Accepted: 02/04/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Lymphovascular invasion (LVI) status facilitates the selection of optimal therapeutic strategy for breast cancer patients, but in clinical practice LVI status is determined in pathological specimens after resection. PURPOSE To explore the use of dynamic contrast-enhanced (DCE)-magnetic resonance imaging (MRI)-based radiomics for preoperative prediction of LVI in invasive breast cancer. STUDY TYPE Prospective. POPULATION Ninety training cohort patients (22 LVI-positive and 68 LVI-negative) and 59 validation cohort patients (22 LVI-positive and 37 LVI-negative) were enrolled. FIELD STRENGTH/SEQUENCE 1.5 T and 3.0 T, T1 -weighted DCE-MRI. ASSESSMENT Axillary lymph node (ALN) status for each patient was evaluated based on MR images (defined as MRI ALN status), and DCE semiquantitative parameters of lesions were calculated. Radiomic features were extracted from the first postcontrast DCE-MRI. A radiomics signature was constructed in the training cohort with 10-fold cross-validation. The independent risk factors for LVI were identified and prediction models for LVI were developed. Their prediction performances and clinical usefulness were evaluated in the validation cohort. STATISTICAL TESTS Mann-Whitney U-test, chi-square test, kappa statistics, least absolute shrinkage and selection operator (LASSO) regression, logistic regression, receiver operating characteristic (ROC) analysis, DeLong test, and decision curve analysis (DCA). RESULTS Two radiomic features were selected to construct the radiomics signature. MRI ALN status (odds ratio, 10.452; P < 0.001) and the radiomics signature (odds ratio, 2.895; P = 0.031) were identified as independent risk factors for LVI. The value of the area under the curve (AUC) for a model combining both (0.763) was higher than that for MRI ALN status alone (0.665; P = 0.029) and similar to that for the radiomics signature (0.752; P = 0.857). DCA showed that the combined model added more net benefit than either feature alone. DATA CONCLUSION The DCE-MRI-based radiomics signature in combination with MRI ALN status was effective in predicting the LVI status of patients with invasive breast cancer before surgery. LEVEL OF EVIDENCE 1 Technical Efficacy Stage: 2 J. Magn. Reson. Imaging 2019;50:847-857.
Collapse
Affiliation(s)
- Zhuangsheng Liu
- Department of Radiology, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, Guangdong, China
| | - Bao Feng
- Department of Radiology, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, Guangdong, China.,School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi, China
| | - Changlin Li
- School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi, China
| | - Yehang Chen
- School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi, China
| | - Qinxian Chen
- Department of Radiology, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, Guangdong, China
| | - Xiaoping Li
- Department of Gastrointestinal Surgery, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, Guangdong, China
| | - Jianhua Guan
- Department of Thyroid and Breast Surgery, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, Guangdong, China
| | - Xiangmeng Chen
- Department of Radiology, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, Guangdong, China
| | - Enming Cui
- Department of Radiology, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, Guangdong, China
| | - Ronggang Li
- Department of Pathology, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, Guangdong, China
| | - Zhi Li
- School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi, China
| | - Wansheng Long
- Department of Radiology, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, Guangdong, China
| |
Collapse
|
91
|
A Review of the Role of Augmented Intelligence in Breast Imaging: From Automated Breast Density Assessment to Risk Stratification. AJR Am J Roentgenol 2019; 212:259-270. [DOI: 10.2214/ajr.18.20391] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
92
|
Radiomic analysis of imaging heterogeneity in tumours and the surrounding parenchyma based on unsupervised decomposition of DCE-MRI for predicting molecular subtypes of breast cancer. Eur Radiol 2019; 29:4456-4467. [PMID: 30617495 DOI: 10.1007/s00330-018-5891-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 10/02/2018] [Accepted: 11/13/2018] [Indexed: 10/27/2022]
Abstract
OBJECTIVES This study aimed to predict the molecular subtypes of breast cancer via intratumoural and peritumoural radiomic analysis with subregion identification based on the decomposition of contrast-enhanced magnetic resonance imaging (DCE-MRI). METHODS The study included 211 women with histopathologically confirmed breast cancer. We utilised a completely unsupervised convex analysis of mixtures (CAM) method by unmixing dynamic imaging series from heterogeneous tissues. Each tumour and the surrounding parenchyma were thus decomposed into multiple subregions, representing different vascular characterisations, from which radiomic features were extracted. A random forest model was trained and tested using a leave-one-out cross-validation (LOOCV) method to predict breast cancer subtypes. The predictive models from tumoural and peritumoural subregions were fused for classification. RESULTS Tumour and peritumour DCE-MR images were decomposed into three compartments, representing plasma input, fast-flow kinetics, and slow-flow kinetics. The tumour subregion related to fast-flow kinetics showed the best performance among the subregions for differentiating between patients with four molecular subtypes (area under the receiver operating characteristic curve (AUC) = 0.832), exhibiting an AUC value significantly (p < 0.0001) higher than that obtained with the entire tumour (AUC = 0.719). When the tumour- and parenchyma-based predictive models were fused, the performance, measured as the AUC, increased to 0.897; this value was significantly higher than that obtained with other tumour partition methods. CONCLUSIONS Radiomic analysis of intratumoural and peritumoural heterogeneity based on the decomposition of image time-series signals has the potential to more accurately identify tumour kinetic features and serve as a valuable clinical marker to enhance the prediction of breast cancer subtypes. KEY POINTS • Decomposition of image time-series signals has the potential to more accurately identify tumour kinetic features. • Fusion of intratumoural- and peritumoural-based predictive models improves the prediction of breast cancer subtypes.
Collapse
|
93
|
Artificial Intelligence for Breast MRI in 2008-2018: A Systematic Mapping Review. AJR Am J Roentgenol 2019; 212:280-292. [PMID: 30601029 DOI: 10.2214/ajr.18.20389] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The purpose of this study is to review literature from the past decade on applications of artificial intelligence (AI) to breast MRI. MATERIALS AND METHODS In June 2018, a systematic search of the literature was performed to identify articles on the use of AI in breast MRI. For each article identified, the surname of the first author, year of publication, journal of publication, Web of Science Core Collection journal category, country of affiliation of the first author, study design, dataset, study aim(s), AI methods used, and, when available, diagnostic performance were recorded. RESULTS Sixty-seven studies, 58 (87%) of which had a retrospective design, were analyzed. When journal categories were considered, 36% of articles were identified as being included in the radiology and imaging journal category. Contrast-enhanced sequences were used for most AI applications (n = 50; 75%) and, on occasion, were combined with other MRI sequences (n = 8; 12%). Four main clinical aims were addressed: breast lesion classification (n = 36; 54%), image processing (n = 14; 21%), prognostic imaging (n = 9; 13%), and response to neoadjuvant therapy (n = 8; 12%). Artificial neural networks, support vector machines, and clustering were the most frequently used algorithms, accounting for 66%. The performance achieved and the most frequently used techniques were then analyzed according to specific clinical aims. Supervised learning algorithms were primarily used for lesion characterization, with the AUC value from ROC analysis ranging from 0.74 to 0.98 (median, 0.87) and with that from prognostic imaging ranging from 0.62 to 0.88 (median, 0.80), whereas unsupervised learning was mainly used for image processing purposes. CONCLUSION Interest in the application of advanced AI methods to breast MRI is growing worldwide. Although this growth is encouraging, the current performance of AI applications in breast MRI means that such applications are still far from being incorporated into clinical practice.
Collapse
|
94
|
Computerized Image Analysis to Differentiate Benign and Malignant Breast Tumors on Magnetic Resonance Diffusion Weighted Image. J Comput Assist Tomogr 2019; 43:93-97. [DOI: 10.1097/rct.0000000000000793] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
95
|
Luo J, Ning Z, Zhang S, Feng Q, Zhang Y. Bag of deep features for preoperative prediction of sentinel lymph node metastasis in breast cancer. ACTA ACUST UNITED AC 2018; 63:245014. [DOI: 10.1088/1361-6560/aaf241] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
96
|
Cain EH, Saha A, Harowicz MR, Marks JR, Marcom PK, Mazurowski MA. Multivariate machine learning models for prediction of pathologic response to neoadjuvant therapy in breast cancer using MRI features: a study using an independent validation set. Breast Cancer Res Treat 2018; 173:455-463. [PMID: 30328048 DOI: 10.1007/s10549-018-4990-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 10/01/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE To determine whether a multivariate machine learning-based model using computer-extracted features of pre-treatment dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) can predict pathologic complete response (pCR) to neoadjuvant therapy (NAT) in breast cancer patients. METHODS Institutional review board approval was obtained for this retrospective study of 288 breast cancer patients at our institution who received NAT and had a pre-treatment breast MRI. A comprehensive set of 529 radiomic features was extracted from each patient's pre-treatment MRI. The patients were divided into equal groups to form a training set and an independent test set. Two multivariate machine learning models (logistic regression and a support vector machine) based on imaging features were trained to predict pCR in (a) all patients with NAT, (b) patients with neoadjuvant chemotherapy (NACT), and (c) triple-negative or human epidermal growth factor receptor 2-positive (TN/HER2+) patients who had NAT. The multivariate models were tested using the independent test set, and the area under the receiver operating characteristics (ROC) curve (AUC) was calculated. RESULTS Out of the 288 patients, 64 achieved pCR. The AUC values for predicting pCR in TN/HER+ patients who received NAT were significant (0.707, 95% CI 0.582-0.833, p < 0.002). CONCLUSIONS The multivariate models based on pre-treatment MRI features were able to predict pCR in TN/HER2+ patients.
Collapse
Affiliation(s)
- Elizabeth Hope Cain
- Department of Radiology, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27705, USA.
| | - Ashirbani Saha
- Department of Radiology, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27705, USA
| | - Michael R Harowicz
- Department of Radiology, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27705, USA.,Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey R Marks
- Department of Surgery, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27705, USA
| | - P Kelly Marcom
- Department of Medicine, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27705, USA
| | - Maciej A Mazurowski
- Department of Radiology, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27705, USA.,Department of Electrical and Computer Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
97
|
Tan W, Yang M, Yang H, Zhou F, Shen W. Predicting the response to neoadjuvant therapy for early-stage breast cancer: tumor-, blood-, and imaging-related biomarkers. Cancer Manag Res 2018; 10:4333-4347. [PMID: 30349367 PMCID: PMC6188192 DOI: 10.2147/cmar.s174435] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neoadjuvant therapy (NAT) has been used increasingly in patients with locally advanced or early-stage breast cancer. However, the accurate evaluation and prediction of response to NAT remain the great challenge. Biomarkers could prove useful to identify responders or nonresponders, or even to distinguish between early and delayed responses. These biomarkers could include markers from the tumor itself, such as versatile proteins, genes, and ribonucleic acids, various biological factors or peripheral blood cells, and clinical and pathological features. Possible predictive markers could also include multiple features from functional imaging, such as standard uptake values in positron emission tomography, apparent diffusion coefficient in magnetic resonance, or radiomics imaging biomarkers. In addition, cells that indirectly present the immune status of tumor cells and/or their host could also potentially be used as biomarkers, eg, tumor-infiltrating lymphocytes, tumor-associated macrophages, and myeloid-derived suppressor cells. Though numerous biomarkers have been widely investigated, only estrogen and/or progesterone receptors and human epidermal growth factor receptor have been proven to be reliable biomarkers to predict the response to NAT. They are the only biomarkers recommended in several international guidelines. The other aforementioned biomarkers warrant further validation studies. Some multigene profiling assays that are commercially available, eg, Oncotype DX and MammaPrint, should be used with caution when extrapolated to NAT settings. A panel of combined multilevel biomarkers might be able to predict the response to NAT more robustly than individual biomarkers. To establish such a panel and its prediction model, reliable methods and extensive clinical validation are warranted.
Collapse
Affiliation(s)
- Wenyong Tan
- Department of Oncology, Shenzhen Hospital of Southern Medical University, Shenzhen, People's Republic of China, ;
- Clinical Medical Research Center, The Second Clinical Medical College (Shenzhen People Hospital), Jinan University, Shenzhen, People's Republic of China,
| | - Ming Yang
- Shenzhen Jingmai Medical Scientific and Technique Company, Shenzhen, People's Republic of China
| | - Hongli Yang
- Clinical Medical Research Center, The Second Clinical Medical College (Shenzhen People Hospital), Jinan University, Shenzhen, People's Republic of China,
| | - Fangbin Zhou
- Clinical Medical Research Center, The Second Clinical Medical College (Shenzhen People Hospital), Jinan University, Shenzhen, People's Republic of China,
| | - Weixi Shen
- Department of Oncology, Shenzhen Hospital of Southern Medical University, Shenzhen, People's Republic of China, ;
| |
Collapse
|
98
|
Application of Radiomics and Decision Support Systems for Breast MR Differential Diagnosis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2018; 2018:7417126. [PMID: 30344618 PMCID: PMC6174735 DOI: 10.1155/2018/7417126] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/24/2018] [Accepted: 09/04/2018] [Indexed: 01/17/2023]
Abstract
Over the years, MR systems have evolved from imaging modalities to advanced computational systems producing a variety of numerical parameters that can be used for the noninvasive preoperative assessment of breast pathology. Furthermore, the combination with state-of-the-art image analysis methods provides a plethora of quantifiable imaging features, termed radiomics that increases diagnostic accuracy towards individualized therapy planning. More importantly, radiomics can now be complemented by the emerging deep learning techniques for further process automation and correlation with other clinical data which facilitate the monitoring of treatment response, as well as the prediction of patient's outcome, by means of unravelling of the complex underlying pathophysiological mechanisms which are reflected in tissue phenotype. The scope of this review is to provide applications and limitations of radiomics towards the development of clinical decision support systems for breast cancer diagnosis and prognosis.
Collapse
|
99
|
Palmisano A, Esposito A, Rancoita PMV, Di Chiara A, Passoni P, Slim N, Campolongo M, Albarello L, Fiorino C, Rosati R, Del Maschio A, De Cobelli F. Could perfusion heterogeneity at dynamic contrast-enhanced MRI be used to predict rectal cancer sensitivity to chemoradiotherapy? Clin Radiol 2018; 73:911.e1-911.e7. [PMID: 30029837 DOI: 10.1016/j.crad.2018.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 06/04/2018] [Indexed: 12/16/2022]
Abstract
AIM To evaluate whether perfusion heterogeneity of rectal cancer prior to chemoradiotherapy (CRT) using histogram analysis of dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) quantitative parameters can predict response to treatment. MATERIALS AND METHODS Twenty-one patients with histologically proven rectal adenocarcinoma were enrolled prospectively. All patients underwent 1.5 T DCE-MRI before CRT. Tumour volumes were drawn on Ktrans and Ve maps, using T2-weighted (W) images as reference, and the following first-order texture parameters of Ve and Ktrans values were extracted: 25th, 50th, 75th percentile, mean, standard deviation, skewness, and kurtosis. After CRT, patients underwent surgery and according with Rödel's tumour regression grade (TRG), they were classified as poor responders "non-GR" (TRG 0-2) and good responders "GR" (TRG 3-4). Differences between GR and non-GR in DCE-MRI first-order texture parameters were evaluated using the Mann-Whitney test, and their role in the prediction of response was investigated using receiver operating characteristic (ROC) curve analysis. RESULTS Sixteen (76%) patients were classified as GR and five (24%) were non-GR. Skewness and kurtosis of Ve was significantly higher in non-GR (4.886±1.320 and 36.402±24.486, respectively) than in GR patients (1.809±1.280, p=0.003 and 6.268±8.130, p= 0.011). Ve skewness <3.635 was able to predict GR with an area under the ROC curve (AUC) of 0.988, sensitivity 93.8%, specificity 80%, and accuracy 90.5%. Ve kurtosis <21.095 was able to predict response with an AUC of 0.963, sensitivity 93.8%, specificity 80%, and accuracy 90.5%. Other parameters were not different between groups or predictors of response. CONCLUSION Ve skewness and kurtosis seem to be promising in the prediction of response to CRT in rectal cancer patients.
Collapse
Affiliation(s)
- A Palmisano
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - A Esposito
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - P M V Rancoita
- University Centre of Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan, Italy
| | - A Di Chiara
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - P Passoni
- Unit of Radiotherapy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - N Slim
- Unit of Radiotherapy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - M Campolongo
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | - L Albarello
- Department of Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - C Fiorino
- Medical Physics, San Raffaele Hospital, Milan, Italy
| | - R Rosati
- Vita-Salute San Raffaele University, Milan, Italy; Department of Gastrointestinal Surgery, San Raffaele Hospital, Milan, Italy
| | - A Del Maschio
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - F De Cobelli
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
100
|
Valdora F, Houssami N, Rossi F, Calabrese M, Tagliafico AS. Rapid review: radiomics and breast cancer. Breast Cancer Res Treat 2018; 169:217-229. [PMID: 29396665 DOI: 10.1007/s10549-018-4675-4] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/16/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE To perform a rapid review of the recent literature on radiomics and breast cancer (BC). METHODS A rapid review, a streamlined approach to systematically identify and summarize emerging studies was done (updated 27 September 2017). Clinical studies eligible for inclusion were those that evaluated BC using a radiomics approach and provided data on BC diagnosis (detection or characterization) or BC prognosis (response to therapy, morbidity, mortality), or provided data on technical challenges (software application: open source, repeatability of results). Descriptive statistics, results, and radiomics quality score (RQS) are presented. RESULTS N = 17 retrospective studies, all published after 2015, provided BC-related radiomics data on 3928 patients evaluated with a radiomics approach. Most studies were done for diagnosis and/or characterization (65%, 11/17) or to aid in prognosis (41%, 7/17). The mean number of radiomics features considered was 100. Mean RQS score was 11.88 ± 5.8 (maximum value 36). The RQS criteria related to validation, gold standard, potential clinical utility, cost analysis, and open science data had the lowest scores. The majority of studies n = 16/17 (94%) provided correlation with histological outcomes and staging variables or biomarkers. Only 4/17 (23%) studies provided evidence of correlation with genomic data. Magnetic resonance imaging (MRI) was used in most studies n = 14/17 (82%); however, ultrasound (US), mammography, or positron emission tomography with 2-deoxy-2-[fluorine-18]fluoro-D-glucose integrated with computed tomography (18F FDG PET/CT) was also used. Much heterogeneity was found for software usage. CONCLUSIONS The study of radiomics in BC patients is a new and emerging translational research topic. Radiomics in BC is frequently done to potentially improve diagnosis and characterization, mostly using MRI. Substantial quality limitations were found; high-quality prospective and reproducible studies are needed to further potential application.
Collapse
Affiliation(s)
- Francesca Valdora
- Department of Health Sciences, University of Genova, Via L.B. Alberti 2, 16132, Genoa, Italy
| | - Nehmat Houssami
- Sydney School of Public Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Federica Rossi
- Department of Health Sciences, University of Genova, Via L.B. Alberti 2, 16132, Genoa, Italy
| | | | - Alberto Stefano Tagliafico
- Department of Health Sciences, University of Genova, Via L.B. Alberti 2, 16132, Genoa, Italy.
- Ospedale Policlinico San Martino IST, Genoa, Italy.
| |
Collapse
|