Figure - available from: BMC Medical Imaging
This content is subject to copyright. Terms and conditions apply.
Mammogram and histogram analyses of a patient (female, 48 years old) with TNBC in the lateral quadrant of the left breast. A A mammography image shows an irregular nodule in the lateral quadrant of the left breast, with a length of about 7.9 cm and shallow lobes visible on the edge. B The MaZda image segmentation tool was applied to manually delineate the area of interest in the mammography and extract the radiomics features. C The gray level histogram shows the ROI in the lateral quadrant of the left breast

Mammogram and histogram analyses of a patient (female, 48 years old) with TNBC in the lateral quadrant of the left breast. A A mammography image shows an irregular nodule in the lateral quadrant of the left breast, with a length of about 7.9 cm and shallow lobes visible on the edge. B The MaZda image segmentation tool was applied to manually delineate the area of interest in the mammography and extract the radiomics features. C The gray level histogram shows the ROI in the lateral quadrant of the left breast

Source publication
Article
Full-text available
Objective: This study is aimed to explore the value of mammography-based radiomics signature for preoperative prediction of triple-negative breast cancer (TNBC). Materials and methods: Initially, the clinical and X-ray data of patients (n = 319, age of 54 ± 14) with breast cancer (triple-negative-65, non-triple-negative-254) from the First Affil...

Similar publications

Article
Full-text available
Radiomics has been a promising imaging biomarker for many malignant diseases. We developed a novel radiomics strategy that incorporating radiomics features extracted from dual-view mammograms and clinical parameters for identifying benign and malignant breast lesions, and validated whether the radiomics assessment could improve the accurate diagnos...

Citations

... Recently, the increase in computational power and the availability of bigger imaging datasets have enabled the development of successful deep learning artificial intelligence (AI) algorithms in medical imaging. In fact, there are some recently published works addressing the use of AI algorithms to predict the breast cancer molecular subtype in mammography images [12][13][14]. Wang et al. [12] aimed to differentiate TN from non-TN tumors using radiomics. Using a dataset of 51 cases (23 TN, 31 non-TN), the authors extracted 396 features of the segmented tumors and employed the maximum relevance minimum redundancy (mRMR) algorithm and the least absolute shrinkage and selection operator (LASSO) method to eliminate redundant and irrelevant features. ...
... The same goal was addressed by Ge et al. [13]. In this work, the "verification" set used by the authors has a larger number of subjects, with 108 cases. ...
Article
Full-text available
Breast cancer remains a leading cause of mortality among women, with molecular subtypes significantly influencing prognosis and treatment strategies. Currently, identifying the molecular subtype of cancer requires a biopsy—a specialized, expensive, and time-consuming procedure, often yielding to results that must be supported with additional biopsies due to technique errors or tumor heterogeneity. This study introduces a novel approach for predicting breast cancer molecular subtypes using mammography images and advanced artificial intelligence (AI) methodologies. Using the OPTIMAM imaging database, 1397 images from 660 patients were selected. The pretrained deep learning model ResNet-101 was employed to classify tumors into five subtypes: Luminal A, Luminal B1, Luminal B2, HER2, and Triple Negative. Various classification strategies were studied: binary classifications (one vs. all others, specific combinations) and multi-class classification (evaluating all subtypes simultaneously). To address imbalanced data, strategies like oversampling, undersampling, and data augmentation were explored. Performance was evaluated using accuracy and area under the receiver operating characteristic curve (AUC). Binary classification results showed a maximum average accuracy and AUC of 79.02% and 64.69%, respectively, while multi-class classification achieved an average AUC of 60.62% with oversampling and data augmentation. The most notable binary classification was HER2 vs. non-HER2, with an accuracy of 89.79% and an AUC of 73.31%. Binary classification for specific combinations of subtypes revealed an accuracy of 76.42% for HER2 vs. Luminal A and an AUC of 73.04% for HER2 vs. Luminal B1. These findings highlight the potential of mammography-based AI for non-invasive breast cancer subtype prediction, offering a promising alternative to biopsies and paving the way for personalized treatment plans.
... The application of deep learning techniques has demonstrated remarkable efficacy in accurately segmenting TNBC tumors 86 and distinguishing them from non-TNBC cases, marking a significant milestone in diagnosis. 87 However, the true power of machine learning lies in its potential for prediction and prognostication, going beyond tumor classification. By leveraging vast amounts of data and complex algorithms, machine learning can provide insights into the future behavior and outcomes of TNBC patients. ...
Article
Full-text available
Breast cancer is one of the most common and deadly cancers worldwide. Approximately, 20% of all breast cancers are characterized as triple negative (TNBC). TNBC typically is associated with a poorer prognosis relative to other breast cancer subtypes. Due to its aggressiveness and lack of response to hormonal therapy, conventional cytotoxic chemotherapy is the usual treatment; however, this treatment is not always effective, and an important percentage of patients develop recurrence. More recently, immunotherapy has started to be used on some populations with TNBC showing promising results. Unfortunately, immunotherapy is only applicable to a minority of patients and responses in metastatic TNBC have overall been modest in comparison to other cancer types. This situation evidences the need for developing effective biomarkers that help to stratify and personalize patient management. Thanks to recent advances in artificial intelligence (AI), there has been an increasing interest in its use for medical applications aiming at supporting clinical decision making. Several works have used AI in combination with diagnostic medical imaging, more specifically radiology and digitized histopathological tissue samples, aiming to extract disease-specific information that is difficult to quantify by the human eye. These works have demonstrated that analysis of such images in the context of TNBC has great potential for (1) risk-stratifying patients to identify those patients who are more likely to experience disease recurrence or die from the disease and (2) predicting pathologic complete response. In this manuscript, we present an overview on AI and its integration with radiology and histopathological images for developing prognostic and predictive approaches for TNBC. We present state of the art approaches in the literature and discuss the opportunities and challenges with developing AI algorithms regarding further development and clinical deployment, including identifying those patients who may benefit from certain treatments (e.g., adjuvant chemotherapy) from those who may not and thereby should be directed toward other therapies, discovering potential differences between populations, and identifying disease subtypes.
... AI has been applied to imaging analysis of lung, prostate, and breast cancer; however, the task of diagnosing pancreatic cancer remains particularly challenging [78,[85][86][87][88]. The pancreas is highly variable in size, shape, and location. ...
Article
Full-text available
Simple Summary Pancreatic cancer will soon become the second leading cause of cancer-related death mainly due to a lack of early diagnosis. Artificial intelligence is being applied in various aspects of diagnosing medical conditions. In this review, we summarize the current literature on the application of artificial intelligence in the diagnosis and management of premalignant lesions that would otherwise progress to pancreatic cancer. Abstract Pancreatic cancer is projected to become the second leading cause of cancer-related mortality in the United States by 2030. This is in part due to the paucity of reliable screening and diagnostic options for early detection. Amongst known pre-malignant pancreatic lesions, pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasms (IPMNs) are the most prevalent. The current standard of care for the diagnosis and classification of pancreatic cystic lesions (PCLs) involves cross-sectional imaging studies and endoscopic ultrasound (EUS) and, when indicated, EUS-guided fine needle aspiration and cyst fluid analysis. However, this is suboptimal for the identification and risk stratification of PCLs, with accuracy of only 65–75% for detecting mucinous PCLs. Artificial intelligence (AI) is a promising tool that has been applied to improve accuracy in screening for solid tumors, including breast, lung, cervical, and colon cancer. More recently, it has shown promise in diagnosing pancreatic cancer by identifying high-risk populations, risk-stratifying premalignant lesions, and predicting the progression of IPMNs to adenocarcinoma. This review summarizes the available literature on artificial intelligence in the screening and prognostication of precancerous lesions in the pancreas, and streamlining the diagnosis of pancreatic cancer.
... It is effective in its use for various imaging analyses, including X-rays, US, and magnetic resonance imaging. It has been proven to be an efficient and reliable tool for quantitative image analyses, providing more accurate and objective medical diagnoses (12)(13)(14)(15). A logistic regression model is based on a multivariate regression analysis, integrating multiple predictors and using multiple indicators to diagnose or predict the occurrence or progress of diseases (16, 17). ...
Article
Full-text available
Objectives We aimed to develop an ultrasound-based radiomics model to distinguish between sclerosing adenosis (SA) and invasive ductal carcinoma (IDC) to avoid misdiagnosis and unnecessary biopsies.Methods From January 2020 to March 2022, 345 cases of SA or IDC that were pathologically confirmed were included in the study. All participants underwent pre-surgical ultrasound (US), from which clinical information and ultrasound images were collected. The patients from the study population were randomly divided into a training cohort (n = 208) and a validation cohort (n = 137). The US images were imported into MaZda software (Version 4.2.6.0) to delineate the region of interest (ROI) and extract features. Intragroup correlation coefficient (ICC) was used to evaluate the consistency of the extracted features. The least absolute shrinkage and selection operator (LASSO) logistic regression and cross-validation were performed to obtain the radiomics score of the features. Based on univariate and multivariate logistic regression analyses, a model was developed. 56 cases from April 2022 to December 2022 were included for independent validation of the model. The diagnostic performance of the model and the radiomics scores were evaluated by performing the receiver operating characteristic (ROC) analysis. The calibration curve and decision curve analysis (DCA) were used for calibration and evaluation. Leave-One-Out Cross-Validation (LOOCV) was used for the stability of the model.ResultsThree predictors were selected to develop the model, including radiomics score, palpable mass and BI-RADS. In the training cohort, validation cohort and independent validation cohort, AUC of the model and radiomics score were 0.978 and 0.907, 0.946 and 0.886, 0.951 and 0.779, respectively. The model showed a statistically significant difference compared with the radiomics score (p<0.05). The Kappa value of the model was 0.79 based on LOOCV. The Brier score, calibration curve, and DCA showed the model had a good calibration and clinical usefulness.Conclusions The model based on radiomics, ultrasonic features, and clinical manifestations can be used to distinguish SA from IDC, which showed good stability and diagnostic performance. The model can be considered a potential candidate diagnostic tool for breast lesions and can contribute to effective clinical diagnosis.
Article
This study aimed to develop a radiomics nomogram based on grayscale ultrasound (US) to distinguish triple-negative breast cancer (TNBC) from non-triple-negative breast cancer (NTNBC) prior to surgery. A retrospective analysis of 454 breast carcinoma patients confirmed by pathology was conducted, with 317 patients in the training dataset (59 with TNBC) and 137 patients in the validation dataset (27 with TNBC). Clinical information, conventional US features, and radiomics features were collected, and the Radscore model was constructed after feature selection. Independent risk factors were identified using univariate and multivariate logistic regression analysis. The nomogram model was assessed using the receiver operating characteristic (ROC) curve analysis, calibration curve, decision curve analysis (DCA), net reclassification improvement (NRI) and integrated discrimination improvement (IDI). Tumor shape, margin, and calcification were independent risk factors in the clinical prediction model. Additionally, 16 radiomics features were selected to construct the Radscore model out of a total of 474 extracted features. The radiomics nomogram model, which incorporated tumor shape, margin, calcification, and Radscore, achieved an AUC value of 0.837 in the training dataset and 0.813 in the validation dataset, outperforming both the Radscore and clinical models in terms of predictive performance. The significant improvement of NRI and IDI indicated that the Radscore may be useful biomarkers for TNBC. The US-based radiomics nomogram showed satisfactory preoperative prediction of TNBC.