Next Article in Journal
Prevalence of Myocardial Injury and Myocardial Infarction in Patients with a Hypertensive Emergency: A Systematic Review
Next Article in Special Issue
Pregnancy-Associated Breast Cancer: A Diagnostic and Therapeutic Challenge
Previous Article in Journal
Clinical Outcomes of Cataract Surgery in Patients with Sjögren’s Syndrome
Previous Article in Special Issue
Precision Medicine in Breast Cancer: Do MRI Biomarkers Identify Patients Who Truly Benefit from the Oncotype DX Recurrence Score® Test?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Artificial Intelligence in Breast Ultrasound: From Diagnosis to Prognosis—A Rapid Review

by
Nicole Brunetti
1,*,
Massimo Calabrese
2,
Carlo Martinoli
2,3 and
Alberto Stefano Tagliafico
2,3
1
Department of Experimental Medicine (DIMES), University of Genova, Via L.B. Alberti 2, 16132 Genoa, Italy
2
Department of Radiology, IRCCS—Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
3
Department of Health Sciences (DISSAL), University of Genova, Via L.B. Alberti 2, 16132 Genoa, Italy
*
Author to whom correspondence should be addressed.
Diagnostics 2023, 13(1), 58; https://doi.org/10.3390/diagnostics13010058
Submission received: 3 December 2022 / Revised: 19 December 2022 / Accepted: 20 December 2022 / Published: 26 December 2022
(This article belongs to the Special Issue Advances in Breast Cancer Imaging and Treatment)

Abstract

:
Background: Ultrasound (US) is a fundamental diagnostic tool in breast imaging. However, US remains an operator-dependent examination. Research into and the application of artificial intelligence (AI) in breast US are increasing. The aim of this rapid review was to assess the current development of US-based artificial intelligence in the field of breast cancer. Methods: Two investigators with experience in medical research performed literature searching and data extraction on PubMed. The studies included in this rapid review evaluated the role of artificial intelligence concerning BC diagnosis, prognosis, molecular subtypes of breast cancer, axillary lymph node status, and the response to neoadjuvant chemotherapy. The mean values of sensitivity, specificity, and AUC were calculated for the main study categories with a meta-analytical approach. Results: A total of 58 main studies, all published after 2017, were included. Only 9/58 studies were prospective (15.5%); 13/58 studies (22.4%) used an ML approach. The vast majority (77.6%) used DL systems. Most studies were conducted for the diagnosis or classification of BC (55.1%). At present, all the included studies showed that AI has excellent performance in breast cancer diagnosis, prognosis, and treatment strategy. Conclusions: US-based AI has great potential and research value in the field of breast cancer diagnosis, treatment, and prognosis. More prospective and multicenter studies are needed to assess the potential impact of AI in breast ultrasound.

1. Introduction

Breast cancer (BC) is the most common malignancy in women and the second leading cause of cancer death, so the early diagnosis of BC remains crucial [1]. Mammography is the first-line examination for breast cancer screening. In several settings, another fundamental tool is represented by breast ultrasound (US). In particular, US is recommended as a first-line examination in young women, during pregnancy or breastfeeding, and as an additional examination in women with dense breasts after mammography [2]. US presents several advantages, including safety, low cost, rapid execution, and cost-effectiveness. However, US remains an operator-dependent examination. For these reasons, in recent years, there has been a strong development of artificial intelligence (AI) in breast ultrasound. AI systems involve three working steps: image processing, segmentation, and feature extraction. AI is used to provide predictive models based on the analysis of the features extracted from radiological data. The first step is represented by lesion detection and segmentation (unsupervised or supervised). Then, radiomic analysis is performed with biomarker extraction and analysis used to obtain information for diagnosis or prognosis, as will be explained later. AI systems process a large amount of iconographic data from different imaging modalities to obtain output information [3]. In this framework, we can divide these into two main broad approaches:
Machine learning (ML)—a complex multistep process that uses texture analysis to extract quantitative information from radiological images to create prediction models and decision support tools.
Deep learning (DL)—represents an evolution of ML. The system is able to extrapolate inputs directly from images. These inputs come directly to a multilayer neural network. It does not require manual design features required by traditional methods but automatically learns features [4].
Currently, radiomics is a complex process that involves several steps, and generally, its application is not fully automated. Unsupervised machine learning does not need the training phase to work, but it is typically used just for classification purposes. Supervised machine learning has more general applications, for example, for regression and prediction. Deep learning is an extreme modification of machine learning. AI looks promising in diagnosing breast lesions, predicting molecular subtypes of breast cancer, evaluating axillary lymph node status, and evaluating the response to neoadjuvant chemotherapy, as demonstrated by several studies in the literature. The aim of this rapid review was to assess the current development and research status of US-based AI in the field of breast cancer.

2. Materials and Methods

2.1. Identification of Studies

Two investigators with experience in medical research performed literature searching and data extraction on PubMed. The two researchers have had extensive experience in medical research and systematic reviews (N.B. and A.T.), with specific expertise in breast cancer imaging, ultrasound imaging, and radiomics. Literature search on PubMed was conducted with the following search strategy:
(((“diagnostic imaging”(MeSH Subheading) OR (“diagnostic”(All Fields) AND “imaging”(All Fields)) OR “diagnostic imaging”(All Fields) OR “ultrasound”(All Fields) OR “ultrasonography”(MeSH Terms) OR “ultrasonography”(All Fields) OR “ultrasonics”(MeSH Terms) OR “ultrasonics”(All Fields) OR “ultrasounds”(All Fields) OR “ultrasound s”(All Fields)) AND (“breast”(MeSH Terms) OR “breast”(All Fields) OR “breasts”(All Fields) OR “breast s”(All Fields)) AND (“artificial intelligence”(MeSH Terms) OR (“artificial”(All Fields) AND “intelligence”(All Fields)) OR “artificial intelligence”(All Fields)))) AND (2017:2023(pdat)).
Only publications after 2017 and in the English language were considered. Reports or case series, review articles, letters, comments, or studies with incomplete data were excluded. The studies included in this rapid review evaluated the role of artificial intelligence in breast ultrasound providing data on BC diagnosis, prognosis, or BC staging. In Figure 1, a schematic representation of a typical workflow of data extraction from US images is presented. After the segmentation is completed, a radiomic analysis is carried out, extracting quantitative features from the obtained volumes. The extracted data are processed by software for possible clinical use, such as diagnosis, reduction in biopsies number, or as prognostic tools. We only used data available in the published studies without contacting authors.

2.2. Studies Examination and Data Extraction

Two authors independently extracted the data from the eligible study. Fifty-eight main studies were identified, as reported in Figure 2. Discrepancies were resolved by discussion between two authors. From each study, we extracted the following data: first author, publication year, design of the study (retrospective or prospective, single center or multicenter), study population, aim of the study, imaging modality, AI modality, test data set, and training-validation dataset. Papers without solid statistical analysis were excluded from the study. Then, we performed a descriptive analysis of these studies.

3. Results

A total of 58 main studies, all published after 2017, were included. Only 9/58 studies were prospective (15.5%), and 13/58 studies (22.4%) used an ML approach. The vast majority (77.6%) used DL systems. Studies performed with the aim of evaluating the role of AI in the diagnosis of BC was 32/58 (55.1%), assessing lymph node status was 8/58 (13.8%), predicting the response to neoadjuvant chemotherapy (NAC) was 6/58 (10.3%), and predicting molecular subtypes of BC was 8/58 (13.8%). Only four studies investigated the role of AI in the upstage of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) and in the prediction of the BC prognosis. Only 8/58 studies (13.7%) also used shear wave imaging (SWE)/quantitative ultrasound (QUS) and color doppler flow imaging (CDFI) in addition to B-mode US.
Table 1, Table 2, Table 3 and Table 4 show all the included studies, their aims, and the performance of the AI algorithms.
The mean values of sensitivity (true positive rate), specificity (true negative rate) and AUC (area under the curve) were calculated for the main study categories with a meta-analytical approach.
For the BC diagnosis:
  • the average sensitivity value was 84.00 (95% CI 73.85–94.15);
  • the average specificity value was 85.67 (95% CI 73.41–97.92);
  • the average AUC value was 90.6429 (95% CI 88.4898–92.7959).
For the assessment of lymph node status:
  • the average sensitivity value was 66.475 (95% CI 31.3442–101.6058);
  • the average specificity value was 66.455 (95% CI 31.6272–101.2828);
  • the average AUC value was 84.5714 (95% CI 76.8547–92.2882).
For prediction of the response to neoadjuvant chemotherapy:
  • the average sensitivity value was 85.3333 (95% CI 76.0254–94.6412);
  • the average specificity value was 78.1667 (95% CI 61.6080–94.7254);
  • the average AUC value was 85.8333 (95% CI 77.5059–94.1608).
For prediction of the molecular subtypes of BC:
  • the average sensitivity value was 88.00 (95% CI 79.1947–96.8053);
  • the average specificity value was 82.00 (95% CI 69.8337–94.8329);
  • the average AUC value was 86.875 (95% CI 81.4489–92.3011).

4. Discussion

4.1. Diagnosis of Breast Cancer

US represents a fundamental tool to evaluate and characterize breast masses after mammography. Interpreting breast ultrasound is challenging. According to BI-RADS [59], many features have to be analyzed to evaluate a breast nodule, including size, shape, margin, echogenicity, posterior acoustic features, and orientation. Diagnosis depends on the experience of radiologists, and thus, great interobserver variability may happen. The malignancy risk of BI-RADS-4 lesions covers a range from 2% to 95%. After breast US, 5–15% of patients are recalled, and 4–8% of these undergo biopsy with the problem of false positive examinations [2]. One of the advantages of the AI system is the possibility of detecting ultrasound features that cannot be distinguished by the human eye. Especially in BI-RADS 4 breast lesions, AI could help to reduce the rate of unnecessary biopsies. When AI is present in the clinical workflow of radiologists, it can be used as a “second opinion” in the assessment of suspicious breast nodules [60]. Several studies have assessed the value of an AI system in the decision-making process (downgrading and upgrading). According to Lyu et al. [8], an adjusted BI-RADS classification after using AI could lead to a decreased biopsy rate (from 100% to 67.29% in this study), with a great reduction in unnecessary biopsies. As also demonstrated by Wang et al. [11], after hypothetically downgrading with AI, 14 samples would have been avoided without cancer missed. Shen et al.’s paper [12], based on a large amount of data, showed how, helped by artificial intelligence, radiologists could decrease false positive rates by 37.3% and reduce requested biopsies by 27.8%, maintaining the same sensitivity. Gu et al. [27], in an important multicenter study with a large training cohort, internal test cohort, and external test cohorts, demonstrated that a DL system presented a similar performance with respect to the expert and had a significantly higher performance than that of inexperienced radiologists. AI could also have a fundamental role in reducing the US interpretation time, as demonstrated by Lai et al.’s study [34], in which it was reduced by approximately 40% due to AI system support.
Concluding, at present, many studies have shown that AI has excellent diagnostic performance in breast cancer diagnosis [61,62]. However, most studies are only single-center studies, and in most cases, there was no independent external test set.

4.2. Prediction of Molecular Subtypes of Breast Cancer

As known, there are four different molecular subtypes of breast cancer: luminal A, luminal B, HER-2 overexpression, and triple-negative breast cancer (TNBC). Each subtype presents different biological characteristics, imaging characteristics, prognosis, and treatment [63]. Molecular subtypes, in fact, can affect the response to NAC; HER2+ and TNBC cancers have a higher probability of responding well to preoperative therapy, but, on the other hand, patients with nonluminal disease have worse recurrence-free and disease-specific survival [64]. Therefore, it is essential to make a histological and molecular diagnosis before surgery. Currently, the gold standard is the microhistological (core or vacuum) biopsy. One of the main limitations of pathological biopsy is the possible underestimation of the sample because of the heterogeneity of the tumor, and for this reason, the biopsy may sometimes not be representative of the cancer [65]. In the literature, there are several studies based on the prediction of breast cancer molecular subtypes based on the AI US system. Ma et al. [42] developed an ML system to differentiate luminal/HER 2+/TNBC/versus other subtypes, ER-positive versus ER-negative, HER2-positive versus HER2-negative, and high Ki-67 versus low Ki-67 expression, proving that a machine learning model can assist radiologists to evaluate the molecular subtype of breast cancer. Other studies, on the other hand, have focused only on the diagnosis of TN subtypes. Several retrospective studies showed, in fact, that TNBC was more likely to show benign features, such as an oval or round shape or smooth or circumscribed margins, and was less likely to have an echogenic halo [66]. All these characteristics may lead to a diagnostic delay, resulting in a poor outcome. Therefore, the AI-based diagnosis of TN breast cancer is crucial in clinical diagnosis and treatment. Every study included in this paper showed good diagnostic performance in the diagnosis of TNBC versus other histological subtypes. Zhou et al. [41] established a multimodal approach based on US, SWE, and CDFI to predict the subtype of breast cancer. This multimodal AI system performed better than US alone (AUC: 0.89–0.96 vs. 0.81–0.84) and, surprisingly, also performed better than the core needle biopsy (AUC: 0.89–0.99 vs. 0.67–0.82, p < 0.05). In clinical practice, a possible mismatch between the biopsy result and the AI algorithm could lead to a rebiopsy with the sample at another site. This model also achieved excellent results (AUC: 0.934–0.970) in differentiating TNBC and non-TNBC. AI has the potential to provide a noninvasive method of assessing tumor biology before surgical or medical treatment.

4.3. Prediction of Axillary Lymph Node Metastases in Breast Cancer

Identification of lymph node metastasis in breast cancer is crucial for correct diagnostic and therapeutic planning. US has a fundamental role in determining axillary lymph node (ALN) status. The prediction of preoperative lymph node metastases can provide valuable information for determining adjuvant therapy and making surgical plans (lymph node dissection) that are often associated with complications, such as lymphedema. Several studies have described a great deal of breast US characteristics associated with lymph node metastasis and, in addition, lymphatic invasion and the size of the breast cancer are associated with the presence of metastatic cells [67]. At US examination, lymph node metastasis is characterized by unclear margins, irregular shapes, and loss of fatty hilum, but lymph nodes with micrometastases are missed [68]. In addition, all these evaluations are based on expertise and experience, which are operator-dependent. In all the analyzed studies, AI algorithms improved metastatic lymph node detection compared with standard radiological evaluation. The studies mainly focused on predicting the presence or absence of ALN metastasis, but a maximum value of sensitivity below 0.7 could be considered promising but relatively low to enter into clinical practice soon. We acknowledge that differences in cut-off values and reference standards across the different studies involved could influence the results. In addition, we do not know to what extent the relatively low specificity of US in predicting LN metastasis could lead to overtreatment or undertreatment. Prospective, well-designed, and possibly multicentric studies are needed to clarify the role of US in LN metastasis prediction. Guo et al. [50] analyzed 3049 US images of 937 patients and developed a DL radiomics-based prediction model to assess the risk of metastasis of sentinel lymph nodes (SLNs) (AUC = 0.84, sensitivity = 98.4%) and nonsentinel lymph nodes (NSLNs) (AUC = 0.81, sensitivity = 98.4%). Zhou et al. [47] used three different AI algorithms, and all three performed better than the medical radiologist (the sensitivity and specificity of radiologists were 73% and 63%). Other studies also predicted the metastatic burden of ALNs. Zheng et al. [45] showed good results in predicting 1–2 (low metastatic burden) or ≥3 (heavy metastatic burden) ALN metastasis (AUC = 0.90). The prediction of lymph node metastasis by combining the US characteristics of the tumor, US characteristics of the lymph node, and artificial intelligence features could lead to a great diagnostic effect in clinical practice if diagnostic yield above standard radiological evaluation is at least equal to standard sentinel lymph node assessment.

4.4. Prediction Response to NAC in Breast Cancer

According to several guidelines [69,70], neoadjuvant chemotherapy (NAC) is the standard of care for patients with locally advanced breast cancer (LABC), biologically aggressive tumors, or nonsurgical patients. LABC is defined as a tumor greater than 5 cm and with skin and/or chest wall involvement or a tumor with many metastatic lymph nodes [71]. Pathological complete response (pCR) is associated with better clinical outcomes compared with nonresponder or partial responder patients. Combining US imaging technology with AI to extract quantitative information from US images can provide more objective information about the response to neoadjuvant chemotherapy. All the analyzed studies give good results in predicting the response to therapy. Jiang et al. [54] used AI to establish a pCR prediction model based on breast cancer US images before and after neoadjuvant chemotherapy in locally advanced breast cancer. The model had a good predictive value (AUC = 0.94). Gu et al. [55] used US images at different NAC time points to establish a NAC response (before, after the second course, and after the fourth course of NAC), with an AUC value that increased during the course of therapy (AUC of 0.812 after the second courses and an AUC of 0.937 after the fourth courses). Gu et al. [55] also identified 90.5% of nonresponder patients. Jiang et al. [54] also demonstrated that prediction within the hormone receptor–positive/human epidermal growth factor receptor 2 (HER2)–negative, HER2+, and triple-negative subgroups also achieved good discrimination performance, with an AUC of 0.90, 0.95, and 0.93. The prediction of the NAC response before starting treatment could be extremely useful for clinicians for risk stratification and targeting treatment, and in future, this could lead to precision medicine permitting therapy adjustments.

4.5. Upstage of DCIS in IDC

A correct histological diagnosis of BC before surgery is fundamental. In fact, sentinel lymph node biopsy is not usually performed in the case of partial mastectomy for DCIS [72]. This patient could undergo SLN biopsy if DCIS is upgraded postoperatively to invasive cancer. This could lead to a second surgery, with increased costs for the health system. One of the main limitations of microhistological biopsy is the possible underestimation of the sample because of the heterogeneity of the tumor [65]. For this reason, we sometimes observe an upgrade of DCIS to IDC. AI could also have an important role in this kind of prediction. Qian et al. [73] used a DL algorithm to predict whether simple DCIS diagnosed with core needle biopsy would be upgraded to invasive cancer after surgical excision. The proposed model achieved good sensitivity, specificity, and accuracy (0.733, 0.750, and 0.742).

4.6. Predicting BC Prognosis

Classifications of molecular subtypes have a key role in the management strategy of BC. Several studies investigated the role of AI in BC prognosis, especially for TNBC. TNBC had a worse overall survival because of its higher nuclear grade, larger tumor size, and more aggressive proliferative index. Extracting 460 radiomic characteristics, Wang et al. [74] established an ML model for predicting disease-free survival in TNBC. In addition, Yu et al. [75], in a large multicenter study, assessed that radiomics is a promising biomarker for risk stratification for TNBC patients

5. Conclusions

US-based AI has great potential and research value in the field of breast cancer diagnosis, treatment, and prognosis. AI is considered a “hot topic” in radiology, and it has the potential to carry us into the era of personalized medicine. AI looks promising in every field of study evaluated in this review. More prospective and multicentric studies are needed to assess the potential impact of AI in breast ultrasound and to understand how to insert AI into the clinical workflow of radiologists.

Author Contributions

N.B., investigation; A.S.T., data curation; N.B., writing—original draft preparation; A.S.T., writing—review and editing; M.C. and C.M., validation and supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ferlay, J.; Steliarova-Foucher, E.; Lortet-Tieulent, J.; Rosso, S.; Coebergh, J.W.; Comber, H.; Forman, D.; Bray, F. Cancer incidence and mortality patterns in Europe: Estimates for 40 countries in 2012. Eur. J. Cancer 2013, 49, 1374–1403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Yang, L.; Wang, S.; Zhang, L.; Sheng, C.; Song, F.; Wang, P.; Huang, Y. Performance of ultrasonography screening for breast cancer: A systematic review and meta-analysis. BMC Cancer 2020, 20, 499. [Google Scholar] [CrossRef] [PubMed]
  3. Litjens, G.; Kooi, T.; Bejnordi, B.E.; Setio, A.A.A.; Ciompi, F.; Ghafoorian, M.; Van Der Laak, J.A.; Van Ginneken, B.; Sánchez, C.I. A survey on deep learning in medical image analysis. Med. Image Anal. 2017, 42, 60–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. LeCun, Y.; Bengio, Y.; Hinton, G. Deep learning. Nature 2015, 521, 436–444. [Google Scholar] [CrossRef] [PubMed]
  5. Niu, S.; Huang, J.; Li, J.; Liu, X.; Wang, D.; Zhang, R.; Wang, Y.; Shen, H.; Qi, M.; Xiao, Y.; et al. Application of ultrasound artificial intelligence in the differential diagnosis between benign and malignant breast lesions of BI-RADS 4A. BMC Cancer 2020, 20, 959. [Google Scholar] [CrossRef]
  6. Fleury, E.; Marcomini, K. Performance of machine learning software to classify breast lesions using BI-RADS radiomic features on ultrasound images. Eur. Radiol. Exp. 2019, 3, 34. [Google Scholar] [CrossRef] [Green Version]
  7. Romeo, V.; Cuocolo, R.; Apolito, R.; Stanzione, A.; Ventimiglia, A.; Vitale, A.; Verde, F.; Accurso, A.; Amitrano, M.; Insabato, L.; et al. Clinical value of radiomics and machine learning in breast ultrasound: A multicenter study for differential diagnosis of benign and malignant lesions. Eur. Radiol. 2021, 31, 9511–9519. [Google Scholar] [CrossRef]
  8. Lyu, S.-Y.; Zhang, Y.; Zhang, M.-W.; Zhang, B.-S.; Gao, L.-B.; Bai, L.-T.; Wang, J. Diagnostic value of artificial intelligence automatic detection systems for breast BI-RADS 4 nodules. World J. Clin. Cases 2022, 10, 518–527. [Google Scholar] [CrossRef]
  9. Hayashida, T.; Odani, E.; Kikuchi, M.; Nagayama, A.; Seki, T.; Takahashi, M.; Futatsugi, N.; Matsumoto, A.; Murata, T.; Watanuki, R.; et al. Establishment of a deep-learning system to diagnose BI-RADS4a or higher using breast ultrasound for clinical application. Cancer Sci. 2022, 113, 3528. [Google Scholar] [CrossRef]
  10. Fujioka, T.; Kubota, K.; Mori, M.; Kikuchi, Y.; Katsuta, L.; Kasahara, M.; Oda, G.; Ishiba, T.; Nakagawa, T.; Tateishi, U. Distinction between benign and malignant breast masses at breast ultrasound using deep learning method with convolutional neural network. Jpn. J. Radiol. 2019, 37, 466–472. [Google Scholar] [CrossRef]
  11. Wang, X.Y.; Cui, L.G.; Feng, J.; Chen, W. Artificial intelligence for breast ultrasound: An adjunct tool to reduce excessive lesion biopsy. Eur. J. Radiol. 2021, 138, 109624. [Google Scholar] [CrossRef] [PubMed]
  12. Shen, Y.; Shamout, F.E.; Oliver, J.R.; Witowski, J.; Kannan, K.; Park, J.; Wu, N.; Huddleston, C.; Wolfson, S.; Millet, A.; et al. Artificial intelligence system reduces pre-positive findings in the interpretation of breast ultrasound exams. Nat. Commun. 2021, 12, 5645. [Google Scholar] [CrossRef] [PubMed]
  13. Huo, L.; Tan, Y.; Wang, S.; Geng, C.; Li, Y.; Ma, X.; Wang, B.; He, Y.; Yao, C.; Ouyang, T. Machine Learning Models to Improve the Differentiation Between Benign and Malignant Breast Lesions on Ultrasound: A Multicenter External Validation Study. Cancer Manag. Res. 2021, 13, 3367–3379. [Google Scholar] [CrossRef] [PubMed]
  14. Gao, Y.; Liu, B.; Zhu, Y.; Chen, L.; Tan, M.; Xiao, X.; Yu, G.; Guo, Y. Detection and recognition of ultrasound breast nodules based on semi-supervised deep learning: A powerful alternative strategy. Quant. Imaging Med. Surg. 2021, 11, 2265–2278. [Google Scholar] [CrossRef]
  15. Du, R.; Chen, Y.; Li, T.; Shi, L.; Fei, Z.; Li, Y. Discrimination of Breast Cancer Based on Ultrasound Images and Convolutional Neural Network. J. Oncol. 2022, 2022, 7733583. [Google Scholar] [CrossRef]
  16. Zhang, N.; Li, X.T.; Ma, L.; Fan, Z.Q.; Sun, Y.S. Application of deep learning to establish a diagnostic model of breast lesions using two-dimensional grayscale ultrasound imaging. Clin. Imaging 2021, 79, 56–63. [Google Scholar] [CrossRef]
  17. Wan, K.W.; Wong, C.H.; Ip, H.F.; Fan, D.; Yuen, P.L.; Fong, H.Y.; Ying, M. Evaluation of the performance of traditional machine learning algorithms, convolutional neural network and AutoML Vision in ultrasound breast lesions classification: A comparative study. Quant. Imaging Med. Surg. 2021, 11, 1381–1393. [Google Scholar] [CrossRef]
  18. Shia, W.C.; Lin, L.S.; Chen, D.R. Classification of malignant tumours in breast ultrasound using unsupervised machine learning approaches. Sci. Rep. 2021, 11, 1418. [Google Scholar] [CrossRef]
  19. Li, C.; Li, J.; Tan, T.; Chen, K.; Xu, Y.; Wu, R. Application of ultrasonic dual-mode artificially intelligent architecture in assisting radiologists with different diagnostic levels on breast masses classification. Diagn. Interv. Radiol. 2021, 27, 315–322. [Google Scholar] [CrossRef]
  20. Zhang, X.; Liang, M.; Yang, Z.; Zheng, C.; Wu, J.; Ou, B.; Li, H.; Wu, X.; Luo, B.; Shen, J. Deep Learning-Based Radiomics of B-Mode Ultrasonography and Shear-Wave Elastography: Improved Performance in Breast Mass Classification. Front. Oncol. 2020, 10, 1621. [Google Scholar] [CrossRef]
  21. Tanaka, H.; Chiu, S.W.; Watanabe, T.; Kaoku, S.; Yamaguchi, T. Computer-aided diagnosis system for breast ultrasound images using deep learning. Phys. Med. Biol. 2019, 64, 235013. [Google Scholar] [CrossRef] [PubMed]
  22. Qi, X.; Zhang, L.; Chen, Y.; Pi, Y.; Chen, Y.; Lv, Q.; Yi, Z. Automated diagnosis of breast ultrasonography images using deep neural networks. Med. Image Anal. 2019, 52, 185–198. [Google Scholar] [CrossRef] [PubMed]
  23. Byra, M.; Galperin, M.; Ojeda-Fournier, H.; Olson, L.; O’Boyle, M.; Comstock, C.; Andre, M. Breast mass classification in sonography with transfer learning using a deep convolutional neural network and color conversion. Med. Phys. 2019, 46, 746–755. [Google Scholar] [CrossRef] [PubMed]
  24. Xiao, T.; Liu, L.; Li, K.; Qin, W.; Yu, S.; Li, Z. Comparison of Transferred Deep Neural Networks in Ultrasonic Breast Masses Discrimination. BioMed Res. Int. 2018, 2018, 4605191. [Google Scholar] [CrossRef]
  25. Sultan, L.R.; Schultz, S.M.; Cary, T.W.; Sehgal, C.M. Machine learning to improve breast cancer diagnosis by multimodal ultrasound. In Proceedings of the 2018 IEEE International Ultrasonics Symposium (IUS), Kobe, Japan, 22–25 October 2018. [Google Scholar]
  26. Becker, A.S.; Mueller, M.; Stoffel, E.; Marcon, M.; Ghafoor, S.; Boss, A. Classification of breast cancer in ultrasound imaging using a generic deep learning analysis software: A pilot study. Br. J. Radiol. 2018, 91, 20170576. [Google Scholar] [CrossRef]
  27. Gu, Y.; Xu, W.; Bin Lin, B.; An, X.; Tian, J.; Ran, H.; Ren, W.; Chang, C.; Yuan, J.; Kang, C.; et al. Deep learning based on ultrasound images assists breast lesion diagnosis in China: A multicenter diagnostic study. Insights Imaging 2022, 13, 124. [Google Scholar] [CrossRef]
  28. Wei, Q.; Yan, Y.-J.; Wu, G.-G.; Ye, X.-R.; Jiang, F.; Liu, J.; Wang, G.; Wang, Y.; Song, J.; Pan, Z.-P.; et al. The diagnostic performance of ultrasound computer-aided diagnosis system for distinguishing breast masses: A prospective multicenter study. Eur. Radiol. 2022, 32, 4046–4055. [Google Scholar] [CrossRef]
  29. Wilding, R.C.; Sheraton, V.M.; Soto, L.; Chotai, N.; Tan, E.Y. Deep learning applied to breast imaging classification and segmentation with human expert intervention. J. Ultrasound. 2022, 25, 659–666. [Google Scholar] [CrossRef]
  30. O’Connell, A.M.; Bartolotta, T.V.; Orlando, A.; Jung, S.H.; Baek, J.; Parker, K.J.J. Diagnostic Performance of an Artificial Intelligence System in Breast Ultrasound. Ultrasound Med. 2022, 41, 97–105. [Google Scholar] [CrossRef]
  31. Ma, H.; Tian, R.; Li, H.; Sun, H.; Lu, G.; Liu, R.; Wang, Z. Fus2Net: A novel Convolutional Neural Network for classification of benign and malignant breast tumor in ultrasound images. Biomed. Eng. Online 2021, 20, 112. [Google Scholar] [CrossRef]
  32. Chowdhury, A.; Razzaque, R.R.; Muhtadi, S. Ultrasound classification of breast masses using a comprehensive Nakagami imaging and machine learning framework. Ultrasonics 2022, 124, 106744. [Google Scholar] [CrossRef] [PubMed]
  33. Li, J.; Bu, Y.; Lu, S.; Pang, H.; Luo, C.; Liu, Y.; Qian, L.J. Development of a Deep Learning-Based Model for Diagnosing Breast Nodules with Ultrasound. Ultrasound Med. 2021, 40, 513–520. [Google Scholar] [CrossRef] [PubMed]
  34. Lai, Y.C.; Chen, H.H.; Hsu, J.F.; Hong, Y.J.; Chiu, T.T.; Chiou, H.J. Evaluation of physician performance using a concurrent-read artificial intelligence system to support breast ultrasound interpretation. Breast 2022, 65, 124–135. [Google Scholar] [CrossRef]
  35. Li, Y.; Gu, H.; Wang, H.; Qin, P.; Wang, J. BUSnet: A Deep Learning Model of Breast Tumor Lesion Detection for Ultrasound Images. Front. Oncol. 2022, 12, 848271. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, H.; Han, L.; Chen, K.; Peng, Y.; Lin, J. Diagnostic Efficiency of the Breast Ultrasound Computer-Aided Prediction Model Based on Convolutional Neural Network in Breast Cancer. J. Digit. Imaging 2020, 33, 1218–1223. [Google Scholar] [CrossRef] [PubMed]
  37. Ye, H.; Hang, J.; Zhang, M. Automatic identification of triple negative breast cancer in ultrasonography using a deep convolutional neural network. Sci. Rep. 2021, 11, 20474. [Google Scholar] [CrossRef] [PubMed]
  38. Zhang, X.; Li, H.; Wang, C.; Cheng, W.; Zhu, Y.; Li, D.; Jing, H.; Li, S.; Hou, J.; Li, J.; et al. Evaluating the Accuracy of Breast Cancer and Molecular Subtype Diagnosis by Ultrasound Image Deep Learning Model. Front. Oncol. 2021, 11, 623506. [Google Scholar] [CrossRef]
  39. Xu, Z.; Yang, Q.; Li, M.; Gu, J.; Du, C.; Chen, Y.; Li, B. Predicting HER2 Status in Breast Cancer on Ultrasound Images Using Deep Learning Method. Front. Oncol. 2022, 12, 829041. [Google Scholar] [CrossRef]
  40. Wu, T.; Sultan, L.R.; Tian, J.; Cary, T.W.; Sehgal, C.M. Machine learning for diagnostic ultrasound of triple-negative breast cancer. Breast Cancer Res. Treat. 2019, 173, 365–373. [Google Scholar] [CrossRef]
  41. Zhou, B.-Y.; Wang, L.-F.; Yin, H.-H.; Wu, T.-F.; Ren, T.-T.; Peng, C.; Li, D.-X.; Shi, H.; Sun, L.-P.; Zhao, C.-K.; et al. Decoding the molecular subtypes of breast cancer seen on multimodal ultrasound images using an assembled convolutional neural network model: A prospective and multicentre study. EBioMedicine 2021, 74, 103684. [Google Scholar] [CrossRef]
  42. Ma, M.; Liu, R.; Wen, C.; Xu, W.; Xu, Z.; Wang, S.; Wu, J.; Pan, D.; Zheng, B.; Qin, G.; et al. Predicting the molecular subtype of breast cancer and identifying interpretable imaging features using machine learning algorithms. Eur. Radiol. 2022, 32, 1652–1662. [Google Scholar] [CrossRef] [PubMed]
  43. Guo, Y.; Hu, Y.; Qiao, M.; Wang, Y.; Yu, J.; Li, J.; Chang, C. Radiomics Analysis on Ultrasound for Prediction of Biologic Behavior in Breast Invasive Ductal Carcinoma. Clin. Breast Cancer 2018, 18, e335–e344. [Google Scholar] [CrossRef] [PubMed]
  44. Jiang, M.; Zhang, D.; Tang, S.C.; Luo, X.M.; Chuan, Z.R.; Lv, W.Z.; Jiang, F.; Ni, X.J.; Cui, X.W.; Dietrich, C.F. Deep learning with convolutional neural network in the assessment of breast cancer molecular subtypes based on US images: A multicenter retrospective study. Eur. Radiol. 2021, 31, 3673–3682. [Google Scholar] [CrossRef] [PubMed]
  45. Zheng, X.; Yao, Z.; Huang, Y.; Yu, Y.; Wang, Y.; Liu, Y.; Mao, R.; Li, F.; Xiao, Y.; Wang, Y.; et al. Deep learning radiomics can predict axillary lymph node status in early-stage breast cancer. Nat. Commun. 2020, 11, 1236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Jiang, M.; Li, C.-L.; Luo, X.-M.; Chuan, Z.-R.; Chen, R.-X.; Tang, S.-C.; Lv, W.-Z.; Cui, X.-W.; Dietrich, C.F. Radiomics model based on shear-wave elastography in the assessment of axillary lymph node status in early-stage breast cancer. Eur. Radiol. 2022, 32, 2313–2325. [Google Scholar] [CrossRef]
  47. Zhou, L.-Q.; Wu, X.-L.; Huang, S.-Y.; Wu, G.-G.; Ye, H.-R.; Wei, Q.; Bao, L.-Y.; Deng, Y.-B.; Li, X.-R.; Cui, X.-W.; et al. Lymph Node Metastasis Prediction from Primary Breast Cancer US Images Using Deep Learning. Radiology 2020, 294, 19–28. [Google Scholar] [CrossRef]
  48. Tahmasebi, A.; Qu, E.; Sevrukov, A.; Liu, J.-B.; Wang, S.; Lyshchik, A.; Yu, J.; Eisenbrey, J.R. Assessment of Axillary Lymph Nodes for Metastasis on Ultrasound Using Artificial Intelligence. Ultrason. Imaging 2021, 43, 329–336. [Google Scholar] [CrossRef]
  49. Ozaki, J.; Fujioka, T.; Yamaga, E.; Hayashi, A.; Kujiraoka, Y.; Imokawa, T.; Takahashi, K.; Okawa, S.; Yashima, Y.; Mori, M.; et al. Deep learning method with a convolutional neural network for image classification of normal and metastatic axillary lymph nodes on breast ultrasonography. Jpn. J. Radiol. 2022, 40, 814–822. [Google Scholar] [CrossRef]
  50. Guo, X.; Liu, Z.; Sun, C.; Zhang, L.; Wang, Y.; Li, Z.; Shi, J.; Wu, T.; Cui, H.; Zhang, J.; et al. Deep learning radiomics of ultrasonography: Identifying the risk of axillary non-sentinel lymph node involvement in primary breast cancer. EBioMedicine 2020, 60, 103018. [Google Scholar] [CrossRef]
  51. Lee, Y.W.; Huang, C.S.; Shih, C.C.; Chang, R.F. Axillary lymph node metastasis status prediction of early-stage breast cancer using convolutional neural networks. Comput. Biol. Med. 2021, 130, 104206. [Google Scholar] [CrossRef]
  52. Sun, S.; Mutasa, S.; Liu, M.Z.; Nemer, J.; Sun, M.; Siddique, M.; Desperito, E.; Jambawalikar, S.; Ha, R.S. Deep learning prediction of axillary lymph node status using ultrasound images. Comput. Biol. Med. 2022, 143, 105250. [Google Scholar] [CrossRef]
  53. DiCenzo, D.; Quiaoit, K.; Fatima, K.; Bhardwaj, D.; Sannachi, L.; Gangeh, M.; Sadeghi-Naini, A.; Dasgupta, A.; Kolios, M.C.; Trudeau, M.; et al. Quantitative ultrasound radiomics in predicting response to neoadjuvant chemotherapy in patients with locally advanced breast cancer: Results from multi-institutional study. Cancer Med. 2020, 9, 5798–5806. [Google Scholar] [CrossRef] [PubMed]
  54. Jiang, M.; Li, C.-L.; Luo, X.-M.; Chuan, Z.-R.; Lv, W.-Z.; Li, X.; Cui, X.-W.; Dietrich, C.F. Ultrasound-based deep learning radiomics in the assessment of pathological complete response to neoadjuvant chemotherapy in locally advanced breast cancer. Eur. J. Cancer 2021, 147, 95–105. [Google Scholar] [CrossRef] [PubMed]
  55. Gu, J.; Tong, T.; He, C.; Xu, M.; Yang, X.; Tian, J.; Jiang, T.; Wang, K. Deep learning radiomics of ultrasonography can predict response to neoadjuvant chemotherapy in breast cancer at an early stage of treatment: A prospective study. Eur. Radiol. 2022, 32, 2099–2109. [Google Scholar] [CrossRef] [PubMed]
  56. Taleghamar, H.; Jalalifar, S.A.; Czarnota, G.J.; Sadeghi-Naini, A. Deep learning of quantitative ultrasound multi-parametric images at pre-treatment to predict breast cancer response to chemotherapy. Sci. Rep. 2022, 12, 2244. [Google Scholar] [CrossRef]
  57. Xie, J.; Shi, H.; Du, C.; Song, X.; Wei, J.; Dong, Q.; Wan, C. Dual-Branch Convolutional Neural Network Based on Ultrasound Imaging in the Early Prediction of Neoadjuvant Chemotherapy Response in Patients with Locally Advanced Breast Cancer. Front. Oncol. 2022, 12, 812463. [Google Scholar] [CrossRef]
  58. Byra, M.; Dobruch-Sobczak, K.; Klimonda, Z.; Piotrzkowska-Wroblewska, H.; Litniewski, J. Early Prediction of Response to Neoadjuvant Chemotherapy in Breast Cancer Sonography Using Siamese Convolutional Neural Networks. IEEE J. Biomed. Health Inform. 2021, 25, 797–805. [Google Scholar] [CrossRef]
  59. D’Orsi, C.; Sickles, E.; Mendelson, E.; Morris, E. ACR BI-RADS® Atlas, Breast Imaging Reporting and Data System, 5th ed.; American College of Radiology: Reston, VA, USA, 2013. [Google Scholar]
  60. Hosny, A.; Parmar, C.; Quackenbush, J.; Schwartz, L.H.; Aerts, H. Artificial intelligence in radiology. Nat. Rev. Cancer 2018, 18, 500–510. [Google Scholar] [CrossRef]
  61. Fujioka, T.; Mori, M.; Kubota, K.; Oyama, J.; Yamaga, E.; Yashima, Y.; Katsuta, L.; Nomura, K.; Nara, M.; Oda, G.; et al. The Utility of Deep Learning in Breast Ultrasonic Imaging: A Review. Diagnostics 2020, 10, 1055. [Google Scholar] [CrossRef]
  62. Wu, G.-G.; Zhou, L.-Q.; Xu, J.-W.; Wang, J.-Y.; Wei, Q.; Deng, Y.-B.; Cui, X.-W.; Dietrich, C.F. Artificial intelligence in breast ultrasound. World J. Radiol. 2019, 11, 19–26. [Google Scholar] [CrossRef]
  63. Prat, A.; Pineda, E.; Adamo, B.; Galván, P.; Fernández, A.; Gaba, L.; Díez, M.; Viladot, M.; Arance, A.; Muñoz, M. Clinical implications of the intrinsic molecular subtypes of breast cancer. Breast 2015, 24 (Suppl. 2), S26–S35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Chen, X.S.; Wu, J.Y.; Huang, O.U.; Chen, C.M.; Wu, J.; Lu, J.S.; Shao, Z.M.; Shen, Z.Z.; Shen, K.W. Molecular subtype can predict the response and outcome of Chinese locally advanced breast cancer patients treated with preoperative therapy. Oncol. Rep. 2010, 23, 1213–1220. [Google Scholar] [PubMed]
  65. Schueller, G.; Jaromi, S.; Ponhold, L.; Fuchsjaeger, M.; Memarsadeghi, M.; Rudas, M.; Weber, M.; Liberman, L.; Helbich, T.H. US-guided 14-gauge core-needle breast biopsy: Results of a validation study in 1352 cases. Radiology 2008, 248, 406–413. [Google Scholar] [CrossRef] [PubMed]
  66. Costantini, M.; Belli, P.; Bufi, E.; Asunis, A.M.; Ferra, E.; Bitti, G.T. Association between sonographic appearances of breast cancers and their histopathologic features and biomarkers. J. Clin. Ultrasound 2016, 44, 26–33. [Google Scholar] [CrossRef] [PubMed]
  67. Fujii, T.; Yajima, R.; Tatsuki, H.; Suto, T.; Morita, H.; Tsutsumi, S.; Kuwano, H. Significance of lymphatic invasion combined with size of primary tumor for predicting sentinel lymph node metastasis in patients with breast cancer. Anticancer Res. 2015, 35, 3581–3584. [Google Scholar] [PubMed]
  68. De Boer, M.; Van Deurzen, C.H.; Van Dijck, J.A.; Borm, G.F.; Van Diest, P.J.; Adang, E.M.; Nortier, J.W.; Rutgers, E.J.; Seynaeve, C.; Menke-Pluymers, M.B.; et al. Micrometastases or isolated tumor cells and the outcome of breast cancer. N. Engl. J. Med. 2009, 361, 653–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Available online: https://old-prod.asco.org/practice-patients/guidelines/breast-cancer (accessed on 29 November 2022).
  70. Available online: https://www.esmo.org/guidelines/guidelines-by-topic/breast-cancer (accessed on 29 November 2022).
  71. Hortobagyi, G.N. Comprehensive management of locally advanced breast cancer. Cancer 1990, 66 (Suppl. 6), 1387–1391. [Google Scholar] [CrossRef]
  72. Lyman, G.H.; Somerfield, M.R.; Giuliano, A.E. Sentinel Lymph Node Biopsy for Patients with Early-Stage Breast Cancer: 2016 American Society of Clinical Oncology Clinical Practice Guideline Update Summary. J. Oncol. Pract. 2017, 13, 196–198. [Google Scholar] [CrossRef]
  73. Qian, L.; Lv, Z.; Zhang, K.; Wang, K.; Zhu, Q.; Zhou, S.; Chang, C.; Tian, J. Application of deep learning to predict underestimation in ductal carcinoma in situ of the breast with ultrasound. Ann. Transl. Med. 2021, 9, 295. [Google Scholar] [CrossRef]
  74. Wang, H.; Li, X.; Yuan, Y.; Tong, Y.; Zhu, S.; Huang, R.; Shen, K.; Guo, Y.; Wang, Y.; Chen, X. Association of machine learning ultrasound radiomics and disease outcome in triple negative breast cancer. Am. J. Cancer Res. 2022, 12, 152–164. [Google Scholar]
  75. Yu, F.; Hang, J.; Deng, J.; Yang, B.; Wang, J.; Ye, X.; Liu, Y. Radiomics features on ultrasound imaging for the prediction of disease-free survival in triple negative breast cancer: A multi-institutional study. Br. J. Radiol. 2021, 94, 20210188. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart: lesion detection, segmentation, validation, and test dataset.
Figure 1. Flowchart: lesion detection, segmentation, validation, and test dataset.
Diagnostics 13 00058 g001
Figure 2. Flowchart of literature search.
Figure 2. Flowchart of literature search.
Diagnostics 13 00058 g002
Table 1. Specifications of studies on BC diagnosis included in this review.
Table 1. Specifications of studies on BC diagnosis included in this review.
StudyAimModalityDesignMethodTest DatasetTraining and Validation DatasetPerformance
Niu et al. 2020 [5]Diagnosis of BC among BI-RADS 4a lesionsUSSingle-center retrospectiveDL 206 images AUC = 0.8746,
ACC = 88.75%,
SENS = 87.35%,
SPEC = 90.32%
Fleury et al. 2019 [6]Benign or malignant lesionsUS Single-center prospective ML206 lesions (144 benign, 62 malignant) AUC = 0.840,
SENS = 71.4%, SPEC = 76.9%
Romeo et al. 2021 [7] Noncystic benign and malignant breast lesions USMulticenter retrospectiveML 174 patients, 201 lesions135 patientsAUC = 0.82, ACC = 82%, SENS = 93%,
SPEC = 57%, PPV = 82%, NPV = 80%
Lyu et al. 2022 [8]Reduce the probability of BI-RADS 4 biopsyUSSingle-center prospectiveDL107 lesions SENS = 93.48%, SPEC = 67.21%, ACC = 78.50%, VPP = 68.25%, VPN = 93.18%
Hayashida et al. 2019 [9]Classify lesion in BI-RADS 3 or lower or BI-RADS 4a or higherUS, Single-center retrospectiveDL 3656 lesionsAUC = 0.95, SENS = 91.2%, SPEC = 90.7%
Fujioka et al. 2019 [10]Distinguish between benign and malignant massesUSSingle-center retrospectiveDL 48 benign masses, 72 malignant masses 480 images of 96 benign masses and 467 images of 144 malignant masses SENS = 0.958, SPEC = 0.875, ACC = 0.925, AUC = 0.913
Wang et al. 2021 [11]Reduce excessive lesion biopsyUSSingle-center prospectiveDL167 patients with 173 lesions SENS = 94.9, SPEC = 69.4, AUC = 0.82, ACC = 80.9
Shen et al.
2021 [12]
Identify malignant lesions and reduce false positive examinationsUS, CDFIMulticenter retrospective DL5442907 images 4584271 imagesAUC = 0.976 for internal test set,
AUC = 0.927 for external test set
Huo et al. 2021 [13] Distinguish between malignant and benign lesions USMulticenter retrospectiveML 1125 cases 1965 cases AUC = 0.906, ACC = 0.772, precision = 0.528, recall = 0.936, F1-score = 0.675
Gao et al. 2021 [14] Distinguish between malignant and benign lesions USMulticenter retrospectiveDL 8966 nodules 5396 nodules AUC = 0.934
Du et al 2022 [15] Distinguish between malignant and benign lesions USSingle-center retrospectiveDL1181 images1031 imagesACC = 92.6%
Zhang et al. 2021 [16]Establish an AI model USSingle-center retrospectiveDL1311 lesions1109 lesionsAUC = 0.846, ACC = 77.0%, SENS = 82.0%, SPEC = 71.7%, PPV = 79.3%, NPV = 75.1%
Wan et al. 2021 [17] Distinguish between malignant and benign lesions USRetrospectiveML895 images800 imagesAUC = 0.95, ACC = 0.86, SENS = 0.84, SPEC = 0.88
Shia et al. 2021 [18] Distinguish and classify between malignant and benign lesions USSingle-center retrospective DL677 images AUC = 0.938, SENS = 94.34 %, SPEC = 93.22 %, PPV = 92.6%, NPV = 94.8%
Li et al. 2021 [19]Radiologist performance with and without AIUS, SWERetrospectiveDL2967 images2368 imagesAUC = 0.892, ACC = 92.4%, SENS = 81.5%, SPEC = 96.9%
Zhang et al. 2020 [20]Classify breast lesionsUS, SWERetrospective DL291 masses198 massesUS-AUC = 0.99, SWE-AUC = 0.99
Tanaka et al. 2019 [21]Classify breast lesionsUSMulticenter retrospective DL1536 masses1382 massesAUC = 0.951, ACC = 89.0%, SENS = 90.9%, SPEC = 87.0%
Qi et al. 2019 [22]Distinguish malignant tumors and
solid nodules
USSingle-center retrospectiveDL8145 images6786 imagesAUC = 0.982, ACC = 94.48%, SENS = 95.65%, SPEC = 93.88%
Byra et al. 2019 [23]Classify breast mass USRetrospectiveDL882 images732 imagesAUC = 0.936, ACC = 0.887, SENS = 0.848, SPEC = 0.897
Xiao et al. 2018 [24]Differentiate benign and malignant tumorsUSSingle-center retrospectiveDL2058 lesion AUC = 0.93, ACC = 89.44%, SENS = 88.73%, SPEC = 89.91%
Sultan et al.
2018 [25]
Breast cancer
diagnosis
US + CDFISingle-center retrospectiveML160 lesions AUC = 0.89, SENS = 79%, SPEC = 89%
Becker et al.
2018 [26]
Classify breast cancerUSSingle-center retrospectiveDL637 lesions,
637 images
637 imagesAUC = 0.84, SENS = 84.2%, SPEC = 80.3%, PPV = 32.0%, NPV = 97.9%
Gu et al. 2022 [27] Differentiating benign from malignant breast lesions USMulticenter prospectiveDL 14,043 US images 4149 AUC = 0.913, SENS = 88.84%, SPEC = 83.77%, ACC = 86.40%
Wei et al. 2022 [28] Distinguishing benign and malignant breast masses USMulticenter prospectiveML 901 breast masses ACC = 89.0%, SENS = 91.4%, SPEC = 87.7%, PVV = 80.8%, NPV = 94.7%
Wilding et al. 2022 [29]Benign and malignant classesUSSingle-center retrospectiveML953 masses ACC = 96%
O’Connell et al. 2022 [30]Assist radiologists in the diagnosis of breast cancerUSMulticenter retrospective DL299 lesions SENS, SPEC and ACC greater than 0.8
Ma et al. 2021 [31] Assist radiologists in the diagnosis of benign and malignant tumors USSingle-center retrospective DL952 lesions 1052 images ACC = 92%, SENS = 95.65%, SPEC = 88.89%, AUC = 0.97
Chowdhury et al. 2022 [32] Assist in the diagnosis by helping to reduce false positive diagnoses USRetrospectiveML ACC = 93.08%, AUC = 0.9712, FNR = 0%, FPR = 8.65%
Li et al. 2021 [33] Distinguish benign from malignant lesions USMulticenter retrospective DL 692 images 271 malignant nodules, 1053 benign nodules and 2144 images of the contralateral normal breast AUC = 0.943, SENS = 73.3%, SPEC = 94.9%
Lai et al. 2022 [34] Compare the diagnostic performance between reading without AI USSingle-center retrospective DL172 lesions Increase in the average AUC ROC from 0.7582 to 0.8294
Li et al. 2022 [35] Enhance the detection accuracy with AI system USSingle-center retrospective DL156 images624 imagesAccuracy benign = 0.651
Accuracy malignant = 0.579
Zhang et al. 2020 [36] Prediction of breast cancer USSingle-center retrospective DL1007 images5000 imagesAUC = 0.913
Table 2. Specifications of studies on prediction of molecular subtypes of BC included in this review.
Table 2. Specifications of studies on prediction of molecular subtypes of BC included in this review.
StudyAimModalityDESIGNMethodTest DatasetTraining and Validation DatasetPerformance
Ye et al. 2021 [37]Identify TNBC from non-TNBCUSSingle-center retrospectiveDL 204 images (102 NTN and 102 TN) AUC = 0.8746, ACC = 88.75%, SENS = 87.35%, SPEC = 90.32%
Zhang et al. 2021 [38]Predict molecular subtypesUS Multicenter retrospective DL1235 images, 790 patientsPatients/images: 684/986
HR+ = 417/588, HER2+ = 149/212, TNBC = 118/186
AUC 0.864, 0.811 and 0.837 for TN, HER2 (+) and HR (+) subtype
Xu et al. 2022 [39]Predict the expression of HER2USSingle-center retrospectiveDL 144 patients144 patientsAUC = 0.84, ACC = 80.56%, SENS = 72.73%, SPEC = 84.00%, PPV = 66.67%, NPV = 87.5%
Wu et al. 2019 [40]Identify TNBC from non-TNBCUSSingle-center retrospectiveML140 images131 patientsAUC = 0.88, SENS = 86.96%, SPEC = 82.91%
Zhou et al. 2021 [41]Predict four- and five-classification molecular subtypesUS, SWE, CDFIMulticenter prospectiveDL 807 patients, 818 images 534 patients, 545 breast cancersTest A: AUC = 0.92, SENS = 91.67%, SPEC = 78.57%, ACC = 81.72%
Test B: AUC = 0.96, SENS = 87.50%, SPEC = 87.50%, ACC = 82.11%
Ma et al. 2022 [42]Predict molecular subtypesMammography, USSingle-center retrospectiveML600 patients450 patientsTNBC vs. other: AUC = 0.971, ACC = 0.947, SENS = 0.905, SPEC = 0.941
Luminal vs. other: AUC = 0.900, ACC = 0.860, SENS = 0.871, SPEC = 0.886
HER2 vs. other: AUC = 0.855, ACC = 0.893, SENS = 0.900, SPEC = 0.724
ER-positive vs. ER-negative: AUC = 0.878, ACC = 0.867, SENS = 0.940, SPEC = 0.788
Guo et al. 2018 [43]Assess biological characteristicsUSSingle-center retrospectiveML215 patients SENS = 0.979, SPEC = 0.601, AUC = 0.760
Jiang et al. 2021 [44]Luminal disease from non luminal diseaseUSMulticenter retrospective DL1132 images, 845 patients4828 US images from 1275 patientsTest A: Luminal A: AUC = 0.89, ACC = 86.96%
Luminal Test B: AUC = 0.86, ACC = 80.07%
HER2+: AUC = 0.80, ACC = 90.88%
TNBC: AUC = 0.82, ACC = 97.02%
Table 3. Specifications of studies on prediction of axillary lymph node metastases in BC included in this review.
Table 3. Specifications of studies on prediction of axillary lymph node metastases in BC included in this review.
StudyAimModalityDesignMethodTest DatasetTraining and Validation DatasetPerformance
Zheng et al. 2020 [45]Predict ALN status (N0-N1)USProspectiveDL584 lesions 466 lesionsAUC = 0.902, ACC = 81.0%, SENS = 81.6%, SPEC = 83.6%, PPV = 78.4%, NPV = 86.2%
Jiang et al. 2021 [46]ALN burden in early-stage breast cancerUS, SWE Multicenter retrospective DL 303 + 130 patientsC-index of 0.845 for the training cohort and 0.817 for the validation cohort
Zhou et al. 2020 [47] Clinically negative axillary lymph node metastasis USMulticenter retrospectiveDL834 patients, 1055 images680 patients, 877 imagesAUC = 0.89, ACC = 79%, SENS = 85%, SPEC = 73%, PPV = 76%, NPV = 81%
Tahmasebi et al. 2021 [48]Benign and malignant ALNUSSingle-center retrospectiveML317 images64 imagesAUC = 0.78, ACC = 69.5%, SENS = 74.0%, SPEC = 64.4%, PPV = 68.3%, NPV = 72.6%
Ozaki et al. 2022 [49]Normal and metastatic axillary lymph nodeUSSingle-center retrospectiveDL 300 images normal and 328 images metastatic 100 images (50 normal, 50 metastases)SENS = 94%, SPEC = 88%, AUC = 0.966
Guo et al. 2020 [50] Predict the risk of sentinel LN and nonsentinel LN metastasis USMulticenter retrospectiveDL3049 images542 patients (SLN), 180 patients (NSLN)SLN: AUC = 0.848, VPP = 61.5%, VPN = 97%
NSLN: AUC = 0.81, VPP = 78.55%, VPN = 91.7%
Lee at al. 2021 [51] Predict the axillary lymph node (ALN) metastatic status in patients with early-stage breast cancer USSingle-center retrospectiveDL153 patients153 patientsAUC = 0.805, ACC = 81.05%, SENS = 81.36%, SPEC = 80.85%, PPV = 72.73%, NPV = 87.36%
Sun et al. 2022 [52]Predict ALN
metastasis
USSingle-center retrospectiveDL338 images248 imagesAUC = 0.72, ACC = 72.6%, SENS = 65.5%, SPEC = 78.9%
Table 4. Specifications of studies on prediction of response to NAC included in this review.
Table 4. Specifications of studies on prediction of response to NAC included in this review.
StudyAimModalityDesignMethodTest DatasetTraining and Validation DatasetPerformance
DiCenzo et al. 2020 [53]Assess pCR to NACUS, QUSMulticenter prospectiveML82 patients SENS = 91.2, SPEC = 83.3, AUC = 0.726
Jiang et al. 2021 [54]Assess pCR to NACUS Multicenter retrospective DL592 patients356 patientsSENS = 89.3%, SPEC = 81.3%, AUC = 0.94
Gu et al. 2021 [55]Response to NAC after the second course and fourth course of therapyUSSingle-center prospectiveDL (two different models)592 patients356 patientsDLR-2: AUC = 0.812, ACC = 69.1%, SENS = 90.5%, SPEC = 47.6%, NPV = 83.3%
DLR-4: AUC = 0.937, ACC = 85.7%, SENS = 81.0%, SPEC = 90.5%, NPV = 82.6%
Taleghamar et al. 2022 [56]Responders and nonresponders before starting therapyQUSSingle-center retrospectiveDL181 patients131 patientsAUC = 0.86, ACC = 88%,
SENS = 70%, SPEC = 92.5%
Xie et al. 2022 [57]Predict pCR before and after the first stage of NACUSSingle-center retrospectiveDL114 patients, 968 images776 imagesAUC = 0.939, ACC = 87.50%, SENS = 90.67%, SPEC = 85.67%, PPV = 80.00%, NPV = 93.46%
Byra et al. 2021 [58]Responders and nonrespondersUSSingle-center retrospectiveDL39 tumors After the second NAC: AUC = 0.847, ACC = 76.9%, SENS = 78.9%, SPEC = 75.2%
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Brunetti, N.; Calabrese, M.; Martinoli, C.; Tagliafico, A.S. Artificial Intelligence in Breast Ultrasound: From Diagnosis to Prognosis—A Rapid Review. Diagnostics 2023, 13, 58. https://doi.org/10.3390/diagnostics13010058

AMA Style

Brunetti N, Calabrese M, Martinoli C, Tagliafico AS. Artificial Intelligence in Breast Ultrasound: From Diagnosis to Prognosis—A Rapid Review. Diagnostics. 2023; 13(1):58. https://doi.org/10.3390/diagnostics13010058

Chicago/Turabian Style

Brunetti, Nicole, Massimo Calabrese, Carlo Martinoli, and Alberto Stefano Tagliafico. 2023. "Artificial Intelligence in Breast Ultrasound: From Diagnosis to Prognosis—A Rapid Review" Diagnostics 13, no. 1: 58. https://doi.org/10.3390/diagnostics13010058

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop