Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cancer Imaging ; 24(1): 48, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38576031

RESUMO

BACKGROUND: Ductal Carcinoma In Situ (DCIS) can progress to invasive breast cancer, but most DCIS lesions never will. Therefore, four clinical trials (COMET, LORIS, LORETTA, AND LORD) test whether active surveillance for women with low-risk Ductal carcinoma In Situ is safe (E. S. Hwang et al., BMJ Open, 9: e026797, 2019, A. Francis et al., Eur J Cancer. 51: 2296-2303, 2015, Chizuko Kanbayashi et al. The international collaboration of active surveillance trials for low-risk DCIS (LORIS, LORD, COMET, LORETTA),  L. E. Elshof et al., Eur J Cancer, 51, 1497-510, 2015). Low-risk is defined as grade I or II DCIS. Because DCIS grade is a major eligibility criteria in these trials, it would be very helpful to assess DCIS grade on mammography, informed by grade assessed on DCIS histopathology in pre-surgery biopsies, since surgery will not be performed on a significant number of patients participating in these trials. OBJECTIVE: To assess the performance and clinical utility of a convolutional neural network (CNN) in discriminating high-risk (grade III) DCIS and/or Invasive Breast Cancer (IBC) from low-risk (grade I/II) DCIS based on mammographic features. We explored whether the CNN could be used as a decision support tool, from excluding high-risk patients for active surveillance. METHODS: In this single centre retrospective study, 464 patients diagnosed with DCIS based on pre-surgery biopsy between 2000 and 2014 were included. The collection of mammography images was partitioned on a patient-level into two subsets, one for training containing 80% of cases (371 cases, 681 images) and 20% (93 cases, 173 images) for testing. A deep learning model based on the U-Net CNN was trained and validated on 681 two-dimensional mammograms. Classification performance was assessed with the Area Under the Curve (AUC) receiver operating characteristic and predictive values on the test set for predicting high risk DCIS-and high-risk DCIS and/ or IBC from low-risk DCIS. RESULTS: When classifying DCIS as high-risk, the deep learning network achieved a Positive Predictive Value (PPV) of 0.40, Negative Predictive Value (NPV) of 0.91 and an AUC of 0.72 on the test dataset. For distinguishing high-risk and/or upstaged DCIS (occult invasive breast cancer) from low-risk DCIS a PPV of 0.80, a NPV of 0.84 and an AUC of 0.76 were achieved. CONCLUSION: For both scenarios (DCIS grade I/II vs. III, DCIS grade I/II vs. III and/or IBC) AUCs were high, 0.72 and 0.76, respectively, concluding that our convolutional neural network can discriminate low-grade from high-grade DCIS.


Assuntos
Neoplasias da Mama , Carcinoma Ductal de Mama , Carcinoma Intraductal não Infiltrante , Aprendizado Profundo , Humanos , Feminino , Carcinoma Intraductal não Infiltrante/diagnóstico por imagem , Carcinoma Intraductal não Infiltrante/patologia , Estudos Retrospectivos , Participação do Paciente , Conduta Expectante , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/patologia , Mamografia , Carcinoma Ductal de Mama/diagnóstico , Carcinoma Ductal de Mama/patologia , Carcinoma Ductal de Mama/cirurgia
2.
J Magn Reson Imaging ; 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38581127

RESUMO

In breast imaging, there is an unrelenting increase in the demand for breast imaging services, partly explained by continuous expanding imaging indications in breast diagnosis and treatment. As the human workforce providing these services is not growing at the same rate, the implementation of artificial intelligence (AI) in breast imaging has gained significant momentum to maximize workflow efficiency and increase productivity while concurrently improving diagnostic accuracy and patient outcomes. Thus far, the implementation of AI in breast imaging is at the most advanced stage with mammography and digital breast tomosynthesis techniques, followed by ultrasound, whereas the implementation of AI in breast magnetic resonance imaging (MRI) is not moving along as rapidly due to the complexity of MRI examinations and fewer available dataset. Nevertheless, there is persisting interest in AI-enhanced breast MRI applications, even as the use of and indications of breast MRI continue to expand. This review presents an overview of the basic concepts of AI imaging analysis and subsequently reviews the use cases for AI-enhanced MRI interpretation, that is, breast MRI triaging and lesion detection, lesion classification, prediction of treatment response, risk assessment, and image quality. Finally, it provides an outlook on the barriers and facilitators for the adoption of AI in breast MRI. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY: Stage 6.

3.
IEEE Trans Med Imaging ; PP2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38530714

RESUMO

Pulmonary nodules may be an early manifestation of lung cancer, the leading cause of cancer-related deaths among both men and women. Numerous studies have established that deep learning methods can yield high-performance levels in the detection of lung nodules in chest X-rays. However, the lack of gold-standard public datasets slows down the progression of the research and prevents benchmarking of methods for this task. To address this, we organized a public research challenge, NODE21, aimed at the detection and generation of lung nodules in chest X-rays. While the detection track assesses state-of-the-art nodule detection systems, the generation track determines the utility of nodule generation algorithms to augment training data and hence improve the performance of the detection systems. This paper summarizes the results of the NODE21 challenge and performs extensive additional experiments to examine the impact of the synthetically generated nodule training images on the detection algorithm performance.

4.
J Med Imaging (Bellingham) ; 11(1): 014001, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38162417

RESUMO

Purpose: We developed a segmentation method suited for both raw (for processing) and processed (for presentation) digital mammograms (DMs) that is designed to generalize across images acquired with systems from different vendors and across the two standard screening views. Approach: A U-Net was trained to segment mammograms into background, breast, and pectoral muscle. Eight different datasets, including two previously published public sets and six sets of DMs from as many different vendors, were used, totaling 322 screen film mammograms (SFMs) and 4251 DMs (2821 raw/processed pairs and 1430 only processed) from 1077 different women. Three experiments were done: first training on all SFM and processed images, second also including all raw images in training, and finally testing vendor generalization by leaving one dataset out at a time. Results: The model trained on SFM and processed mammograms achieved a good overall performance regardless of projection and vendor, with a mean (±std. dev.) dice score of 0.96±0.06 for all datasets combined. When raw images were included in training, the mean (±std. dev.) dice score for the raw images was 0.95±0.05 and for the processed images was 0.96±0.04. Testing on a dataset with processed DMs from a vendor that was excluded from training resulted in a difference in mean dice varying between -0.23 to +0.02 from that of the fully trained model. Conclusions: The proposed segmentation method yields accurate overall segmentation results for both raw and processed mammograms independent of view and vendor. The code and model weights are made available.

5.
Med Image Anal ; 92: 103044, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38043455

RESUMO

Multi-sequence MRIs can be necessary for reliable diagnosis in clinical practice due to the complimentary information within sequences. However, redundant information exists across sequences, which interferes with mining efficient representations by learning-based models. To handle various clinical scenarios, we propose a sequence-to-sequence generation framework (Seq2Seq) for imaging-differentiation representation learning. In this study, not only do we propose arbitrary 3D/4D sequence generation within one model to generate any specified target sequence, but also we are able to rank the importance of each sequence based on a new metric estimating the difficulty of a sequence being generated. Furthermore, we also exploit the generation inability of the model to extract regions that contain unique information for each sequence. We conduct extensive experiments using three datasets including a toy dataset of 20,000 simulated subjects, a brain MRI dataset of 1251 subjects, and a breast MRI dataset of 2101 subjects, to demonstrate that (1) top-ranking sequences can be used to replace complete sequences with non-inferior performance; (2) combining MRI with our imaging-differentiation map leads to better performance in clinical tasks such as glioblastoma MGMT promoter methylation status prediction and breast cancer pathological complete response status prediction. Our code is available at https://github.com/fiy2W/mri_seq2seq.


Assuntos
Glioblastoma , Imageamento por Ressonância Magnética , Humanos , Mama
6.
Invest Radiol ; 59(3): 230-242, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-37493391

RESUMO

ABSTRACT: Primary systemic therapy (PST) is the treatment of choice in patients with locally advanced breast cancer and is nowadays also often used in patients with early-stage breast cancer. Although imaging remains pivotal to assess response to PST accurately, the use of imaging to predict response to PST has the potential to not only better prognostication but also allow the de-escalation or omission of potentially toxic treatment with undesirable adverse effects, the accelerated implementation of new targeted therapies, and the mitigation of surgical delays in selected patients. In response to the limited ability of radiologists to predict response to PST via qualitative, subjective assessments of tumors on magnetic resonance imaging (MRI), artificial intelligence-enhanced MRI with classical machine learning, and in more recent times, deep learning, have been used with promising results to predict response, both before the start of PST and in the early stages of treatment. This review provides an overview of the current applications of artificial intelligence to MRI in assessing and predicting response to PST, and discusses the challenges and limitations of their clinical implementation.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/terapia , Neoplasias da Mama/tratamento farmacológico , Inteligência Artificial , Mama/patologia , Imageamento por Ressonância Magnética , Aprendizado de Máquina
7.
Insights Imaging ; 14(1): 213, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38051355

RESUMO

BACKGROUND: Calcifications on mammography can be indicative of breast cancer, but the prognostic value of their appearance remains unclear. This systematic review and meta-analysis aimed to evaluate the association between mammographic calcification morphology descriptors (CMDs) and clinicopathological factors. METHODS: A comprehensive literature search in Medline via Ovid, Embase.com, and Web of Science was conducted for articles published between 2000 and January 2022 that assessed the relationship between CMDs and clinicopathological factors, excluding case reports and review articles. The risk of bias and overall quality of evidence were evaluated using the QUIPS tool and GRADE. A random-effects model was used to synthesize the extracted data. This systematic review is reported according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA). RESULTS: Among the 4715 articles reviewed, 29 met the inclusion criteria, reporting on 17 different clinicopathological factors in relation to CMDs. Heterogeneity between studies was present and the overall risk of bias was high, primarily due to small, inadequately described study populations. Meta-analysis demonstrated significant associations between fine linear calcifications and high-grade DCIS [pooled odds ratio (pOR), 4.92; 95% confidence interval (CI), 2.64-9.17], (comedo)necrosis (pOR, 3.46; 95% CI, 1.29-9.30), (micro)invasion (pOR, 1.53; 95% CI, 1.03-2.27), and a negative association with estrogen receptor positivity (pOR, 0.33; 95% CI, 0.12-0.89). CONCLUSIONS: CMDs detected on mammography have prognostic value, but there is a high level of bias and variability between current studies. In order for CMDs to achieve clinical utility, standardization in reporting of CMDs is necessary. CRITICAL RELEVANCE STATEMENT: Mammographic calcification morphology descriptors (CMDs) have prognostic value, but in order for CMDs to achieve clinical utility, standardization in reporting of CMDs is necessary. SYSTEMATIC REVIEW REGISTRATION: CRD42022341599 KEY POINTS: • Mammographic calcifications can be indicative of breast cancer. • The prognostic value of mammographic calcifications is still unclear. • Specific mammographic calcification morphologies are related to lesion aggressiveness. • Variability between studies necessitates standardization in calcification evaluation to achieve clinical utility.

9.
Cell Rep Med ; 4(8): 101131, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37490915

RESUMO

Digital health data used in diagnostics, patient care, and oncology research continue to accumulate exponentially. Most medical information, and particularly radiology results, are stored in free-text format, and the potential of these data remains untapped. In this study, a radiological repomics-driven model incorporating medical token cognition (RadioLOGIC) is proposed to extract repomics (report omics) features from unstructured electronic health records and to assess human health and predict pathological outcome via transfer learning. The average accuracy and F1-weighted score for the extraction of repomics features using RadioLOGIC are 0.934 and 0.934, respectively, and 0.906 and 0.903 for the prediction of breast imaging-reporting and data system scores. The areas under the receiver operating characteristic curve for the prediction of pathological outcome without and with transfer learning are 0.912 and 0.945, respectively. RadioLOGIC outperforms cohort models in the capability to extract features and also reveals promise for checking clinical diagnoses directly from electronic health records.


Assuntos
Doenças Mamárias , Radiologia , Humanos , Registros Eletrônicos de Saúde , Curva ROC , Atenção à Saúde
10.
Semin Radiat Oncol ; 32(4): 330-342, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36202436

RESUMO

Automatic image registration plays an important role in many aspects of the radiation oncology workflow ranging from treatment simulation, image guided and adaptive radiotherapy, motion management and response evaluation. Traditional automatic registration algorithms are often time-consuming and further improvements in registration accuracy are required. Recently, a variety of AI-driven strategies for automatic image registrations have been developed. In this review an overview of the many applications of automatic image registration in radiation oncology is provided. Different learning strategies and network architectures have been reviewed and the current status of AI based automatic image registration algorithms in radiation oncology has been described. AI based strategies for automatic image registration typically do not outperform traditional strategies yet. Various promising approaches to further improve AI based image registrations are being explored. Therefore AI based automatic image registration may be the method of choice in the foreseeable future.


Assuntos
Inteligência Artificial , Radioterapia (Especialidade) , Algoritmos , Humanos
11.
Med Image Anal ; 79: 102464, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35596966

RESUMO

We propose a Deep learning-based weak label learning method for analyzing whole slide images (WSIs) of Hematoxylin and Eosin (H&E) stained tumor tissue not requiring pixel-level or tile-level annotations using Self-supervised pre-training and heterogeneity-aware deep Multiple Instance LEarning (DeepSMILE). We apply DeepSMILE to the task of Homologous recombination deficiency (HRD) and microsatellite instability (MSI) prediction. We utilize contrastive self-supervised learning to pre-train a feature extractor on histopathology tiles of cancer tissue. Additionally, we use variability-aware deep multiple instance learning to learn the tile feature aggregation function while modeling tumor heterogeneity. For MSI prediction in a tumor-annotated and color normalized subset of TCGA-CRC (n=360 patients), contrastive self-supervised learning improves the tile supervision baseline from 0.77 to 0.87 AUROC, on par with our proposed DeepSMILE method. On TCGA-BC (n=1041 patients) without any manual annotations, DeepSMILE improves HRD classification performance from 0.77 to 0.81 AUROC compared to tile supervision with either a self-supervised or ImageNet pre-trained feature extractor. Our proposed methods reach the baseline performance using only 40% of the labeled data on both datasets. These improvements suggest we can use standard self-supervised learning techniques combined with multiple instance learning in the histopathology domain to improve genomic label classification performance with fewer labeled data.


Assuntos
Neoplasias da Mama , Neoplasias Colorretais , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Feminino , Humanos , Instabilidade de Microssatélites , Coloração e Rotulagem
12.
Semin Nucl Med ; 52(5): 584-596, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35339259

RESUMO

This review gives an overview of the current state of deep learning research in breast cancer imaging. Breast imaging plays a major role in detecting breast cancer at an earlier stage, as well as monitoring and evaluating breast cancer during treatment. The most commonly used modalities for breast imaging are digital mammography, digital breast tomosynthesis, ultrasound and magnetic resonance imaging. Nuclear medicine imaging techniques are used for detection and classification of axillary lymph nodes and distant staging in breast cancer imaging. All of these techniques are currently digitized, enabling the possibility to implement deep learning (DL), a subset of Artificial intelligence, in breast imaging. DL is nowadays embedded in a plethora of different tasks, such as lesion classification and segmentation, image reconstruction and generation, cancer risk prediction, and prediction and assessment of therapy response. Studies show similar and even better performances of DL algorithms compared to radiologists, although it is clear that large trials are needed, especially for ultrasound and magnetic resonance imaging, to exactly determine the added value of DL in breast cancer imaging. Studies on DL in nuclear medicine techniques are only sparsely available and further research is mandatory. Legal and ethical issues need to be considered before the role of DL can expand to its full potential in clinical breast care practice.


Assuntos
Neoplasias da Mama , Aprendizado Profundo , Inteligência Artificial , Mama/diagnóstico por imagem , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/patologia , Feminino , Humanos , Mamografia/métodos
13.
Radiology ; 303(1): 54-62, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34981975

RESUMO

Background Improving diagnosis of ductal carcinoma in situ (DCIS) before surgery is important in choosing optimal patient management strategies. However, patients may harbor occult invasive disease not detected until definitive surgery. Purpose To assess the performance and clinical utility of mammographic radiomic features in the prediction of occult invasive cancer among women diagnosed with DCIS on the basis of core biopsy findings. Materials and Methods In this Health Insurance Portability and Accountability Act-compliant retrospective study, digital magnification mammographic images were collected from women who underwent breast core-needle biopsy for calcifications that was performed at a single institution between September 2008 and April 2017 and yielded a diagnosis of DCIS. The database query was directed at asymptomatic women with calcifications without a mass, architectural distortion, asymmetric density, or palpable disease. Logistic regression with regularization was used. Differences across training and internal test set by upstaging rate, age, lesion size, and estrogen and progesterone receptor status were assessed by using the Kruskal-Wallis or χ2 test. Results The study consisted of 700 women with DCIS (age range, 40-89 years; mean age, 59 years ± 10 [standard deviation]), including 114 with lesions (16.3%) upstaged to invasive cancer at subsequent surgery. The sample was split randomly into 400 women for the training set and 300 for the testing set (mean ages: training set, 59 years ± 10; test set, 59 years ± 10; P = .85). A total of 109 radiomic and four clinical features were extracted. The best model on the test set by using all radiomic and clinical features helped predict upstaging with an area under the receiver operating characteristic curve of 0.71 (95% CI: 0.62, 0.79). For a fixed high sensitivity (90%), the model yielded a specificity of 22%, a negative predictive value of 92%, and an odds ratio of 2.4 (95% CI: 1.8, 3.2). High specificity (90%) corresponded to a sensitivity of 37%, positive predictive value of 41%, and odds ratio of 5.0 (95% CI: 2.8, 9.0). Conclusion Machine learning models that use radiomic features applied to mammographic calcifications may help predict upstaging of ductal carcinoma in situ, which can refine clinical decision making and treatment planning. © RSNA, 2022.


Assuntos
Neoplasias da Mama , Calcinose , Carcinoma in Situ , Carcinoma Ductal de Mama , Carcinoma Intraductal não Infiltrante , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/diagnóstico por imagem , Carcinoma Ductal de Mama/patologia , Carcinoma Intraductal não Infiltrante/diagnóstico por imagem , Carcinoma Intraductal não Infiltrante/patologia , Feminino , Humanos , Masculino , Mamografia , Pessoa de Meia-Idade , Estudos Retrospectivos
14.
Clin Transl Radiat Oncol ; 32: 6-14, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34765748

RESUMO

BACKGROUND/PURPOSE: Oropharyngeal cancer (OPC) primary gross tumor volume (GTVp) segmentation is crucial for radiotherapy. Multiparametric MRI (mpMRI) is increasingly used for OPC adaptive radiotherapy but relies on manual segmentation. Therefore, we constructed mpMRI deep learning (DL) OPC GTVp auto-segmentation models and determined the impact of input channels on segmentation performance. MATERIALS/METHODS: GTVp ground truth segmentations were manually generated for 30 OPC patients from a clinical trial. We evaluated five mpMRI input channels (T2, T1, ADC, Ktrans, Ve). 3D Residual U-net models were developed and assessed using leave-one-out cross-validation. A baseline T2 model was compared to mpMRI models (T2 + T1, T2 + ADC, T2 + Ktrans, T2 + Ve, all five channels [ALL]) primarily using the Dice similarity coefficient (DSC). False-negative DSC (FND), false-positive DSC, sensitivity, positive predictive value, surface DSC, Hausdorff distance (HD), 95% HD, and mean surface distance were also assessed. For the best model, ground truth and DL-generated segmentations were compared through a blinded Turing test using three physician observers. RESULTS: Models yielded mean DSCs from 0.71 ± 0.12 (ALL) to 0.73 ± 0.12 (T2 + T1). Compared to the T2 model, performance was significantly improved for FND, sensitivity, surface DSC, HD, and 95% HD for the T2 + T1 model (p < 0.05) and for FND for the T2 + Ve and ALL models (p < 0.05). No model demonstrated significant correlations between tumor size and DSC (p > 0.05). Most models demonstrated significant correlations between tumor size and HD or Surface DSC (p < 0.05), except those that included ADC or Ve as input channels (p > 0.05). On average, there were no significant differences between ground truth and DL-generated segmentations for all observers (p > 0.05). CONCLUSION: DL using mpMRI provides reasonably accurate segmentations of OPC GTVp that may be comparable to ground truth segmentations generated by clinical experts. Incorporating additional mpMRI channels may increase the performance of FND, sensitivity, surface DSC, HD, and 95% HD, and improve model robustness to tumor size.

15.
Radiother Oncol ; 165: 52-59, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34688808

RESUMO

BACKGROUND AND PURPOSE: Large radiotherapy (RT) planning imaging datasets with consistently contoured cardiovascular structures are essential for robust cardiac radiotoxicity research in thoracic cancers. This study aims to develop and validate a highly accurate automatic contouring model for the heart, cardiac chambers, and great vessels for RT planning computed tomography (CT) images that can be used for dose-volume parameter estimation. MATERIALS AND METHODS: A neural network model was trained using a dataset of 127 expertly contoured planning CT images from RT treatment of locally advanced non-small-cell lung cancer (NSCLC) patients. Evaluation of geometric accuracy and quality of dosimetric parameter estimation was performed on 50 independent scans with contrast and without contrast enhancement. The model was further evaluated regarding the clinical acceptability of the contours in 99 scans randomly sampled from the RTOG-0617 dataset by three experienced radiation oncologists. RESULTS: Median surface dice at 3 mm tolerance for all dedicated thoracic structures was 90% in the test set. Median absolute difference between mean dose computed with model contours and expert contours was 0.45 Gy averaged over all structures. The mean clinical acceptability rate by majority vote in the RTOG-0617 scans was 91%. CONCLUSION: This model can be used to contour the heart, cardiac chambers, and great vessels in large datasets of RT planning thoracic CT images accurately, quickly, and consistently. Additionally, the model can be used as a time-saving tool for contouring in clinic practice.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Aprendizado Profundo , Neoplasias Pulmonares , Humanos , Órgãos em Risco , Planejamento da Radioterapia Assistida por Computador , Tomografia Computadorizada por Raios X
16.
Magn Reson Imaging ; 84: 35-40, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34560230

RESUMO

PURPOSE: In this study, we compare readout-segmented echo-planar imaging (rs-EPI) Diffusion Weighted Imaging (DWI) to a work-in-progress single-shot EPI with modified Inversion Recovery Background Suppression (ss-EPI-mIRBS) sequence at 3 T using a b-value of 2000 s/mm2 on image quality, lesion visibility and evaluation time. METHOD: From September 2017 to December 2018, 23 women (one case used for training) with known breast cancer were included in this study, after providing signed informed consent. Women were scanned with the conventional rs-EPI sequence and the work-in-progress ss-EPI-mIRBS during the same examination. Four breast radiologists (4-13 years of experience) independently scored both series for overall image quality (1: extremely poor to 9: excellent). All lesions (47 in total, 36 malignant, and 11 benign and high-risk) were evaluated for visibility (1: not visible, 2: visible if location is given, 3: visible) and probability of malignancy (BI-RADS 1 to 5). ADC values were determined by measuring signal intensity in the lesions using dynamic contrast-enhanced (DCE) images for reference. Evaluation times for all assessments were automatically recorded. Results were analyzed using the visual grading characteristics (VGC) and the resulting area under the curve (AUCVGC) method. Statistical analysis was performed in SPSS, with McNemar tests, and paired t-tests used for comparison. RESULTS: No significant differences were detected between the two sequences in image quality (AUCVGC: 0.398, p = 0.087) and lesion visibility (AUCVGC: 0.534, p = 0.336) scores. Lesion characteristics (e.g benign and high-risk, versus malignant; small (≤10 mm) vs. larger (>10 mm)) did not result in different image quality or lesion visibility between sequences. Sensitivity (rs-EPI: 72.2% vs. ss-EPImIRBS: 78.5%, p = 0.108) and specificity (70.5% vs. 56.8%, p = 0.210, respectively) were comparable. In both sequences the mean ADC value was higher for benign and high-risk lesions than for malignant lesions (ss-EPI-mIRBS: p = 0.022 and rs-EPI: p = 0.055). On average, ss-EPI-mIRBS resulted in decreased overall reading time by 7.7 s/case (p = 0.067); a reduction of 17%. For malignant lesions, average reading time was significantly shorter using ss-EPI-mIRBS compared to rs-EPI (64.0 s/lesion vs. 75.9 s/lesion, respectively, p = 0.039). CONCLUSION: Based on this study, the ss-EPI sequence using a b-value of 2000 s/mm2 enables for a mIRBS acquisition with quality and lesion conspicuity that is comparable to conventional rs-EPI, but with a decreased reading time.


Assuntos
Neoplasias da Mama , Imagem de Difusão por Ressonância Magnética , Mama/diagnóstico por imagem , Neoplasias da Mama/diagnóstico por imagem , Imagem de Difusão por Ressonância Magnética/métodos , Imagem Ecoplanar/métodos , Feminino , Humanos
17.
Med Phys ; 48(10): 5897-5907, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34370886

RESUMO

PURPOSE: We propose a deep learning-based computer-aided detection (CADe) method to detect breast lesions in ultrafast DCE-MRI sequences. This method uses both the 3D spatial information and temporal information obtained from the early-phase of the dynamic acquisition. METHODS: The proposed CADe method, based on a modified 3D RetinaNet model, operates on ultrafast T1 weighted sequences, which are preprocessed for motion compensation, temporal normalization, and are cropped before passing into the model. The model is optimized to enable the detection of relatively small breast lesions in a screening setting, focusing on detection of lesions that are harder to differentiate from confounding structures inside the breast. RESULTS: The method was developed based on a dataset consisting of 489 ultrafast MRI studies obtained from 462 patients containing a total of 572 lesions (365 malignant, 207 benign) and achieved a detection rate, sensitivity, and detection rate of benign lesions of 0.90 (0.876-0.934), 0.95 (0.934-0.980), and 0.81 (0.751-0.871) at four false positives per normal breast with 10-fold cross-testing, respectively. CONCLUSIONS: The deep learning architecture used for the proposed CADe application can efficiently detect benign and malignant lesions on ultrafast DCE-MRI. Furthermore, utilizing the less visible hard-to-detect lesions in training improves the learning process and, subsequently, detection of malignant breast lesions.


Assuntos
Neoplasias da Mama , Aprendizado Profundo , Mama/diagnóstico por imagem , Neoplasias da Mama/diagnóstico por imagem , Meios de Contraste , Feminino , Humanos , Imageamento por Ressonância Magnética , Movimento (Física)
18.
Eur Radiol ; 31(11): 8682-8691, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33948701

RESUMO

OBJECTIVES: Digital breast tomosynthesis (DBT) increases sensitivity of mammography and is increasingly implemented in breast cancer screening. However, the large volume of images increases the risk of reading errors and reading time. This study aims to investigate whether the accuracy of breast radiologists reading wide-angle DBT increases with the aid of an artificial intelligence (AI) support system. Also, the impact on reading time was assessed and the stand-alone performance of the AI system in the detection of malignancies was compared to the average radiologist. METHODS: A multi-reader multi-case study was performed with 240 bilateral DBT exams (71 breasts with cancer lesions, 70 breasts with benign findings, 339 normal breasts). Exams were interpreted by 18 radiologists, with and without AI support, providing cancer suspicion scores per breast. Using AI support, radiologists were shown examination-based and region-based cancer likelihood scores. Area under the receiver operating characteristic curve (AUC) and reading time per exam were compared between reading conditions using mixed-models analysis of variance. RESULTS: On average, the AUC was higher using AI support (0.863 vs 0.833; p = 0.0025). Using AI support, reading time per DBT exam was reduced (p < 0.001) from 41 (95% CI = 39-42 s) to 36 s (95% CI = 35- 37 s). The AUC of the stand-alone AI system was non-inferior to the AUC of the average radiologist (+0.007, p = 0.8115). CONCLUSIONS: Radiologists improved their cancer detection and reduced reading time when evaluating DBT examinations using an AI reading support system. KEY POINTS: • Radiologists improved their cancer detection accuracy in digital breast tomosynthesis (DBT) when using an AI system for support, while simultaneously reducing reading time. • The stand-alone breast cancer detection performance of an AI system is non-inferior to the average performance of radiologists for reading digital breast tomosynthesis exams. • The use of an AI support system could make advanced and more reliable imaging techniques more accessible and could allow for more cost-effective breast screening programs with DBT.


Assuntos
Inteligência Artificial , Neoplasias da Mama , Mama/diagnóstico por imagem , Neoplasias da Mama/diagnóstico por imagem , Detecção Precoce de Câncer , Feminino , Humanos , Mamografia
19.
J Med Imaging (Bellingham) ; 8(2): 024501, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33796604

RESUMO

Purpose: A computer-aided diagnosis (CADx) system for breast masses is proposed, which incorporates both handcrafted and convolutional radiomic features embedded into a single deep learning model. Approach: The model combines handcrafted and convolutional radiomic signatures into a multi-view architecture, which retrieves three-dimensional (3D) image information by simultaneously processing multiple two-dimensional mass patches extracted along different planes through the 3D mass volume. Each patch is processed by a stream composed of two concatenated parallel branches: a multi-layer perceptron fed with automatically extracted handcrafted radiomic features, and a convolutional neural network, for which discriminant features are learned from the input patches. All streams are then concatenated together into a final architecture, where all network weights are shared and the learning occurs simultaneously for each stream and branch. The CADx system was developed and tested for diagnosis of breast masses ( N = 284 ) using image datasets acquired with independent dedicated breast computed tomography systems from two different institutions. The diagnostic classification performance of the CADx system was compared against other machine and deep learning architectures adopting handcrafted and convolutional approaches, and three board-certified breast radiologists. Results: On a test set of 82 masses (45 benign, 37 malignant), the proposed CADx system performed better than all other model architectures evaluated, with an increase in the area under the receiver operating characteristics curve (AUC) of 0.05 ± 0.02 , and achieving a final AUC of 0.947, outperforming the three radiologists ( AUC = 0.814 - 0.902 ). Conclusions: In conclusion, the system demonstrated its potential usefulness in breast cancer diagnosis by improving mass malignancy assessment.

20.
Med Image Anal ; 71: 102061, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33910108

RESUMO

The two-dimensional nature of mammography makes estimation of the overall breast density challenging, and estimation of the true patient-specific radiation dose impossible. Digital breast tomosynthesis (DBT), a pseudo-3D technique, is now commonly used in breast cancer screening and diagnostics. Still, the severely limited 3rd dimension information in DBT has not been used, until now, to estimate the true breast density or the patient-specific dose. This study proposes a reconstruction algorithm for DBT based on deep learning specifically optimized for these tasks. The algorithm, which we name DBToR, is based on unrolling a proximal-dual optimization method. The proximal operators are replaced with convolutional neural networks and prior knowledge is included in the model. This extends previous work on a deep learning-based reconstruction model by providing both the primal and the dual blocks with breast thickness information, which is available in DBT. Training and testing of the model were performed using virtual patient phantoms from two different sources. Reconstruction performance, and accuracy in estimation of breast density and radiation dose, were estimated, showing high accuracy (density <±3%; dose <±20%) without bias, significantly improving on the current state-of-the-art. This work also lays the groundwork for developing a deep learning-based reconstruction algorithm for the task of image interpretation by radiologists.


Assuntos
Neoplasias da Mama , Aprendizado Profundo , Mama/diagnóstico por imagem , Densidade da Mama , Neoplasias da Mama/diagnóstico por imagem , Feminino , Humanos , Mamografia , Doses de Radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA