Algorithmic Representation and Use Examples of Interface Intelligence Models

2018 ◽  
Vol 07 (04) ◽  
pp. 193-199
Author(s):  
烨斌 林
2021 ◽  
Vol 4 (1) ◽  
Author(s):  
Albert T. Young ◽  
Kristen Fernandez ◽  
Jacob Pfau ◽  
Rasika Reddy ◽  
Nhat Anh Cao ◽  
...  

AbstractArtificial intelligence models match or exceed dermatologists in melanoma image classification. Less is known about their robustness against real-world variations, and clinicians may incorrectly assume that a model with an acceptable area under the receiver operating characteristic curve or related performance metric is ready for clinical use. Here, we systematically assessed the performance of dermatologist-level convolutional neural networks (CNNs) on real-world non-curated images by applying computational “stress tests”. Our goal was to create a proxy environment in which to comprehensively test the generalizability of off-the-shelf CNNs developed without training or evaluation protocols specific to individual clinics. We found inconsistent predictions on images captured repeatedly in the same setting or subjected to simple transformations (e.g., rotation). Such transformations resulted in false positive or negative predictions for 6.5–22% of skin lesions across test datasets. Our findings indicate that models meeting conventionally reported metrics need further validation with computational stress tests to assess clinic readiness.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Amir Baghdadi ◽  
Sanju Lama ◽  
Rahul Singh ◽  
Hamidreza Hoshyarmanesh ◽  
Mohammadsaleh Razmi ◽  
...  

AbstractSurgical error and resulting complication have significant patient and economic consequences. Inappropriate exertion of tool-tissue force is a common variable for such error, that can be objectively monitored by sensorized tools. The rich digital output establishes a powerful skill assessment and sharing platform for surgical performance and training. Here we present SmartForceps data app incorporating an Expert Room environment for tracking and analysing the objective performance and surgical finesse through multiple interfaces specific for surgeons and data scientists. The app is enriched by incoming geospatial information, data distribution for engineered features, performance dashboard compared to expert surgeon, and interactive skill prediction and task recognition tools to develop artificial intelligence models. The study launches the concept of democratizing surgical data through a connectivity interface between surgeons with a broad and deep capability of geographic reach through mobile devices with highly interactive infographics and tools for performance monitoring, comparison, and improvement.


2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Xinran Wang ◽  
Liang Wang ◽  
Hong Bu ◽  
Ningning Zhang ◽  
Meng Yue ◽  
...  

AbstractProgrammed death ligand-1 (PD-L1) expression is a key biomarker to screen patients for PD-1/PD-L1-targeted immunotherapy. However, a subjective assessment guide on PD-L1 expression of tumor-infiltrating immune cells (IC) scoring is currently adopted in clinical practice with low concordance. Therefore, a repeatable and quantifiable PD-L1 IC scoring method of breast cancer is desirable. In this study, we propose a deep learning-based artificial intelligence-assisted (AI-assisted) model for PD-L1 IC scoring. Three rounds of ring studies (RSs) involving 31 pathologists from 10 hospitals were carried out, using the current guideline in the first two rounds (RS1, RS2) and our AI scoring model in the last round (RS3). A total of 109 PD-L1 (Ventana SP142) immunohistochemistry (IHC) stained images were assessed and the role of the AI-assisted model was evaluated. With the assistance of AI, the scoring concordance across pathologists was boosted to excellent in RS3 (0.950, 95% confidence interval (CI): 0.936–0.962) from moderate in RS1 (0.674, 95% CI: 0.614–0.735) and RS2 (0.736, 95% CI: 0.683–0.789). The 2- and 4-category scoring accuracy were improved by 4.2% (0.959, 95% CI: 0.953–0.964) and 13% (0.815, 95% CI: 0.803–0.827) (p < 0.001). The AI results were generally accepted by pathologists with 61% “fully accepted” and 91% “almost accepted”. The proposed AI-assisted method can help pathologists at all levels to improve the PD-L1 assay (SP-142) IC assessment in breast cancer in terms of both accuracy and concordance. The AI tool provides a scheme to standardize the PD-L1 IC scoring in clinical practice.


Sign in / Sign up

Export Citation Format

Share Document