Next Article in Journal
A Fault Diagnosis Approach Utilizing Artificial Intelligence for Maritime Power Systems within an Integrated Digital Twin Framework
Previous Article in Journal
The Impact of Job Insecurity on Miner Safety Behavior—A Study Based on SEM and fsQCA
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Explainable Artificial Intelligence (XAI) for Oncological Ultrasound Image Analysis: A Systematic Review

by
Lucie S. Wyatt
1,2,
Lennard M. van Karnenbeek
1,
Mark Wijkhuizen
1,
Freija Geldof
1 and
Behdad Dashtbozorg
1,*
1
Image-Guided Surgery, Department of Surgery, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
2
Technical Medicine, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The Netherlands
*
Author to whom correspondence should be addressed.
Appl. Sci. 2024, 14(18), 8108; https://doi.org/10.3390/app14188108
Submission received: 26 June 2024 / Revised: 4 September 2024 / Accepted: 7 September 2024 / Published: 10 September 2024
(This article belongs to the Section Applied Biosciences and Bioengineering)

Abstract

:
This review provides an overview of explainable AI (XAI) methods for oncological ultrasound image analysis and compares their performance evaluations. A systematic search of Medline Embase and Scopus between 25 March and 14 April 2024 identified 17 studies describing 14 XAI methods, including visualization, semantics, example-based, and hybrid functions. These methods primarily provided specific, local, and post hoc explanations. Performance evaluations focused on AI model performance, with limited assessment of explainability impact. Standardized evaluations incorporating clinical end-users are generally lacking. Enhanced XAI transparency may facilitate AI integration into clinical workflows. Future research should develop real-time methodologies and standardized quantitative evaluative metrics.

1. Introduction

Artificial intelligence (AI) technologies are omnipresent, with the healthcare industry experiencing exponential integration in recent years. Projections indicate further expansion of the global AI healthcare market, with an expected compound annual growth rate (CAGR) exceeding 36% from 2024 to 2030 [1,2]. The rapid growth is driven by several key factors, including the accumulation of patient health-related digital information, the increased demand for (personalized) medicine, and the intensified need to reduce care expenses and tackle healthcare workforce shortages [1,3,4].
As digital health data become increasingly available, so too does the pursuit to derive deeper insights from them. In response to the mounting pressure on healthcare systems, AI technologies are applied to offer diverse solutions [5,6]. Widely adopted and integrated AI algorithms can facilitate accurate and early-stage disease prediction, analyze complex medical images, and assist in the medical decision-making process [1,7]. These advancements have the promise to improve diagnosis, treatment, prognosis, and overall quality of life for patients across various conditions [8].
In the medical imaging field, ongoing research is exploring novel algorithms, architectures, and methodologies [2,9]. These innovations show the potential to aid clinicians in extracting decisive insights from medical images, identifying abnormalities, facilitating precision-guided interventions, and improving patient outcomes [7,10]. Machine learning (ML) and deep learning (DL) are specific subsets of AI. Specifically, DL algorithms employ neural networks and have demonstrated significant success in tasks such as image preprocessing, registration, segmentation, and classification, even surpassing human performance levels [11,12]. Research aimed at developing innovative and powerful system architectures for healthcare tasks has resulted in increasingly complex AI solutions [11,13]. However, the complex layered architecture of such neural networks presents challenges in directly interpreting results, due to their inherent “black box” properties. This limits the provision of intuitive explanations for the processes and outcomes of AI systems [11]. The need for transparent decision-making processes becomes apparent in healthcare settings, where AI-based support systems directly influence patient care. Additionally, to gain the trust of clinicians, the inner workings of medical AI systems should be understandable [11,14]. Furthermore, legal and ethical frameworks, such as the European Union’s General Data Protection Regulation (GDPR) and the European AI Act (AIA), mandate meaningful explanations of algorithmic decisions to ensure compliance and protect patient rights [15]. Similarly, identifying errors and potential biases is essential in developing and applying trustworthy AI systems in healthcare [16].
In response to the growing demand for explainability, the field of explainable AI (XAI) has emerged [17,18]. XAI encompasses “techniques that enable stakeholders to understand an AI algorithm and its decisions better” [11,17]. The global XAI market has been valued at USD 5 billion (2022) and is expected to reach over USD 24 billion by 2030, with a CAGR of 21.5% [19]. XAI for medicine is the second largest field in the trend, showing exponential growth [20]. Such explanations of intricate DL processes and outcomes for clinical applications impact functional benefits, clinical confidence, and patient outcomes [21]. Particularly, in the fight against cancer, where timely and accurate decisions are critical and DL model results can directly influence treatment decisions, the integration of XAI is fundamental [22]. Researchers have proposed various strategies for qualitative and quantitative XAI, including the use of comprehensible text, mathematics, and visualizations. However, a widely accepted benchmark for XAI is still lacking.
This need for XAI is underscored by the rapidly increasing global cancer burden. Over 35 million new cancer cases are predicted in 2050, depicting a 77% increase since 2022. The rapidly growing global cancer burden reflects both population aging and growth, as well as increased exposure to risk factors such as tobacco, obesity, and pollution [23]. In the past several years, researchers have developed AI tools based on deep knowledge that have been highly effective and have demonstrated great accuracy in oncological image analyses [24,25] and predicting various types of cancer, including breast, brain, lung, liver, and prostate cancer [8].
One particular area where XAI can provide significant advantages is in ultrasound (US) imaging. This widely used real-time, noninvasive, and relatively low-cost modality plays a crucial role in diagnosing and monitoring various cancers [26,27]. During oncological surgeries, it can be used, for example, for tumor localization and/or margin assessment [28,29,30]. Despite its benefits, US imaging faces challenges such as variability in operator performance and image quality, as well as the need for extensive training to accurately interpret images due to artifacts, noise, and the lack of distinct global anatomical structures [31]. These challenges present opportunities for XAI methods to enhance US image interpretation by providing transparent, understandable insights that aid clinicians in making fast and accurate decisions during clinical procedures. By offering real-time, interpretable feedback in terms of additional information or by highlighting areas of concern on images, XAI may enhance diagnostic accuracy, clinician confidence, and patient trust in AI-driven assessments.
While recent studies have started integrating XAI into AI-driven healthcare applications, a noticeable gap in research exists. Despite several reviews exploring XAI methods in general medical imaging applications [11,14,32], they lack a specific focus on oncological ultrasound imaging contexts. Moreover, these reviews fail to provide an extensive comparison of the advantages and disadvantages of various XAI methods for clinical use, as well as a comprehensive overview of the evaluation metrics employed. To the best of our knowledge, no review investigates the utilization and comparative effectiveness of XAI methods in oncological ultrasound imaging. This review aims to address this gap by investigating XAI methods for real-time ultrasound imaging data. This review will specifically analyze the role of XAI methods in segmentation, classification, and localization tasks. Our primary objective is to offer an overview of identified XAI methods and assess their qualitative and/or quantitative impact.

2. Materials and Methods

2.1. Search Strategy and Data Collection

Between 25 March 2024 and 14 April 2024, comprehensive searches were conducted in the Medline Embase [33] and Scopus [34] databases to identify relevant articles. The search strategy comprised terms related to explainable artificial intelligence, ultrasound imaging, and image analysis tasks, including segmentation, classification, and margin assessment (see Appendix A). No filters regarding publication dates were applied. Boolean operators were used to combine the major topic terms and their synonyms. The search was constrained to articles published in English, and duplicate articles were removed. Articles were included based on their focus on applying or developing XAI methods for ultrasound image analysis tasks.

2.2. Study Eligibility Criteria

Articles were systematically selected following the PRISMA 2020 statement [35]. The titles and abstracts of identified articles were screened by a single assessor (L.S.W.). Articles were excluded if they met one or more of the predefined exclusion criteria: (i) nonhuman studies, (ii) imaging modalities not aligned with ultrasound imaging, (iii) studies unrelated to cancer or oncological procedures, (iv) absence of XAI method discussion or analysis, (v) lack of focus on segmentation, classification, or margin assessment tasks, (vi) reviews, editorials, conference abstracts, letters to the editor, or studies with insufficient methodological detail, (vii) studies without medical topic, focus, or application. Articles with inaccessible full texts were excluded as well. The remaining full-text articles were further assessed for eligibility and solely included if none of the exclusion criteria were met.

2.3. Data Extraction

Data extraction was performed independently for all included studies. General study characteristics were collected, such as researched cancer type, clinical application, study design, and dataset specifics. The AI models developed in the studies were analyzed to provide contextual knowledge of the intrinsic data structures the XAI methods they aimed to explain. The types of XAI methods applied in the included articles were identified to provide an overview of the explanatory techniques employed in the AI-driven analysis of ultrasound imaging data within an oncological context. The characteristics of each identified XAI method were cataloged, including their operational mechanisms, explanatory function, input requirements, outputs, and evaluation metrics.
The operational mechanisms of methods were categorized using taxonomic criteria proposed in previous research [11,17,22,32]; these comprise the following:
  • Model-specific vs. model-agnostic: Model-specific explanation methods are limited to application on particular AI model architectures, e.g., a specific convolutional network model (CNN) (see Figure 1). These methods leverage the underlying network’s internal characteristics and use reverse engineering to generate their explanations. Model-agnostic explanation methods only operate on the model input and output; thus, they are independent of the model architecture. They aim to clarify the model’s underlying f ( x ) function, for example, by approximating f ( x ) with another, simpler model that should be explainable. Other agnostic methods attribute weights to each model variable, depending on its influence on the f ( x ) , to decompose the importance between variables [36].
  • Global vs. local scope: Global-scope explanations provide general relationships learned by the model, by assessing common patterns in the overall dataset that drive the model’s predictions (see Figure 2). Local-scope methods offer explanations of the model’s specific prediction for a given input or single case.
  • Intrinsic vs. post hoc explanation: Intrinsic explanation models can construct general, internal relationships between input and output made during predictions, due to their simple structure (e.g., decision trees, linear regression model, or support vector machine); see Figure 3. Post hoc explanations are applied to analyze models after these have completed training, providing insight into the learned relationships. The important difference is that post hoc explanations train a neural network and attempt to explain the behavior of the black box network after that. In contrast, intrinsic explanations force the neural network to be explainable itself [32].

2.4. Main Outcomes

The primary outcome was to provide an overview of XAI methods utilizing real-time ultrasound data for cancer-focused classification, segmentation, or localization tasks. The function-specific performance of XAI methods, which include explaining by examples, semantics, or, visualization, was assessed. This categorization of explanatory functions is based on proposed deep learning frameworks for medical image analysis [14,32]. Further elaboration focused on the advantages and disadvantages of each function’s identified XAI methods, emphasizing their clinical relevance and contribution. Additionally, the extent and impact of quantitative versus qualitative evaluations of XAI were explored across the included studies. Special emphasis was given to studies that integrated quantitative evaluations of XAI capabilities and assessments by clinical end-users, highlighting the practical impact of these technologies in clinical settings.

3. Results

3.1. Data Collection

On 29 May 2024, the search strategy identified a total of 222 articles from the Medline Embase (n = 57) and Scopus (n = 165) databases. After the removal of 65 duplicates, 157 articles remained for the initial title and abstract screening. The screening phase resulted in the exclusion of 129 articles due to nonhuman study models (n = 30), lack of real-time ultrasound imaging data (n = 51), irrelevance to cancer (n = 95), absence of XAI applications (n = 25), focus on impertinent tasks (n = 18), inappropriate study types (n = 19), and no medical application (n = 51). Excluded articles complied with one (n = 29) or multiple exclusion (n = 100) criteria.
Subsequently, the remaining 28 articles underwent full-text analysis. Two articles were excluded during the retrieval, due to their unavailability of full text. The full-text analysis of the remaining 26 articles resulted in an additional exclusion of 9 articles based on the predefined criteria. These exclusions encompassed articles that (i) investigated nonhuman subjects i.e., objects (n = 2) and animals (n = 1); (ii) utilized inappropriate imaging data types such as static photographs (n = 2), monomodal CT scans (n = 1), schematic images (n = 1), or the use of ultrasound for navigational purposes instead of image analysis (n = 1); (iii) lacked sufficient relevance to cancer or oncology (n = 4); (iv) only mentioned XAI as a future recommendation (n = 2) or in mathematical expressions (n = 2); (v) focused on peripheral tasks such as captioning (n = 1), eye-gaze mapping (n = 1), or lacked a specific task (n = 1); and (vi) were directed towards nonmedical image description applications (n = 3). Excluded articles complied with one (n = 4) or multiple (n = 4) exclusion criteria. Ultimately, 17 articles were eligible and selected for further in-depth analysis [37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]. Figure 4 provides a visual overview of the article selection process.

3.2. General Study Characteristics

The general study characteristics of all included articles (n = 17) were extracted and compared; see Table 1. The studies comprise a variety of cancer types, with the majority focused on breast (n = 8) or thyroid (n = 5) cancers. Further, most studies focus on diagnostic applications (n = 11), and only a few address surgical applications (n = 2). Regarding study type, most studies are retrospective (n = 14), indicating a common reliance on historical data for analysis.
Several studies employ extensive datasets, highlighting the significant data-handling capabilities of current medical image analysis technologies. In the study by Hassan et al. (2022) over 600,000 prostate multimodal MRI and US images were used [42]. In the studies by Song et al. (2023) and Qian et al. (2021), more than 19,000 and 10,000 breast US images were analyzed, respectively [47,49]. The number of included lesions was highest in Song (2023) et al. [49], Han (2022) et al. [41], and Karimzadeh (2023) et al. [43], all counting 2917 or more unique lesions.
The distribution of tissue types varies, encompassing two (n = 13) or three (n = 4) different tissue types. Studies with tripartite tissues distinguished between healthy, benign, and malignant in three studies [38,45,48], with a single study differentiating between three malignant variants [53]. The thirteen studies with binary tissue labels discriminated between benign and malignant tissue. In the twelve studies describing datasets’ class balance, eight studies were unbalanced, with malignant data as the minority in six studies [38,44,45,49,50,52] and the majority in two studies [48,53]. Four studies showed relative class balance [37,39,40,41].

3.3. Imaging Modality

Most of the included studies solely used ultrasound as imaging modality (n = 16), while one study combined US with MRI [42]. A detailed overview of the ultrasound systems covered in the included studies can be found in Appendix A.
Fourteen studies employed continuous US acquisition, underscoring a dynamic, real-time imaging trend. Rezazadeh (2022) et al. and Zheng (2024) et al. highlight the importance of live feedback during oncological procedures [48,53]. Two studies acquired 3D volume scans [44,45]. All studies mention a trend towards real-time imaging combined with AI to enhance clinical accuracy and patient outcomes.

3.4. AI Models

Properties of the AI models studied can be found in Table 2 and Table 3. The developed models primarily focused on classification, followed by segmentation and localization; see Figure 5. Six studies applied ML models for either classification (n = 5) or segmentation (n = 1) tasks. Fourteen studies applied DL models for classification (n = 12), segmentation (n = 1), or localization (n = 1) tasks.
Prominent DL architectures for classification were VGG-16, DenseNets, and ResNets. The predicted classes were compared to labels originating from (histo)pathology reports (n = 8), clinical diagnosis (n = 8), or expert annotations (n = 2). Four studies encompassed multiclass tasks where models predicted multiple malignancy risk-level descriptors [41,43,46,47]. The average reported classification accuracy was 0.89 (0.78–0.95) across binary models and 0.85 (0.78–0.96) across multiclass models (Table 2). The highest performing models combined either DenseNets [41] or ResNet-18 [47] with an attention module for final class prediction. Segmentation tasks were performed using a U-Net architecture in one out of two studies [43], with a reported Dice similarity coefficient (DSC) of 0.83 (Table 3). The localization task was performed on a global level. The results from segmentation and localization tasks were compared to ground truth manual annotation masks.

3.5. XAI Methods

In total, fourteen distinctive XAI methods were identified across the included studies. The main characteristics of each XAI method were cataloged and can be found in Table 4. The operational mechanisms of the identified XAI methods showed the following distribution:
  • Model: Specific (71.4%) vs. agnostic (28.6%).
  • Scope: Local (85.7%) vs. global scope (14.3%).
  • Explanation: Intrinsic (42.9%) vs. post hoc (57.1%).
Within the nine DL model-specific XAI methods, eight methods were specific to CNNs and one to Transformers. Two CNN-specific methods, BI-RADS-Net and MT-BI-RADS, implemented multitask learning (MTL) to improve AI-model classification and segmentation performance, respectively [43,52]. The XNML segmentation model, described by Martizzi et al. (2021), was the only XAI method that showed ML model-specific attributes and was tailored to their in-study developed PRISMTM platform [45]. The remaining four XAI methods were model-agnostic, solely operating on input and output, independent of the AI model architecture. The most often occurring explanations comprised a local scope and post hoc application; see Figure 6. The local scope indicated a focus on the explanation of specific patient cases. The post hoc applications of XAI indicated models used for explaining model outputs, for better comprehension of the decision-making process of AI models.

3.6. XAI Functions

The main XAI function implemented in the identified methods was visualization (n = 9), followed by semantics (n = 5) and example-based (n = 3); see Figure 7 and Figure 8. All three XAI methods with an example-based function were combined with additional semantics (n = 2) or visualization (n = 1) functions.

3.6.1. Visualization

In classification tasks, visualization provided insight into model behavior realized through backpropagation, perturbation, optimization, or feature attribution approaches.
Backpropagation-based approaches include saliency maps to visually represent the importance of different areas of the input image in the prediction process. These maps highlight the pixels with the highest impact on the predicted class output. These approaches provide model-specific, local-scope, post hoc explanations. Byra et al. (2022) and Tasnim et al. (2024) used class activation mapping (CAM), which replaces the fully connected layers at the end of CNNs with global average pooling layers to spatially aggregate the feature maps from the last convolutional layer [39,50]. The layer outputs are used to create a heat map, by weighting the class-specific spatial grid with the output weights connected to the class predictions. This heat map can thereafter be overlaid on the original image to show the discriminative regions used by the network to identify that class.
A generalized, gradient-weighted form of CAM, Grad-CAM, is used for classification tasks in six studies [37,41,47,49,53]. Grad-CAM is model-specific to CNNs and can be used without global average pooling, allowing the application to a wider variety of CNN architectures. Grad-CAM first computes the gradients of the target output concerning the feature maps of the last convolutional layer, which are global-average-pooled to obtain the importance weights for each feature map. These weights are multiplied with the forward activation maps to create the localization map and passed through a ReLU function to preserve the features that positively influence the class of interest. The result is an overlay of the produced heat map highlighting the most discriminative areas on the input image; see an example in Figure 9.
Perturbation-based approaches perturb the input image to assess the importance of image areas for the proposed task (e.g., classification). Local interpretable model-agnostic explanations (LIME) are used in Hassan et al. (2022) [42]. LIME provides a local explanation by replacing a complex model locally with simpler models, for example, by approximating a CNN by a linear model; see Figure 10. The input data are perturbed and the simplified model is used to learn the change in output. The similarity of the perturbed input to the original input is used as a weight, to ensure that explanations provided by the simple models with highly perturbed inputs have less effect on the final explanation. The simple model is trained on the dataset of perturbed samples using the prediction labels and the assigned weights. The simple model’s coefficients serve as the explanation for the instance. In the case of classification tasks, the coefficients reveal the contribution of each feature to the prediction of a specific class. Unlike CAM and Grad-CAM, LIME is model-agnostic and can be applied to any type of (ensemble) model.
Optimization-based approaches involve optimizing the input to see what features the network learns to recognize as belonging to a certain class, rather than observing changes in output due to perturbations or backpropagation of error. Tasnim et al. (2024) proposed activation maximization, a technique where the model’s parameters are adjusted to maximize the output activation of a specific class or feature [50]. The input image is iteratively adjusted until the pattern that most excites a particular neuron or layer is found. Activation maximizations allow the exploration of what each part of a neural network is looking for in its input.
Feature attribution approaches identify and quantify the contribution of individual features or regions in the input data to the output prediction. The region of evidence (ROE) method described by Dong et al. (2021) focuses on highlighting discriminative regions (ROEs) in ultrasound images that the CNN relies on for making class decisions, aligning with feature attribution through visualization [40]. The Explainer framework designed by Song et al. (2023) generates heat maps, or “E maps”, from the convolution of feature maps to highlight the region a CNN model uses for determining the output class [49]. These E maps, which have the same dimensions as the input feature maps, assign values between 0 and 1 to represent the importance of each pixel. Subsequently, these E maps are multiplied by the feature maps, with their resultant map used for the classification. This method produces salient mappings directly aligned with the model’s reasoning process, providing intrinsic explanations as it leverages the model’s internal architectures and computations to attribute features directly contributing to the output. Pixels deemed unimportant are assigned a value of 0, effectively excluding the features they represent from the classification. This approach contrasts with Grad-CAM, which identifies significant features considered by the model, whereas the Explainer focuses on the features the model actually uses to make predictions [49]. Although there may be overlap, these two sets of features are not identical. Additionally, the Explainer framework reshapes feature maps from different layers into the same size, allowing it to combine inputs from multiple layers rather than being limited to the last convolution layer.
In segmentation tasks, integrated visualization approaches empower visual evaluations of the model’s ability to separate different regions (tumor vs. healthy tissue) within the images. Integrated visualization techniques enhance explainability without perturbing the input or analyzing backpropagation gradients directly, as they inherently integrate visualization into the model’s architecture. The XNML method integrates data-centric learning and causal explanations directly into the learning process. It focuses on generating explainable ML predictions in medical anomaly detection by generating color-coded masks ranking lesion candidates [45]. In Karimzadeh et al. (2023), the developed MTL approach combined classification, segmentation, and quantitative explainability using semantic Shapley values [43]. Their MT-BI-RADS model outputs the predicted category of each BI-RADS risk assessment descriptor and provides visual explanations by highlighting tumor regions with image segmentation.
In localization tasks, object detection frameworks localize and highlight critical areas (lesions) in an image using backpropagation-based heat maps that allow for visual interpretation by experts. The activation heat map in Basu et al. (2023) was generated from global image features [38]. This method involves backpropagation of gradients from the output back to the input image, resulting in a map localizing salient regions in the images. The activation heat map is subjected to Otsu binarization to determine the highest activated region, and a bounding box is selected that covers this local region of interest (ROI); see Figure 11.

3.6.2. Semantics

XAI methods involving semantics aim to produce meaningful descriptions or labels that correlate closely with the features and decision-making criteria used by clinical experts. This approach enhances the explainability of AI systems by providing explanations that are not only technically accurate but are also understandable and clinically relevant. The identified semantics methods present feature-based or transparency-based approaches and are all applied in classification tasks.
Feature-based approaches emphasize individual or groups of features that substantially influence model predictions. These semantic explanations provide insights into the intrinsic decision-making processes of models by correlating computational assessments with clinically relevant features. The bag-of-features (BoF) method transforms complex image data into discrete features, known as “visual words”, simplifying the interpretation of transformer models’ decisions. The method segments images into local patches and identifies recurrent patterns, creating a vocabulary of visual words that represent clusters of similar patches. These visual words are then used to generate histograms that serve as input to classifiers, allowing models to make predictions based on the distribution of these words. This approach not only simplifies image data but also aligns closely with clinical interpretations, focusing on patterns and textures indicative of pathological conditions [38]. Developed by Zhang et al. (2021), BI-RADS-Net employs BI-RADS descriptors such as shape, margins, and densities as features to elucidate their impact on tumor malignancy classifications. This method leverages standardized clinical lexicons, enhancing the transparency and applicability of AI in clinical settings [52].
The SHAP (Shapley additive explanations) method quantifies the contribution of each input feature to model predictions using Shapley values, a concept from cooperative game theory, to ensure fair and consistent explanations across all possible combinations of feature inputs. SHAP has been applied in various studies to provide quantitative, post hoc explanations. In Karimzadeh et al., SHAP showed the contribution of each BI-RADS descriptor in MTL breast cancer classifications, offering insights into the significance of ultrasound image features like shape and margin [43]. Lombardi et al. (2023) utilized SHAP to identify key features in dermatoscopic images most indicative of malignancy or benignity, highlighting the diagnostic importance of specific visual patterns [44]. Rezazadeh et al. (2022) applied SHAP to an ensemble model for breast cancer diagnosis, detailing how statistical texture features impact malignant or benign class predictions [48], see Figure 12. These associations supported more informed clinical decisions.
Transparency-based approaches are designed to be inherently explainable. They may also enhance existing models with additional mechanisms that clarify their internal decision processes. These approaches ensure that the features learned by the model are clinically meaningful and do not just represent simulated correlations or artifacts of the training data. The image similarity technique (AIBx) in Thomas et al. (2020) integrated clinically significant patterns and semantic descriptors into AI models. The AI predictions were aligned with medical standards by embedding diagnostic categories, molecular markers, treatment responses, and recurrence status directly into the decision-making process [51]. The LTQ-E method enhances the local texture quantization (LTQ) model by using intermediate decision labels for added transparency. In Morris et al. (2023), LTQ-E extracted and aggregated embeddings from the penultimate layer of each separate decision-label model into a unified feature set. Hereafter, these collected features were used by linear or neural network model classifiers to predict thyroid-related TI-RADS-level labels [46]. Incorporating clinical knowledge into the computational explanation process aligns such methods with clinical reasoning steps to increase the models’ trustworthiness and transparency.

3.6.3. Example-Based

In XAI with example-based functions, methodologies provide insights and understanding about model decisions by highlighting similarities to previously encountered cases, specific features, or patterns that the model has learned are indicative of a particular class or output. Example-based methods allow clinicians to compare the AI’s findings in classification and segmentation tasks with decisive features or established cases, enhancing confidence in and acceptance of model predictions.
Feature attribution approaches included BI-RADS-Net for classification. BI-RADS-Net employs specific descriptors that align with terms derived from medical standards to explain class decisions [52]. Each descriptor acts as a feature example, illustrating how the model classifies breast ultrasound images based on standardized criteria.
Analogical reasoning approaches employ a case-based approach in AIBx and a counterfactual approach in XNML. In Thomas et al. (2020), AIBx actively involved operating clinicians in the decision process by, upon the input of an initial image for analysis, generating a collection of visually and clinically similar images based on previously confirmed cases, alongside an initial classification. Clinicians review these images to confirm or adjust the classification, where the image similarity mirrors clinicians’ memory and expertise to deepen the trust in the AI system [51]. The XNML model leveraged causal and counterfactual explanations to enhance ML models’ explainability. It linked statistical data with clinical outcomes, to provide rationales for each segmentation and to aid anomaly detection [45].

3.7. XAI Advantages and Disadvantages

Comparison of XAI methods reveals relative advantages and disadvantages across visualization, semantics, and example-based functions, highlighting the varied clinical relevance and potential impact of each method.
Visualization methods such as CAM and Grad-CAM provide direct and intuitive visual insights into influential areas within an image, particularly useful for tumor detection or classification tasks. These methods preserve high-resolution spatial information and are class-discriminative, making them suitable for differentiating between various conditions across a wide range of CNN architectures, without the need for architectural modifications. However, their requirement of a CNN architecture (with global average pooling) limits their broader application. This limited choice of algorithms could potentially exclude (neural) networks that could better fit the data. Additionally, saliency-based methods like Grad-CAM and LIME often rely on external validation to verify their accuracy. LIME uses simple models for local approximations, making its explanations easily understandable. The perturbation of the input shows the resultant change in the model output and can be used to explain which regions are driving the predictions directly. Despite being model-agnostic and providing specific, locally focused explanations, LIME may not generalize across different datasets. The method’s reliability heavily depends on how perturbations are implemented, and managing feature contributions in high-dimensional data can be challenging, necessitating careful interpretation to prevent misleading outcomes [42]. Clinically, visualization XAI methods could be implemented in US for tumor detection in various cancer types. This could result in a more thorough evaluation of the US images.
Semantic methods in XAI aim to bridge the gap between complex algorithmic image analysis and practical clinical interpretation by incorporating clinically relevant lexicons for higher confidence in class decisions. The bag of features (BoF) method concentrates on well-defined, localized features for detailed US image analysis. In Basu et al. (2023), BoF was used to identify features unknown to radiologists lexicons, but present in 78% of tumor-labeled cases. This feature could be added to the lexicon after its characterization is determined [38]. Other semantic methods, such as BI-RADS-Net, utilize BI-RADS descriptors to align class predictions more with clinical practice, effectively integrating the explanations into clinical workflows [52]. MT-BI-RADS expands upon this by providing both visual and semantic explanations of the significance of each BI-RADS descriptor in the decision-making processes. The Shapley values offer quantifiable insight into individual feature importance, explaining complex patterns in histopathological images and aiding clinical decision making [43]. Despite these advantages, the semantic methods identified were mainly developed for classification tasks, which could hinder their applications for segmentation or localization. Additionally, while methods like LTQ-E are effective in pattern extraction for TI-RADS labels at the image level, their complexity may potentially limit routine clinical use [46]. Semantic methods could be used to explain the AI models’ way of interpreting the difference between malignant and benign tumors in US images.
Example-based methods provide substantial educational and practical advantages in AI model validation and debugging by aligning model reasoning with clinical expert judgment. These methods are particularly beneficial in reducing human subjectivity, as demonstrated by the AIBx image similarity in Thomas et al. (2020), which reduced the number of unnecessary biopsies in clinical studies compared to decisions made without similarity support [51]. The task-specific design and use of familiar diagnostic categories for example-based XAI enhance user trust and integration into clinical systems. In addition, example-based methods can potentially serve an educational purpose, providing real-life case studies for less experienced radiologists or students. However, the effectiveness of these methods is limited by the availability and representativeness of example cases. A lack of clinical validation and reliance on high-quality, well-curated databases can hinder wider adoption. An example for the clinical use of example-based XAI models could be in thyroid nodule risk assessment. The XAI model can show similar-looking nodules to the one being assessed in real time to aid in decision making.
XAI methods that combine functions can leverage individual strengths of trustworthiness, interpretability, and operational efficiency while addressing limitations such as opacity, limited applicability, and clinical integration challenges. For example, in Karimzadeh et al. (2023), the MT-BI-RADS framework integrated tumor mask visualizations with quantitative SHAP values for breast cancer diagnosis, improving transparency and diagnostic confidence by aligning AI outputs with familiar BI-RADS categories [43]. This method addressed the limitations of opacity in traditional computer-aided diagnosis (CAD) systems by providing both intrinsic and post hoc insights. Additionally, the image similarity algorithm AIBx in Thomas et al. (2020) provided a valuable decision-support tool for breast cancer that enhanced diagnostic accuracy by correlating semantic image features with clinically relevant analogous examples [51]. Overall, the combined XAI approaches have the potential to improve accuracy, increase efficiency, and facilitate smoother integration of AI models into existing clinical workflows.

3.8. XAI Evaluation

The evaluation process of XAI methods was based on qualitative (QL) and/or quantitative (QT) metrics, as seen in Table 4, with four studies including both QL and QT evaluation metrics [43,44,49,50].

3.8.1. Qualitative

All but one XAI method using QL evaluation performed visual inspections of the XAI output, i.e., saliency maps and segmentation masks, to determine XAI method performance. Visual inspections generally consisted of examining AI-generated saliency maps overlaid on original images to ensure that the highlighted regions in the saliency maps corresponded to medically significant areas, qualitatively validating the model’s focus areas and their alignment with medical knowledge. In the study of Tasnim et al. (2024), the objective was to confirm that the CAMs correctly focused on lesion regions rather than the surrounding tissues to determine reliable diagnosis [50]. Some studies compared visual inspections to previous methods or manual segmentations. For example, Al-Jebrni et al. (2023) showed that their model’s Grad-CAMs consistently and accurately identified thyroid nodules, outperforming previous studies’ Grad-CAMs, which had scattered focus areas [37]. Hassan et al. (2022) compared LIME-generated explanations with radiologist-annotated regions containing malignancy indicators, confirming the model’s predictions [42]. The degree of improvement attributed to applying XAI was not explicitly expressed with a (qualitative) performance metric.
In most studies, clinician involvement within the visual inspections was not mentioned, with the evaluation primarily focused on the technical processes of generating and inspecting the XAI methods’ computational means. However, manual segmentations [39] or ROI annotations [38,42] by experienced radiologists during the dataset preparation phase indicated some clinical input in multiple studies. Formalized evaluations of XAI contributions with standardized questions or metrics were often lacking. Zheng et al. (2024) analyzed how the model’s attention shifted during different diagnostic stages, comparing this to the step-by-step approach of human experts [53]. Experts provided qualitative feedback on the Grad-CAM heat maps’ clarity and relevance, assessing whether these visual explanations improved their understanding of the model’s decisions and clinical decision making. Interpretability scores were given, although specific details on the scoring were not mentioned.

3.8.2. Quantitative

The QT metrics focus mainly on semantics and generally comprised standardized performance metrics in the field of AI-based image analysis. Classification performance is quantified by confusion matrix scores, e.g., accuracy, precision, sensitivity, specificity, F1-score, or area under the receiver operating characteristic curve (AUROC). Accuracy is the most common and provides a general indication of the proportion of correct predictions (true positives and true negatives) out of all class predictions made. Segmentation performance is quantified using DSC, determining the overlap between the predicted and ground truth mask segments. Scores range from 0 (no overlap) to 1 (perfect overlap), indicating the model’s success in delineating the tumor boundaries within the images. Localization performance is quantified with the common intersection over union (IoU) metric. The metric evaluates how precisely the tumors are localized within the image, considering both the accuracy of the boundary box and the class prediction. These standard metrics provide quantitative insights by statistically evaluating the AI model’s performance in intended tasks. They are essential for tuning the model, understanding its strengths and weaknesses, and comparing its performance with other AI models. However, these metrics primarily measure the performance of AI models itself and do not quantify the impact of XAI on improving model explainability in these evaluations.
A few studies involved clinicians during the XAI evaluation phase [40,44,47,49,53]. Song et al. (2023) conducted reader studies with nine physicians who used heat maps generated by their XAI Explainer method to assist in diagnosing 200 test set US images (100 malignant) [49]. The physicians initially diagnosed without XAI assistance to establish a baseline; thereafter, they used the heat maps to aid their decisions. Performance improvements were measured using accuracy, precision, recall, F1-score, and AUROC curves, showing higher performance with the Explainer visualizations, especially among junior physicians. In Qian et al. (2021), a two-part reader study with seven radiologists (14 years of experience on average) compared the performance of BI-RADS-Net with and without Grad-CAM heat maps. Reviewing 152 breast lesions (44 malignant) from 141 patients in the test set, the radiologists initially diagnosed without XAI assistance and then reassessed using the heat maps. Their study found that visualizations with heat maps improved sensitivity, specificity, and overall diagnostic accuracy, highlighting the technique’s added value in clinical practice [47].
Uncommon QT evaluation metrics for XAI methods were found in several studies:
  • Shapley values are derived from game theory and provide a method to distribute the impact among contributors (features) in a cooperative game (prediction model). Each feature value’s contribution is determined by assessing the change in prediction when a feature is added or removed for all possible combinations of features. The aim is to fairly attribute the model’s output to its input features, providing insights into which features are most important for predictions. In three studies, SHAP values were used to determine which clinical features (e.g., tumor size, shape, or texture) most significantly impact the models’ classification of a tumor as benign or malignant [43,44,48].
  • The zero-mean normalized cross-correlation (ZNCC) score presents a statistical measure to assess the similarity between two images. It calculates the degree of similarity between two images through a normalized cross-correlation measurement formula, subtracting their mean and dividing by their standard deviation. The ZNCC score ranges from −1 to 1, where 1 indicates perfect correlation, 0 indicates no correlation, and −1 indicates perfect inverse correlation. In Tasnim et al. (2024) the ZNCC score quantitatively assessed the feature separation ability of the activation maximization generated images in a benign-malignant (i.e., binary) classification problem [50].
  • The pointing game metric is a QT evaluation method used to assess how well the areas identified by saliency maps align with relevant regions in medical images. It evaluates whether the most significant activation points in the saliency map correspond to specific anatomical or pathological features in the analyzed images. Byra et al. (2022) used the pointing game to verify if the CAM saliency maps highlighted significant regions for accurate diagnosis, i.e., breast mass region, peritumoral region, or region below the breast mass [39]; see Figure 13.
  • The resemblance votes metric was used in Dong et al. (2021) for QT evaluation of how well the ROE identified by the AI aligns with the regions considered important by physicians for making diagnostic decisions [40]. The metric categorizes the ROE into three resemblance levels as perceived by clinicians: High resemblance (HR), where the ROE closely matches the features used by physicians; medium resemblance (MR), where the ROE partially matches; and low resemblance (LR), where there is little to no match. The contribution of clinicians in scoring the resemblance of AI predictions can aid in validating and improving the model’s explainability and utility in clinical practice.
Regarding the additional QT metrics identified, the ZNCC score primarily evaluates the AI model’s performance in terms of image similarity and feature activation. The evaluation contributes to understanding model behavior by illustrating AI model responses to variations in input; however, it does not directly assess the explainability. The pointing game specifically evaluates the explainability, as it measures the alignment between the model’s focus (saliency maps) and the clinically relevant areas in the images. Similarly, the resemblance votes method evaluates the explainability as it quantifies the correspondence between the ROE and the diagnostic areas identified by physicians, rather than the accuracy of the underlying AI system. Comparably, the Shapley values quantify the contribution of individual clinical features to the output. These explanations of which features the model uses to make a specific decision are as important as the decision itself.

4. Discussion

4.1. Key Findings

This systematic literature review aimed to investigate the use of explainable AI in oncological ultrasound image analysis, given the need for transparent and trustworthy AI systems in healthcare. Specifically, it sought to provide an overview of the identified XAI methods, including their operational mechanisms, explanatory function, and evaluation metrics. Additionally, this review aimed to compare the advantages and disadvantages of these XAI methods for clinical use.
Fourteen different XAI methods were identified. Most methods employed visualization functions that intuitively highlight influential image areas in prediction processes. However, these methods typically required specific architectures and depended on external validation, potentially limiting their broader application. Other methods incorporated semantic or example-based functions, aligning with clinical reasoning to reduce human subjectivity. Despite their advantages, semantic methods often face challenges in more complex tasks, and the effectiveness of example-based methods depends on the quality of examples used. Hybrid approaches that combine multiple explanatory functions have shown promise in leveraging the strengths and mitigating the limitations of individual methods, thus enhancing user trust and understanding.
Integrating XAI into oncological imaging, particularly in real-time applications, holds significant potential for enhancing analysis accuracy, surgical precision, and clinical decision making. For example, Song et al. (2023) showed performance improvements in US-based thyroid cancer diagnosis using their XAI Explainer method, particularly benefiting junior physicians [49]. In the study of Qian et al. (2021), breast lesion diagnosis aided by Grad-CAM heat map visualizations improved sensitivity, specificity, and overall diagnostic accuracy, highlighting the technique’s added value in clinical practice [47]. Zheng et al. (2024) demonstrated that XAI during live endoscopic US procedures can improve diagnostic accuracy by identifying discriminative tumor features in US images, supporting real-time surgical decisions and outcomes, particularly in distinguishing between visually similar tumors [53].
A significant gap identified in the current literature is the absence of standardized evaluation metrics that specifically quantify the impact of explanations provided by XAI methods in cancer-focused image analysis utilizing real-time ultrasound data. Current studies predominantly employ subjective assessments that rely on visual inspections to evaluate XAI methods. Researchers typically conduct these qualitative assessments without the involvement of clinical end-users. This approach limits the practical applicability and relevance of such evaluations. Some subjective assessments attempt to integrate quantitative metrics, such as the pointing game metric, which aligns network attention with clinical expectations to enhance trust in AI decisions. This metric is designed to elucidate why a model makes certain decisions, addressing the “black box” nature of models [39]. However, inconsistency in the application of these evaluations complicates comparisons across studies. The traditional quantitative evaluative metrics used (e.g., confusion matrix, DSC, IoU) are indispensable for objectively describing the accuracy and reliability of AI models, but they primarily focus on the AI model’s performance itself rather than the explainability aspects of XAI outputs. There is, thus, a need for metrics specifically designed to assess how transparent and interpretable AI models are to clinical end-users. Such metrics could include user studies or (subjective) clinical ratings of the transparency and effectiveness of explanatory tools. Currently, to our best knowledge, no comprehensive benchmark exists that specifically addresses XAI evaluation for medical image analysis. Although a pioneering evaluative benchmark designed for XAI visualization in general image analysis tasks is noted [54], it lacks a focus on medical or cancer applications and does not consider semantic or example-based XAI.
Moreover, evaluations often miss objective judgments and feedback from clinical end-users, which are crucial components for practical implementation. Studies using tailored quantitative metrics such as ZNCC scores focus more on technical validation, while methods involving clinicians ensure that AI aligns with clinical reasoning, stimulating better integration [38]. Only a few studies actively incorporated clinicians in their XAI evaluations [40,44,46,47,49,53]. Dong et al. (2021) actively involved physicians in assessing the resemblance levels of the ROE to ensure that AI-generated evidence was clinically relevant and aligned with their diagnostic criteria [40]. Additionally, three studies [44,46,49] iteratively consulted clinicians in the development of the XAI, to ensure that explanations were clinically relevant and useful. Clinicians participated in usability testing sessions to evaluate the effectiveness and clarity of the explanations provided by the XAI systems. Thereafter, their feedback was used to refine the explanations to match clinical needs better [46,49]. Lombardi et al. (2023) specifically employed a human-centered design to involve healthcare professionals throughout the design, implementation, and evaluation phases of the proposed XAI pipeline [44]. The remaining two studies solely incorporated radiologists [47] or endoscopists [53] to qualitatively assess models’ performance in providing understandable visual outputs.

4.2. Comparison with the Existing Literature

Despite the broad interest in XAI method development, it remains challenging to compare different evaluations due to the absence of standardized evaluation metrics. This need for a common framework of metrics that can be applied universally is repeatedly concluded in the literature [12,16,55]. Similar to our findings, Abrantes et al. (2024) mention a focus on visual and post hoc explanations, pointing out the unclear impact these explanations have on clinical outcomes. Additionally, they note the risk of overreliance on the model when providing users with insubstantial explanations, potentially even reducing decision performance. They emphasize the importance of improving AI’s operational transparency in future XAI developments [12]. Similarly, Antoniadi et al. (2021) observed that XAI could boost clinicians’ confidence and generate causal hypotheses in clinical decision support systems, increasing the systems’ trustworthiness and acceptability. However, they mention a distinct shortage in XAI evaluations, particularly in user studies that address clinician needs [16]. In support, Amann et al. (2020) argue that neglecting explainability in clinical decision support systems poses a threat to individual and public health [55]. The variability and scarcity of objective evaluations complicate effective comparisons of XAI methods’ impacts. Developing standardized, clinically relevant evaluation metrics for XAI is needed to assess both the technical performance and explainability of AI models.

4.3. Limitations

The results of this review should be interpreted while taking several limitations into account. No articles were identified that applied XAI for intraoperative margin assessments.
The ambiguous taxonomy of XAI might have resulted in missed studies during our search strategy, despite efforts to create a broad search string to minimize this risk. Future research could expand on this by using broader search terms or additional databases.
The included studies varied significantly in design and outcome measures, which complicates direct comparisons. This heterogeneity should be considered when interpreting the findings.
The findings of this review are limited to the cancer types covered in the included studies, primarily breast and thyroid cancers. Further research is required to validate the use of XAI in ultrasound imaging for other cancer types. The proposed XAI methods might need significant modifications to handle new data and applications. This limits their generalizability across various clinical settings. Furthermore, many XAI methods have not been evaluated in real clinical settings, limiting their practical adoption. Moreover, feedback from end-users is absent in most of the articles regarding XAI. This, together with the minimal clinical assessment of these models, poses a significant shortcoming in the development and evaluation of XAI methods. The lack of discussion on computational time in the studies is also concerning. Many existing XAI methods use parts of the data or derivatives of real-time images. Time is an essential feature in real-time environments. Consequently, our conclusions regarding the expected clinical potential and impact of XAI in cancer-based image analysis remain uncertain. Furthermore, these models are also limited by the US hardware. Even though most of the recently manufactured US devices allow for AI integration, the computational time should be calculated using the same specifications as the end product. Additionally, XAI methods could help identify and address biases in AI predictions, but most of the reviewed studies did not explicitly focus on bias mitigation. Ensuring fairness in AI predictions is crucial, particularly in diverse patient populations. Future XAI methods should focus on developing approaches that can handle dynamic and complex image data, e.g., for better integration into oncological surgery.

4.4. Strengths

This systematic review has several strengths regarding comprehensiveness and relevance. The identification of 14 methods and their categorization into three main XAI functions (visualization, semantics, example-based) aligns with the existing literature [14,32]. The extracted characteristics of XAI methods show a similar distribution to those in the study by Van der Velden et al. (2022), which surveyed over 200 papers on XAI for DL-based medical image analysis. This study found that most XAI methods provided CNN-specific, local, post hoc explanations using saliency maps [32]. Reflecting these findings reinforces the relevance of the categorized XAI methods.
Another notable strength of this review is the provided overview comparing XAI methods for cancer-focused image analysis tasks, encompassing classification, segmentation, and localization. To the best of our knowledge, this is the first review to specifically address XAI for real-time ultrasound imaging in cancer applications.
Additionally, this review underscores the need for standardized and widely accepted evaluative metrics in XAI, which are currently missing. It reinforces the need for clinical end-user integration in developing and evaluating XAI methods, consistent with previous research [12,16,55]. Addressing existing gaps and aligning with the established literature, this review provides a foundation for future advancements in XAI for cancer-focused image analysis.

4.5. Implications and Future Research

The findings in this review have several implications for the current research, development, and clinical implementation of XAI. XAI has shown the potential to enhance clinician confidence and act as a second reader, improving clinical accuracy and decision making. However, while explainability can improve transparency, it can lead to overreliance on AI systems. This is problematic when models are uncertain or incorrect, potentially reducing the decision performance [12]. Therefore, it is necessary to integrate clinical end-users in the development and evaluation processes, focusing on the quality and utility of the explanations provided. Future research should prioritize this by creating “explainability for end-users”. Interactive explanations, where end-users can refine and adjust explanations, should be explored. Additionally. the traditional XAI methods like LIME or SHAP focus on feature attributions, which may not align with human conceptual reasoning. Future research should explore concept-based XAI methods, as proposed by Longo et al. (2024) [21]. These explanations embed human-understandable concepts into AI outputs, facilitating the integration of expert clinical knowledge. These methods should be developed for tasks beyond computer vision applications, such as classification and regression in oncological contexts. Initial examples can already be seen in the BoF [38] and BI-RADS-Net [52] methods.
XAI methods combining functions and scopes can provide rich explanations that align more closely with human cognitive processes [21]. Such hybrid methods ensure technical accuracy whilst enhancing transparency and user trust, particularly important in critical care areas such as oncology. However, many XAI methods identified present generalization issues across different datasets or conditions, indicating the need for further refinement of XAI to ensure robustness and sophistication. Policymakers should consider the balance between the benefits of explainability and the potential monetary and safety costs [15].
Future research should address identified shortcomings to advance the application and effectiveness of XAI in cancer-focused ultrasound image analysis. These include the need for standardized evaluation metrics and the development of an evaluation framework that addresses the clinically relevant impact of the explanations provided. Such a framework should ensure that XAI methods are consistently and quantitatively evaluated for increased reliability and comparability.
Subsequently, various other studies should be performed to really grasp the impact of XAI in a clinical setting for US imaging. It is of interest to understand how XAI aids clinical decision making or changes treatment plans compared to conventional methods. The cost of implementing XAI in clinical practice is not well documented in current studies. A cost–benefit analysis would be valuable for hospitals considering adoption, weighing potential improvements in diagnosis accuracy against financial costs. Another important topic that requires further research is the user-friendliness of the output of XAI models, especially focusing on the interobserver variability of the end-users and whether the XAI output is rational, instead of derived from biases. Lastly, given the gaps in demographic data, particularly concerning sex and ethnicity, the generalizability of these studies to the broader population may be limited. Future research should prioritize comprehensive reporting of patient demographics to enhance the applicability and relevance of study findings.

5. Conclusions

In conclusion, this review provides an overview of various XAI methods applied to cancer-focused image analysis using real-time ultrasound imaging data, aimed at addressing the “black box” nature of advanced AI models. The enhanced transparency resulting from XAI implementations can help clinicians make more informed decisions, foster trust, and facilitate the integration of AI into clinical workflows. The identified XAI methods show promise, particularly when combining functions and actively incorporating clinical end-users. However, challenges remain regarding the XAI method’s novelty, ambiguous evaluations, and data dependency. Addressing the identified gaps and proposing methodological improvements will be essential for promoting the practical implementation and clinical efficacy of XAI. The expansion of XAI towards real-time cancer applications and developing standardized quantitative evaluative metrics are crucial steps in this advancement.

Author Contributions

Conceptualization, F.G. and B.D.; methodology, L.S.W., L.M.v.K., M.W., F.G. and B.D.; software, L.S.W.; validation, L.S.W., L.M.v.K., M.W., F.G. and B.D.; formal analysis, L.S.W., F.G. and B.D.; investigation, L.S.W. and F.G.; resources, B.D.; data curation, L.S.W. and F.G.; writing—original draft preparation, L.S.W. and F.G.; writing—review and editing, L.S.W., L.M.v.K., M.W., F.G. and B.D.; visualization, L.S.W. and F.G.; supervision, F.G. and B.D.; project administration, B.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Research at the Netherlands Cancer Institute is supported by institutional grants of the Dutch Cancer Society and of the Dutch Ministry of Health, Welfare and Sport.

Conflicts of Interest

The authors declare no conflicts of interest.

Appendix A

Medline search string:
  • ((((explainab*) ADJ12 (artificial-intelligen* OR AI OR deep-learn* OR machine-learn* OR neural-network* OR machine-intellig* OR automated-reason* OR machine-reason*)) OR XAI).ab,ti,kf.) AND (exp Ultrasonography/ OR Ultrasonics/ OR (ultrasound* OR ultrasonic OR echograph* OR ultrasonograph*).ab,ti,kf.) AND (Classification/ OR exp Neoplasm Staging/ OR Margins of Excision/ OR (segmentat* or classificat* OR margin* OR delineation* OR staging).ab,ti,kf.)
Scopus search string:
  • TITLE-ABS-KEY(((((explainab*) W/12 (artificial-intelligen* OR AI OR deep-learn* OR machine-learn* OR neural-network* OR machine-intellig* OR automated-reason* OR machine-reason*)) OR XAI)) AND ((ultrasound* OR ultrasonic OR echograph* OR ultrasonograph*)) AND ((segmentat* or classificat* OR margin* OR delineation* OR staging)))

Appendix B

Table A1. Characteristics of ultrasound imaging systems utilized per study.
Table A1. Characteristics of ultrasound imaging systems utilized per study.
Author (Year)US ModalitySystem (Manufacturer)Probe/TransducerFrequencyAcquisition
Al-Jebrni, A.H. (2023) [37]USEPIQ 7 (Philips) + DU8 (Technos)NANAContinuous
Basu, S. (2023) [38]Transabdominal USLogic S8 (GE Healthcare)Convex low-frequency1.0–5.0 MHzMultiple shot, static B-mode
Byra, M. (2022) [39]Breast USSonixTouch Research (Ultrasonix)L14-5/38 linear probe10 MHzSingle shot, static B-mode
Dong, F. (2021) [40]USResona 7 (Mindray)L11-3U linear array2.0–11 MHzContinuous
Han, X. (2022) [41]USNA (Siemens), NA (Philips)NANAContinuous
Hassan, M. R. (2022) [42]Transrectal USHi-Vision 5500 (Hitachi)NA7.5 MHzContinuous
C41V end-fire probe (Nobulus)NA2.0–10 MHzContinuous
Karimzadeh, M. (2023) [43]Breast USLOGIQ E9 + LOGIQ E9 Agile (GE Healthcare)ML6-15-D Matrix linear probeNAContinuous
VIVID 7 (GE Healthcare), LOGIQ E9 (GE Healthcare), EUB-6500 (Hitachi), iU22 (Philips), ACUSON S2000 (Siemens)NANAContinuous
Lombardi, A. (2023) [44]Transvaginal or transabdominal US & 3D USNANA5.0–9.0 MHz (transvaginal), 3.5–5.0 MHz (transabdominal)Continuous or 3D volume scan
Martizzi, D. (2021) [45]Quantitative Transmission USNANANA3D volume scan
Morris, J. (2023) [46]USNANANAContinuous
Qian, X. (2021) [47]USAixplorer (SuperSonic)NANAContinuous
Rezazadeh, A. (2022) [48]Breast USLOGIQ E9 (GE Healthcare) + LOGIQ E9 Agile (GE Healthcare)ML6-15-D Matrix linear probe1.0–5.0 MHzContinuous
Song, D. (2023) [49]USNANANASingle shot
Tasnim, J. (2024) [50]Quantitative USLOGIQ E9 + LOGIQ E9 Agile (GE Healthcare)ML6-15-D Matrix linear probe1.0–5.0 MHzContinuous
Voluson730 scanner (GE Healthcare)S-VNW5-10 small-part transducer5.0–10 MHzContinuous
ACUSON Sequoia C512 (Siemens)17L5 HD linear array transducer8.5 MHzContinuous
iU22 (Philips)Linear probe7.0–15 MHzContinuous
Sonix-Touch Research (Ultrasonix)L14-5/38 linear transducer10 MHzContinuous
Thomas, J. (2020) [51]USNA (GE Healthcare), NA (Philips), NA (Sonosite)NA8.0–13 MHzContinuous
Zhang, B. (2021) [52]Breast USLOGIQ E9 + LOGIQ E9 Agile (GE Healthcare)ML6-15-D Matrix linear probe1.0–5.0 MHzContinuous
VIVID 7 (GE Healthcare), LOGIQ E9 (GE Healthcare), EUB-6500 (Hitachi), iU22 (Philips), ACUSON S2000 (Siemens)L11-3U linear array1.0–5.0 MHzContinuous
Zheng, H. (2024) [53]Endoscopic USNANANAContinuous
NA = uot available, US = ultrasound.

References

  1. Grand View Research. AI In Healthcare Market Size, Share & Trends Analysis Report by Component (Hardware, Services), by Application, by End-Use, by Technology, by Region, and Segment Forecasts, 2024–2030. Available online: https://www.grandviewresearch.com/industry-analysis/artificial-intelligence-ai-healthcare-market (accessed on 1 July 2024).
  2. Sanskrutisathe. AI in Healthcare Market Size and Growth. Available online: https://medium.com/@sanskrutisathe01/ai-in-healthcare-market-size-and-growth-2ae9b8463121 (accessed on 3 September 2024).
  3. World Health Organization. Global Strategy on Human Resources for Health: Workforce 2030. Available online: https://apps.who.int/iris/bitstream/handle/10665/250368/9789241511131-eng.pdf (accessed on 3 September 2024).
  4. Choi, M.; Sempungu, J.K.; Lee, E.H.; Lee, Y.H. Living longer but in poor health: Healthcare system responses to ageing populations in industrialised countries based on the Findings from the Global Burden of Disease Study 2019. BMC Public Health 2024, 24, 576. [Google Scholar] [CrossRef]
  5. Atkinson, S.; Jackson, C. Three in Five Globally Say Their Healthcare System Is Overstretched. Available online: https://www.ipsos.com/en/three-five-globally-say-their-healthcare-system-overstretched (accessed on 3 September 2024).
  6. Page, B.; Irving, D.; Amalberti, R.; Vincent, C. Health services under pressure: A scoping review and development of a taxonomy of adaptive strategies. BMJ Qual. Saf. 2023. [Google Scholar] [CrossRef]
  7. Bohr, A.; Memarzadeh, K. Chapter 2—The rise of artificial intelligence in healthcare applications. In Artificial Intelligence in Healthcare; Academic Press: Cambridge, MA, USA, 2020; pp. 25–60. [Google Scholar] [CrossRef]
  8. Zhang, B.; Shi, H.; Wang, H. Machine Learning and AI in Cancer Prognosis, Prediction, and Treatment Selection: A Critical Approach. J. Multidiscip. Healthc. 2023, 16, 1779–1791. [Google Scholar] [CrossRef] [PubMed]
  9. Wolff, J.; Pauling, J.; Keck, A.; Baumbach, J. The Economic Impact of Artificial Intelligence in Health Care: Systematic Review. J. Med. Internet. Res. 2020, 22, e16866. [Google Scholar] [CrossRef] [PubMed]
  10. Pinto-Coelho, L. How Artificial Intelligence Is Shaping Medical Imaging Technology: A Survey of Innovations and Applications. Bioengineering 2023, 10, 1435. [Google Scholar] [CrossRef] [PubMed]
  11. Borys, K.; Schmitt, Y.A.; Nauta, M.; Seifert, C.; Krämer, N.; Friedrich, C.M.; Nensa, F. Explainable AI in medical imaging: An overview for clinical practitioners—Saliency-based XAI approaches. Eur. J. Radiol. 2023, 162, 110787. [Google Scholar] [CrossRef]
  12. Abrantes, J.; Rouzrokh, P. Explaining explainability: The role of XAI in medical imaging. Eur. J. Radiol. 2024, 173, 111389. [Google Scholar] [CrossRef]
  13. Singh, A.; Sengupta, S.; Lakshminarayanan, V. Explainable Deep Learning Models in Medical Image Analysis. J. Imaging 2020, 6, 52. [Google Scholar] [CrossRef]
  14. Reyes, M.; Meier, R.; Pereira, S.; Silva, C.A.; Dahlweid, F.M.; Tengg-Kobligk, H.v.; Summers, R.M.; Wiest, R. On the Interpretability of Artificial Intelligence in Radiology: Challenges and Opportunities. Radiol. Artif. Intell. 2020, 2, e190043. [Google Scholar] [CrossRef] [PubMed]
  15. Hacker, P.; Passoth, J.H. Varieties of AI Explanations under the Law. From the GDPR to the AIA, and Beyond. In xxAI—Beyond Explainable AI, Lecture Notes in Artificial Intelligence; Springer International Publishing: Berlin/Heidelberg, Germany, 2022; pp. 343–373. [Google Scholar] [CrossRef]
  16. Antoniadi, A.M.; Du, Y.; Guendouz, Y.; Wei, L.; Mazo, C.; Becker, B.A.; Mooney, C. Current Challenges and Future Opportunities for XAI in Machine Learning-Based Clinical Decision Support Systems: A Systematic Review. Appl. Sci. 2021, 11, 5088. [Google Scholar] [CrossRef]
  17. Barredo Arrieta, A.; Díaz-Rodríguez, N.; Del Ser, J.; Bennetot, A.; Tabik, S.; Barbado, A.; Garcia, S.; Gil-Lopez, S.; Molina, D.; Benjamins, R.; et al. Explainable Artificial Intelligence (XAI): Concepts, taxonomies, opportunities and challenges toward responsible AI. Inf. Fusion 2020, 58, 82–115. [Google Scholar] [CrossRef]
  18. Linardatos, P.; Papastefanopoulos, V.; Kotsiantis, S. Explainable AI: A Review of Machine Learning Interpretability Methods. Entropy 2020, 23, 18. [Google Scholar] [CrossRef] [PubMed]
  19. Next Move Strategy Consulting Explainable AI (XAI) Market. Available online: https://www.nextmsc.com/report/explainable-ai-market (accessed on 1 July 2024).
  20. Jacovi, A. Trends in Explainable AI (XAI) Literature. Available online: https://medium.com/@alonjacovi/trends-in-explainable-ai-xai-literature-a1db485e871 (accessed on 1 July 2024).
  21. Longo, L.; Brcic, M.; Cabitza, F.; Choi, J.; Confalonieri, R.; Ser, J.D.; Guidotti, R.; Hayashi, Y.; Herrera, F.; Holzinger, A.; et al. Explainable Artificial Intelligence (XAI) 2.0: A manifesto of open challenges and interdisciplinary research directions. Inf. Fusion 2024, 106, 102301. [Google Scholar] [CrossRef]
  22. Ali, S.; Abuhmed, T.; El-Sappagh, S.; Muhammad, K.; Alonso-Moral, J.M.; Confalonieri, R.; Guidotti, R.; Del Ser, J.; Díaz-Rodríguez, N.; Herrera, F. Explainable Artificial Intelligence (XAI): What we know and what is left to attain Trustworthy Artificial Intelligence. Inf. Fusion 2023, 99, 101805. [Google Scholar] [CrossRef]
  23. World Health Organization. Global Cancer Burden Growing, Amidst Mounting Need for Services. Available online: https://www.who.int/news/item/01-02-2024-global-cancer-burden-growing--amidst-mounting-need-for-services (accessed on 3 September 2024).
  24. Weerarathna, I.N.; Kamble, A.R.; Luharia, A. Artificial Intelligence Applications for Biomedical Cancer Research: A Review. Cureus 2023, 15, e48307. [Google Scholar] [CrossRef]
  25. Jaber, N. Can Artificial Intelligence Help See Cancer in New, and Better, Ways? National Cancer Institute: Mount Warrigal, NSW, Australia, 2022. [Google Scholar]
  26. Wijkhuizen, M.; van Karnenbeek, L.; Geldof, F.; Ruers, T.J.; Dashtbozorg, B. Ultrasound tumor detection using an adapted Mask-RCNN with a continuous objectness score. In Medical Imaging with Deep Learning; 2024; Available online: https://openreview.net/forum?id=IHmvNgX34A (accessed on 3 September 2024).
  27. Natali, T.; Wijkhuizen, M.; Kurucz, L.; Fusaglia, M.; van Leeuwen, P.J.; Ruers, T.J.; Dashtbozorg, B. Automatic real-time prostate detection in transabdominal ultrasound images. In Medical Imaging with Deep Learning; 2024; Available online: https://openreview.net/forum?id=jh1KZ4cyWA (accessed on 3 September 2024).
  28. Hoogteijling, N.; Veluponnar, D.; de Boer, L.; Dashtbozorg, B.; Peeters, M.J.V.; van Duijnhoven, F.; Ruers, T. Toward automatic surgical margin assessment using ultrasound imaging during breast cancer surgery. Eur. J. Surg. Oncol. 2023, 49, e108–e109. [Google Scholar] [CrossRef]
  29. Veluponnar, D.; de Boer, L.L.; Geldof, F.; Jong, L.J.S.; Da Silva Guimaraes, M.; Vrancken Peeters, M.J.T.; van Duijnhoven, F.; Ruers, T.; Dashtbozorg, B. Toward intraoperative margin assessment using a deep learning-based approach for automatic tumor segmentation in breast lumpectomy ultrasound images. Cancers 2023, 15, 1652. [Google Scholar] [CrossRef]
  30. Geldof, F.; Pruijssers, C.W.; Jong, L.J.S.; Veluponnar, D.; Ruers, T.J.; Dashtbozorg, B. Tumor Segmentation in Colorectal Ultrasound Images Using an Ensemble Transfer Learning Model: Towards Intra-Operative Margin Assessment. Diagnostics 2023, 13, 3595. [Google Scholar] [CrossRef]
  31. Weld, A.; Dixon, L.; Anichini, G.; Dyck, M.; Ranne, A.; Camp, S.; Giannarou, S. Identifying Visible Tissue in Intraoperative Ultrasound Images during Brain Surgery: A Method and Application. arXiv 2023, arXiv:2306.01190. [Google Scholar] [CrossRef]
  32. van der Velden, B.H.; Kuijf, H.J.; Gilhuijs, K.G.; Viergever, M.A. Explainable artificial intelligence (XAI) in deep learning-based medical image analysis. Med Image Anal. 2022, 79, 102470. [Google Scholar] [CrossRef]
  33. Medline Embase Database. Available online: https://www.embase.com (accessed on 3 September 2024).
  34. Scopus Bibliographic Database. Available online: https://www.scopus.com (accessed on 3 September 2024).
  35. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Syst. Rev. 2021, 10, 89. [Google Scholar] [CrossRef] [PubMed]
  36. Visani, G. Explainable Machine Learning, XAI Review: Model Agnostic Tools. Available online: https://towardsdatascience.com/explainable-machine-learning-9d1ca0547ae0 (accessed on 3 September 2024).
  37. Al-Jebrni, A.H.; Ali, S.G.; Li, H.; Lin, X.; Li, P.; Jung, Y.; Kim, J.; Feng, D.D.; Sheng, B.; Jiang, L.; et al. SThy-Net: A feature fusion-enhanced dense-branched modules network for small thyroid nodule classification from ultrasound images. Vis. Comput. 2023, 39, 3675–3689. [Google Scholar] [CrossRef]
  38. Basu, S.; Gupta, M.; Rana, P.; Gupta, P.; Arora, C. RadFormer: Transformers with global–local attention for interpretable and accurate Gallbladder Cancer detection. Med Image Anal. 2023, 83, 102676. [Google Scholar] [CrossRef] [PubMed]
  39. Byra, M.; Dobruch-Sobczak, K.; Piotrzkowska-Wroblewska, H.; Klimonda, Z.; Litniewski, J. Explaining a Deep Learning Based Breast Ultrasound Image Classifier with Saliency Maps. J. Ultrason. 2022, 22, 70–75. [Google Scholar] [CrossRef] [PubMed]
  40. Dong, F.; She, R.; Cui, C.; Shi, S.; Hu, X.; Zeng, J.; Wu, H.; Xu, J.; Zhang, Y. One step further into the blackbox: A pilot study of how to build more confidence around an AI-based decision system of breast nodule assessment in 2D ultrasound. Eur. Radiol. 2021, 31, 4991–5000. [Google Scholar] [CrossRef]
  41. Han, X.; Chang, L.; Song, K.; Cheng, L.; Li, M.; Wei, X. Multitask network for thyroid nodule diagnosis based on TI-RADS. Med Phys. 2022, 49, 5064–5080. [Google Scholar] [CrossRef]
  42. Hassan, M.R.; Islam, M.F.; Uddin, M.Z.; Ghoshal, G.; Hassan, M.M.; Huda, S.; Fortino, G. Prostate cancer classification from ultrasound and MRI images using deep learning based Explainable Artificial Intelligence. Future Gener. Comput. Syst. 2022, 127, 462–472. [Google Scholar] [CrossRef]
  43. Karimzadeh, M.; Vakanski, A.; Xian, M.; Zhang, B. Post-Hoc Explainability of BI-RADS Descriptors in a Multi-Task Framework for Breast Cancer Detection and Segmentation. In Proceedings of the 2023 IEEE 33rd International Workshop on Machine Learning for Signal Processing (MLSP), Rome, Italy, 17–20 September 2023; IEEE Computer Society: Washington, DC, USA, 2023. [Google Scholar] [CrossRef]
  44. Lombardi, A.; Arezzo, F.; Sciascio, E.D.; Ardito, C.; Mongelli, M.; Lillo, N.D.; Fascilla, F.D.; Silvestris, E.; Kardhashi, A.; Putino, C.; et al. A human-interpretable machine learning pipeline based on ultrasound to support leiomyosarcoma diagnosis. Artif. Intell. Med. 2023, 146, 102697. [Google Scholar] [CrossRef]
  45. Martizzi, D.; Huang, Y.; Malik, B.; Ray, P.D. Breast mass detection and classification using PRISM™ eXplainable Network based Machine Learning (XNML™) platform for Quantitative Transmission (QT) ultrasound tomography. In Proceedings of the Medical Imaging 2021: Ultrasonic Imaging and Tomography, Online, 15–19 February 2021; Volume 11602. [Google Scholar] [CrossRef]
  46. Morris, J.; Liu, Z.; Liang, H.; Nagala, S.; Hong, X. ThyExp: An explainable AI-assisted Decision Making Toolkit for Thyroid Nodule Diagnosis based on Ultra-sound Images. In Proceedings of the 32nd ACM International Conference on Information and Knowledge Management, Birmingham, UK, 21–25 October 2023; Association for Computing Machiner: New York, NY, USA, 2023; pp. 5371–5375. [Google Scholar] [CrossRef]
  47. Qian, X.; Pei, J.; Zheng, H.; Xie, X.; Yan, L.; Zhang, H.; Han, C.; Gao, X.; Zhang, H.; Zheng, W.; et al. Prospective assessment of breast cancer risk from multimodal multiview ultrasound images via clinically applicable deep learning. Nat. Biomed. Eng. 2021, 5, 522–532. [Google Scholar] [CrossRef]
  48. Rezazadeh, A.; Jafarian, Y.; Kord, A. Explainable Ensemble Machine Learning for Breast Cancer Diagnosis Based on Ultrasound Image Texture Features. Forecasting 2022, 4, 262–274. [Google Scholar] [CrossRef]
  49. Song, D.; Yao, J.; Jiang, Y.; Shi, S.; Cui, C.; Wang, L.; Wang, L.; Wu, H.; Tian, H.; Ye, X.; et al. A new xAI framework with feature explainability for tumors decision-making in Ultrasound data: Comparing with Grad-CAM. Comput. Methods Programs Biomed. 2023, 235, 107527. [Google Scholar] [CrossRef] [PubMed]
  50. Tasnim, J.; Hasan, M.K. CAM-QUS guided self-tuning modular CNNs with multi-loss functions for fully automated breast lesion classification in ultrasound images. Phys. Med. Biol. 2024, 69, 015018. [Google Scholar] [CrossRef]
  51. Thomas, J.; Haertling, T. AIBx, Artificial Intelligence Model to Risk Stratify Thyroid Nodules. Thyroid 2020, 30, 878–884. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, B.; Vakanski, A.; Xian, M. Bi-Rads-Net: An Explainable Multitask Learning Approach for Cancer Diagnosis in Breast Ultrasound Images. In Proceedings of the 2021 IEEE 31st International Workshop on Machine Learning for Signal Processing (MLSP), Gold Coast, QLD, Australia, 25–28 October 2021. [Google Scholar] [CrossRef]
  53. Zheng, H.; Dong, Z.; Liu, T.; Zheng, H.; Wan, X.; Bao, J. Enhancing gastrointestinal submucosal tumor recognition in endoscopic ultrasonography: A novel multi-attribute guided contextual attention network. Expert Syst. Appl. 2024, 242, 122725. [Google Scholar] [CrossRef]
  54. Zhang, Y.; Gu, S.; Song, J.; Pan, B.; Bai, G.; Zhao, L. XAI Benchmark for Visual Explanation. arXiv 2023, arXiv:2310.08537. [Google Scholar] [CrossRef]
  55. Amann, J.; Blasimme, A.; Vayena, E.; Frey, D.; Madai, V.I. Explainability for artificial intelligence in healthcare: A multidisciplinary perspective. BMC Med Inform. Decis. Mak. 2020, 20, 310. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of XAI methods with model-specific (left) or model-agnostic (right) dependencies.
Figure 1. Schematic representation of XAI methods with model-specific (left) or model-agnostic (right) dependencies.
Applsci 14 08108 g001
Figure 2. Schematic representation of XAI methods with global (left) or local (right) scopes.
Figure 2. Schematic representation of XAI methods with global (left) or local (right) scopes.
Applsci 14 08108 g002
Figure 3. Schematic representation of XAI methods with intrinsic (bottom) or post hoc (top) applications.
Figure 3. Schematic representation of XAI methods with intrinsic (bottom) or post hoc (top) applications.
Applsci 14 08108 g003
Figure 4. Flowchart visualizing the results of the PRISMA-based article selection process.
Figure 4. Flowchart visualizing the results of the PRISMA-based article selection process.
Applsci 14 08108 g004
Figure 5. Division of AI-based image analysis tasks in the included studies.
Figure 5. Division of AI-based image analysis tasks in the included studies.
Applsci 14 08108 g005
Figure 6. Frequency of various compositions of XAI methods’ scope and application in the included studies, categorized by function. Note that some XAI methods served multiple functions and were used in multiple studies; hence, the total counts in this figure exceed the number of studies and XAI methods listed previously.
Figure 6. Frequency of various compositions of XAI methods’ scope and application in the included studies, categorized by function. Note that some XAI methods served multiple functions and were used in multiple studies; hence, the total counts in this figure exceed the number of studies and XAI methods listed previously.
Applsci 14 08108 g006
Figure 7. Division of XAI method functions.
Figure 7. Division of XAI method functions.
Applsci 14 08108 g007
Figure 8. Frequency of identified XAI functions in the included studies, categorized by image analysis task. Note that some XAI methods served multiple functions and were used in multiple studies; hence, the total counts in this figure exceed the number of studies and XAI methods listed previously.
Figure 8. Frequency of identified XAI functions in the included studies, categorized by image analysis task. Note that some XAI methods served multiple functions and were used in multiple studies; hence, the total counts in this figure exceed the number of studies and XAI methods listed previously.
Applsci 14 08108 g008
Figure 9. Visualization examples for GIST, leiomyoma, and pancreatic rest tumors with the Grad-CAM plots generated by different methods reflecting the decision basis of different models. The first column presents the original US image. The 2nd column shows the expert annotations. The 3rd–6th columns present the generated Grad-CAM saliency maps using a baseline model, a multiattribute guided network (MAG), a contextual attention network (CA), and a combined MAG–CA network. Adapted from Zheng et al. (2024) [53], with permission from Elsevier.
Figure 9. Visualization examples for GIST, leiomyoma, and pancreatic rest tumors with the Grad-CAM plots generated by different methods reflecting the decision basis of different models. The first column presents the original US image. The 2nd column shows the expert annotations. The 3rd–6th columns present the generated Grad-CAM saliency maps using a baseline model, a multiattribute guided network (MAG), a contextual attention network (CA), and a combined MAG–CA network. Adapted from Zheng et al. (2024) [53], with permission from Elsevier.
Applsci 14 08108 g009
Figure 10. Input of the (a) original input image and (b) radiologist-highlighted region of hypoechoic lesion with mixed echogenicity prostate for malignant case, compared to the resultant (c) simulated image by LIME, which initially locates the regions that could be worthy of investigation given the input image, and (d) the final generated image by LIME explaining why the case was classified as malignant. Adapted from Hassan et al. (2022) [42], with permission from Elsevier.
Figure 10. Input of the (a) original input image and (b) radiologist-highlighted region of hypoechoic lesion with mixed echogenicity prostate for malignant case, compared to the resultant (c) simulated image by LIME, which initially locates the regions that could be worthy of investigation given the input image, and (d) the final generated image by LIME explaining why the case was classified as malignant. Adapted from Hassan et al. (2022) [42], with permission from Elsevier.
Applsci 14 08108 g010
Figure 11. Generation of the ROI and the local patches from the images using the global features. (a) The original image to be used as the input for the global branch. (b) The generated activation heat map of the features. (c) The binarized heat map and the bounding box spanning it. (d) The cropped local patch to be used as the input to the local branch. Adapted from Basu et al. (2023) [38], with permission from Elsevier.
Figure 11. Generation of the ROI and the local patches from the images using the global features. (a) The original image to be used as the input for the global branch. (b) The generated activation heat map of the features. (c) The binarized heat map and the bounding box spanning it. (d) The cropped local patch to be used as the input to the local branch. Adapted from Basu et al. (2023) [38], with permission from Elsevier.
Applsci 14 08108 g011
Figure 12. SHAP explanation example: Results of a malignant case in breast ultrasound images in which the trained ensemble model can be analyzed to provide explainable decision paths within a series of decision trees. In each tree classifier, orange arrows indicate the decision path. The model compares the texture features from the input image (represented by orange numbers at the bottom of each dashed box) with the learned thresholds (indicated by black triangles on each histogram) at each node of the decision tree. Adapted from Rezazadeh et al. (2022) [48] Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license.
Figure 12. SHAP explanation example: Results of a malignant case in breast ultrasound images in which the trained ensemble model can be analyzed to provide explainable decision paths within a series of decision trees. In each tree classifier, orange arrows indicate the decision path. The model compares the texture features from the input image (represented by orange numbers at the bottom of each dashed box) with the learned thresholds (indicated by black triangles on each histogram) at each node of the decision tree. Adapted from Rezazadeh et al. (2022) [48] Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license.
Applsci 14 08108 g012
Figure 13. US images presenting benign (left) and malignant (right) breast masses and the corresponding CAM-generated saliency maps pointing out the three predetermined regions in the US images. The white cross indicates the extreme activation value of CAM responsible for the particular pointing game result. Adapted from Byra et al. (2022) [39]; an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (CC BY-NC-ND).
Figure 13. US images presenting benign (left) and malignant (right) breast masses and the corresponding CAM-generated saliency maps pointing out the three predetermined regions in the US images. The white cross indicates the extreme activation value of CAM responsible for the particular pointing game result. Adapted from Byra et al. (2022) [39]; an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (CC BY-NC-ND).
Applsci 14 08108 g013
Table 1. General characteristics of included studies.
Table 1. General characteristics of included studies.
Author (Year)Cancer TypeClinical ApplicationStudy TypeImagingAcquisition TypeDatasetTotal Patients (Lesions)Total ImagesHealthy Images (%)Benign Images (%)Malignant Images (%)
Al-Jebrni A.H. (2023) [37]ThyroidDiagnosisRUSContinuousSTNU1810 (2068)4136-1884 (45.6%)2252 (54.4%)
Basu S. (2023) [38]GallbladderDiagnosisRUSSingle shotGBUS218 (147)1255432 (34.4%)558 (44.5%)265 (21.1%)
Byra, M. (2022) [39]BreastDiagnosisRUSSingle shotClinicalNA (272)272-149 (54.8%)123 (45.2%)
Dong, F. (2021) [40]BreastDiagnosisRUSContinuousClinical367 (785)579-247 (42.7%)332 (57.3%)
Han, X. (2022) [41]ThyroidDiagnosisPUSSingle ContinuousClinical3906 (3906)3906-1696 (43.4%)2210 (56.6%)
Hassan, M. R. (2022) [42]ProstateDiagnosisRUS + MRIContinuous (US) + Static slice (MRI)Cancer Imaging Archive1151 (NA)611,119-NANA
Karimzadeh, M. (2023) [43]BreastScreeningRUSContinuousBUSI, BUSIS, HMSSNA (2917)2186-NANA
Lombardi, A. (2023) [44]LeiomyogenicSurgery (preoperative)RUSContinuous, 3D volume scanClinical68 (68)68-60 (88.2%)8 (11.8%)
Martizzi, D. (2021) [45]BreastScreeningPUS3D volume scanClinical70 (60)7010 (14.3%)41 (58.6%)19 (27.1%)
Morris, J. (2023) [46]ThyroidDiagnosisRUSContinuousClinical307 (831)831-NANA
Qian, X. (2021) [47]BreastScreeningPUSContinuousClinical634 (721)10,815-NANA
Rezazadeh, A. (2022) [48]BreastDiagnosisRUSContinuousPublic600 (697)780133 (17.1%)210 (26.9%)487 (62.4%)
Song, D. (2023) [49]ThyroidScreeningRUSSingle shotClinical7236 (19,341)19,341-12943 (66.9%)6398 (33.1%)
Tasnim, J. (2024) [50]BreastDiagnosisRUSContinuousBUSI, Mendeley, UDIAT, OMI, BUET-BUSDNA (1494)1494-901 (60.3%)593 (39.7%)
Thomas, J. (2020) [51]ThyroidSurgery (preoperative)RUSContinuousClinical402 (482)2025-NANA
Zhang, B. (2021) [52]BreastDiagnosisRUSContinuousBUSI, BUSISNA (1192)1192-727 (61.0%)465 (39.0%)
Zheng, H. (2024) [53]Gastrointestinal stromal tumorsDiagnosisRUSContinuousClinical261 (261)1900--1900 (100%)
NA = not available, P = prospective, R = retrospective, US = ultrasound, MRI = magnetic resonance imaging.
Table 2. Properties of AI models for classification.
Table 2. Properties of AI models for classification.
Classification (n = 16)
Author (Year)ArchitectureTaskGround TruthAccuracyAUROCOther Performance
Al-Jebrni, A.H. (2023) [37]Inception-V3Tumor class benign or malignantClinical diagnosis0.8740.905Sens 0.895, Spec 0.858
Basu, S. (2023) * [38]BagNets33Tumor class benign or malignant (local-level)Histopathology + radiologist assigned lexicons0.9210.971Sens 0.923, Spec 0.961
Byra, M. (2022) [39]ResNetTumor class benign or malignantPathology0.8870.835Sens 0.801, Spec 0.868
Dong, F. (2021) [40]DenseNet-121Tumor class benign or malignantHistopathology0.8840.899Sens 0.879, Spec 0.892
Han, X. (2022) [41]DenseNets + SGE attention moduleTI-RADS risk level (multiclass) &Clinical diagnosis0.780 (w)-MAE 1.30
Tumor class benign or malignantHistopathology0.9540.981Sens 0.988, Spec 0.912, PPV 0.928, NPV 0.985
Hassan, M. R. (2022) [42]VGG-16 + Random ForestTumor class benign or malignantRadiologist manual annotation0.875--
Karimzadeh, M. (2023) * [43]VGG-16BI-RADS risk level (multiclass) &Clinical diagnosis0.852 (w)--
Tumor class benign or malignantClinical diagnosis0.913-Sens 0.940, Spec 0.858
Lombardi, A. (2023) [44]XGBoostTumor class benign or malignantClinical diagnosis-0.994Sens 0.875, Spec 0.983, F1-score 0.875, Brier score 0.0187
Morris, J. (2023) [46]Local texture quantizationTI-RADS risk level (multiclass)Clinical diagnosis>0.80--
Qian, X. (2021) [47]ResNet-18 + SENetBI-RADS risk level (multiclass)Histopathology-0.955-
Rezazadeh, A. (2022) [48]LightGBMTumor class benign or malignantClinical diagnosis0.910.93Prec 0.94, Rec 0.93, F1-score 0.93
Song, D. (2023) [49]DenseNet-121Tumor class benign or malignantPathology / Clinical diagnosisNA--
Tasnim, J. (2024) [50]ResNet-18 + InceptionV3Tumor class benign or malignantClinical diagnosis0.9150.952Sens 0.894, Spec 0.929, F1-score 0.893, MCC 0.824
Thomas, J. (2020) [51]ResNet 34Tumor class benign or malignantHistopathology0.777-Sens 0.849, Spec 0.743, PPV 0.609, NPV 0.912
Zhang, B. (2021) [52]VGG-16BI-RADS risk level (multiclass) &Clinical diagnosis0.843 (w)--
Tumor class benign or malignant &Clinical diagnosis0.889-Sens 0.838, Spec 0.923
Likelihood of malignancyClinical diagnosis--R2 0.671, MSE 0.153
Zheng, H. (2024) [53]VGG-16Tumor class benign or malignantPathology0.932-Prec 0.932, Rec 0.932, F1-score 0.932
* = Study with AI model encompassing multiple tasks, NA = not available, (w) = weighted score, AUROC = area under the receiver operator curve, Sens = sensitivity, Spec = specificity, MAE = mean absolute error, PPV = positive predictive value, NPV = negative predictive value, MCC = Matthew’s correlation coefficient, Rec = recall, MSE = mean squared error.
Table 3. Properties of AI models for segmentation and localization.
Table 3. Properties of AI models for segmentation and localization.
Segmentation (n = 2)
Author (Year)ArchitectureTaskGround TruthPerformance
Karimzadeh, M. (2023) * [43]U-NetLesion maskNADSC 0.827
Martizzi, D. (2021) [45]Gaussian Mixture Models + Blob detectionROI maskRadiologist manual annotation of ROIRecall 0.83 (benign) Recall 0.95 (malignant)
Localization (n = 1)
Author (Year)ArchitectureTaskGround TruthPerformance
Basu, S. (2023) * [38]ResNet-50ROI identification (global-level)Radiologist bounding box annotationMean IoU 0.484 Mean intersection 0.934
* = Study with AI model encompassing multiple tasks, NA = not available, DSC = Dice similarity coefficient, IoU = intersection over union.
Table 4. Characteristics of identified XAI methods.
Table 4. Characteristics of identified XAI methods.
XAI MethodModel-SpecificScopeExplanationAI TaskXAI FunctionXAI InputXAI OutputAnalysis TypeEvaluation
Activation heat map [38]NoGlobalIntrinsicLocalizationVisualizationImagesHeat map with top activated image featuresVisual inspectionQL
Activation Maximization [50]CNNsGlobalPost hocClassificationVisualizationFeature mapsActivation maximizing output image patternsVisual inspection, ZNCC scoreQL, QT
Bag-of-Features [38]TransformersLocalIntrinsicClassificationSemanticsROI in US imagesMost discriminative features for class predictionFeatures mapped to radiological lexiconQL
BI-RADS-Net [52]MTL using CNNsLocalIntrinsicClassificationSemantics, Example basedBreast US images + feature mapsPredicted class probability + morphological feature explanationsAccuracy, sensitivity, and specificityQT
CAM [39,50]CNNsLocalPost hocClassificationVisualizationImagesSaliency mapVisual inspection, pointing gameQL, QT
Explainer [49]CNNsLocalIntrinsicClassificationVisualizationImages + convoluted feature mapsFeatures the model uses to make the predictionsVisual inspection, accuracy, precision, sensitivity, specificity, F1-score, AUROCQL, QT
Grad-CAM [37,41,47,49,53]CNNsLocalPost hocClassification (n = 6), Segmentation (n = 1)VisualizationImagesSaliency mapVisual inspectionQL
Image similarity AIBx [51])CNNsLocalPost hocClassificationSemantics, example-basedImagesMost similar images in database with known diagnosesConfusion matrixQT
LIME [42]NoLocalPost hocClassificationVisualizationImagesROI identifiedVisual inspectionQL
LTQ-E [46]NoLocalPost hocClassificationSemanticsUS image + Embedded decision label modelPredicted class + morphological feature explanationsAccuracyQT
MT-BI-RADS [43]MTL using CNNsLocalIntrinsicSegmentationVisualizationBreast US imagesTumor maskVisual inspection, DSCQL, QT
Region of Evidence (ROE) [40]CNNsLocalPost hocClassificationVisualizationImagesSaliency map + Resemblance vote by expert physiciansConfusion matrix + breakdown in resemblance votesQT
SHAP [43,44,48]NoLocalPost hocClassificationSemanticsImage + feature mapSignificance (Shapley) value per featureShapley values, Confusion matrixQL, QT
XNML [45]PRISMTM platform (ML)LocalIntrinsicSegmentationVisualization, Example-based3D Quantitative transmission US speed-of-sound mapsColor-coded segmentationsVisual inspectionQL
QL = Qualitative, QT = Quantitative, MTL = Multitask Learning.
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

Wyatt, L.S.; van Karnenbeek, L.M.; Wijkhuizen, M.; Geldof, F.; Dashtbozorg, B. Explainable Artificial Intelligence (XAI) for Oncological Ultrasound Image Analysis: A Systematic Review. Appl. Sci. 2024, 14, 8108. https://doi.org/10.3390/app14188108

AMA Style

Wyatt LS, van Karnenbeek LM, Wijkhuizen M, Geldof F, Dashtbozorg B. Explainable Artificial Intelligence (XAI) for Oncological Ultrasound Image Analysis: A Systematic Review. Applied Sciences. 2024; 14(18):8108. https://doi.org/10.3390/app14188108

Chicago/Turabian Style

Wyatt, Lucie S., Lennard M. van Karnenbeek, Mark Wijkhuizen, Freija Geldof, and Behdad Dashtbozorg. 2024. "Explainable Artificial Intelligence (XAI) for Oncological Ultrasound Image Analysis: A Systematic Review" Applied Sciences 14, no. 18: 8108. https://doi.org/10.3390/app14188108

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