420 research outputs found

    DERMA: A melanoma diagnosis platform based on collaborative multilabel analog reasoning

    Get PDF
    The number of melanoma cancer-related death has increased over the last few years due to the new solar habits. Early diagnosis has become the best prevention method. This work presents a melanoma diagnosis architecture based on the collaboration of several multilabel case-based reasoning subsystems called DERMA. The system has to face up several challenges that include data characterization, pattern matching, reliable diagnosis, and self-explanation capabilities. Experiments using subsystems specialized in confocal and dermoscopy images have provided promising results for helping experts to assess melanoma diagnosis

    Ajuda al Diagnòstic de Càncer de Melanoma amb Raonament Analògic Multietiqueta

    Get PDF
    La mortalitat provocada pel càncer de melanoma ha augmentat en els últims anys a causa, principalment, dels nous hàbits d'exposició al sol. Atenent al criteri mèdic, el diagnòstic precoç s'ha convertit en el millor mètode de prevenció. No és però una tasca trivial ja que els experts del domini han de fer front a un problema caracteritzat per tenir un gran volum de dades, de format heterogeni i amb coneixement parcial. A partir d'aquestes necessitats es proposa la creació d'una eina de suport a la presa de decisions que sigui capaç d'ajudar els experts en melanoma en el seu diagnòstic. El sistema ha de fer front a diversos reptes plantejats, que inclouen la caracterització del domini, la identificació de patrons a les dades segons el criteri dels experts, la classificació de nous pacients i la capacitat d'explicar els pronòstics obtinguts. Aquestes fites s'han materialitzat en la plataforma DERMA, la qual està basada en la col•laboració de diversos subsistemes de raonament analògic multietiqueta. L'experimentació realitzada amb el sistema proposat utilitzant dades d'imatges confocals i dermatoscòpiques ha permès comprovar la fiabilitat del sistema. Els resultats obtinguts han estat validats pels experts en el diagnòstic del melanoma considerant-los positius.La mortalidad a causa del cáncer de melanoma ha aumentado en los últimos años debido, principalmente, a los nuevos hábitos de exposición al sol. Atendiendo al criterio médico, el diagnóstico precoz se ha convertido en el mejor método de prevención, pero no se trata de una tarea trivial puesto que los expertos del dominio deben hacer frente a un problema caracterizado por tener un gran volumen de datos, de formato heterogéneo y con conocimiento parcial. A partir de estas necesidades se propone la creación de una herramienta de ayuda a la toma de decisiones que sea capaz de ayudar a los expertos en melanoma en su diagnóstico. El sistema tiene que hacer frente a diversos retos planteados, que incluyen la caracterización del dominio, la identificación de patrones en los datos según el criterio médico, la clasificación de nuevos pacientes y la capacidad de explicar los pronósticos obtenidos. Estas metas se han materializado en la plataforma DERMA la cual está basada en la colaboración de varios subsistemas de razonamiento analógico multietiqueta. La experimentación realizada con el sistema propuesto utilizando datos de imágenes confocales y dermatoscópicas ha permitido verificar la fiabilidad del sistema. Los resultados obtenidos han sido validados por los expertos en el diagnóstico del melanoma considerándolos positivos.Mortality related to melanoma cancer has increased in recent years, mainly due to new habits of sun exposure. Considering the medical criteria, early diagnosis has become the best method of prevention but this is not trivial because experts are facing a problem characterized by a large volume of data, heterogeneous, and with partial knowledge. Based on these requirements we propose the creation of a decision support system that is able to assist experts in melanoma diagnosis. The system has to cope with various challenges, that include the characterization of the domain, the identification of data patterns attending to medical criteria, the classification of new patients, and the ability to explain predictions. These goals have been materialized in DERMA platform that is based on the collaboration of several analogical reasoning multi-label subsystems. The experiments conducted with the proposed system using confocal and dermoscopic images data have been allowed to ascertain the reliability of the system. The results have been validated by experts in diagnosis of melanoma considering it as positive

    DERMA: A Melanoma Diagnosis Platform Based on Collaborative Multilabel Analog Reasoning

    Get PDF
    The number of melanoma cancer-related death has increased over the last few years due to the new solar habits. Early diagnosis has become the best prevention method. This work presents a melanoma diagnosis architecture based on the collaboration of several multilabel case-based reasoning subsystems called DERMA. The system has to face up several challenges that include data characterization, pattern matching, reliable diagnosis, and self-explanation capabilities. Experiments using subsystems specialized in confocal and dermoscopy images have provided promising results for helping experts to assess melanoma diagnosis

    Towards PACE-CAD Systems

    Get PDF
    Despite phenomenal advancements in the availability of medical image datasets and the development of modern classification algorithms, Computer-Aided Diagnosis (CAD) has had limited practical exposure in the real-world clinical workflow. This is primarily because of the inherently demanding and sensitive nature of medical diagnosis that can have far-reaching and serious repercussions in case of misdiagnosis. In this work, a paradigm called PACE (Pragmatic, Accurate, Confident, & Explainable) is presented as a set of some of must-have features for any CAD. Diagnosis of glaucoma using Retinal Fundus Images (RFIs) is taken as the primary use case for development of various methods that may enrich an ordinary CAD system with PACE. However, depending on specific requirements for different methods, other application areas in ophthalmology and dermatology have also been explored. Pragmatic CAD systems refer to a solution that can perform reliably in day-to-day clinical setup. In this research two, of possibly many, aspects of a pragmatic CAD are addressed. Firstly, observing that the existing medical image datasets are small and not representative of images taken in the real-world, a large RFI dataset for glaucoma detection is curated and published. Secondly, realising that a salient attribute of a reliable and pragmatic CAD is its ability to perform in a range of clinically relevant scenarios, classification of 622 unique cutaneous diseases in one of the largest publicly available datasets of skin lesions is successfully performed. Accuracy is one of the most essential metrics of any CAD system's performance. Domain knowledge relevant to three types of diseases, namely glaucoma, Diabetic Retinopathy (DR), and skin lesions, is industriously utilised in an attempt to improve the accuracy. For glaucoma, a two-stage framework for automatic Optic Disc (OD) localisation and glaucoma detection is developed, which marked new state-of-the-art for glaucoma detection and OD localisation. To identify DR, a model is proposed that combines coarse-grained classifiers with fine-grained classifiers and grades the disease in four stages with respect to severity. Lastly, different methods of modelling and incorporating metadata are also examined and their effect on a model's classification performance is studied. Confidence in diagnosing a disease is equally important as the diagnosis itself. One of the biggest reasons hampering the successful deployment of CAD in the real-world is that medical diagnosis cannot be readily decided based on an algorithm's output. Therefore, a hybrid CNN architecture is proposed with the convolutional feature extractor trained using point estimates and a dense classifier trained using Bayesian estimates. Evaluation on 13 publicly available datasets shows the superiority of this method in terms of classification accuracy and also provides an estimate of uncertainty for every prediction. Explainability of AI-driven algorithms has become a legal requirement after Europe’s General Data Protection Regulations came into effect. This research presents a framework for easy-to-understand textual explanations of skin lesion diagnosis. The framework is called ExAID (Explainable AI for Dermatology) and relies upon two fundamental modules. The first module uses any deep skin lesion classifier and performs detailed analysis on its latent space to map human-understandable disease-related concepts to the latent representation learnt by the deep model. The second module proposes Concept Localisation Maps, which extend Concept Activation Vectors by locating significant regions corresponding to a learned concept in the latent space of a trained image classifier. This thesis probes many viable solutions to equip a CAD system with PACE. However, it is noted that some of these methods require specific attributes in datasets and, therefore, not all methods may be applied on a single dataset. Regardless, this work anticipates that consolidating PACE into a CAD system can not only increase the confidence of medical practitioners in such tools but also serve as a stepping stone for the further development of AI-driven technologies in healthcare

    A Comprehensive Survey of Convolutional Neural Networks for Skin Cancer Classification and Prediction

    Get PDF
    Skin cancer, a prevalent and potentially fatal condition, requires early detection and precise classification to ensure effective treatment. In recent years, there has been a significant rise in the popularity of Convolutional Neural Networks (CNNs) prominence as a robust solution for image processing and analysis, significantly surpassing conventional techniques in skin cancer prediction and classification. This survey paper offers a thorough examination of CNNs and their diverse applications in diagnosing skin cancer, emphasizing their benefits, existing obstacles, and potential avenues for future research

    Dermatologist-like explainable AI enhances trust and confidence in diagnosing melanoma

    Full text link
    Although artificial intelligence (AI) systems have been shown to improve the accuracy of initial melanoma diagnosis, the lack of transparency in how these systems identify melanoma poses severe obstacles to user acceptance. Explainable artificial intelligence (XAI) methods can help to increase transparency, but most XAI methods are unable to produce precisely located domain-specific explanations, making the explanations difficult to interpret. Moreover, the impact of XAI methods on dermatologists has not yet been evaluated. Extending on two existing classifiers, we developed an XAI system that produces text and region based explanations that are easily interpretable by dermatologists alongside its differential diagnoses of melanomas and nevi. To evaluate this system, we conducted a three-part reader study to assess its impact on clinicians' diagnostic accuracy, confidence, and trust in the XAI-support. We showed that our XAI's explanations were highly aligned with clinicians' explanations and that both the clinicians' trust in the support system and their confidence in their diagnoses were significantly increased when using our XAI compared to using a conventional AI system. The clinicians' diagnostic accuracy was numerically, albeit not significantly, increased. This work demonstrates that clinicians are willing to adopt such an XAI system, motivating their future use in the clinic

    Melanoma Skin Cancer Identification with Explainability Utilizing Mask Guided Technique

    Get PDF
    Melanoma is a highly prevalent and lethal form of skin cancer, which has a significant impact globally. The chances of recovery for melanoma patients substantially improve with early detection. Currently, deep learning (DL) methods are gaining popularity in assisting with the identification of diseases using medical imaging. The paper introduces a computational model for classifying melanoma skin cancer images using convolutional neural networks (CNNs) and vision transformers (ViT) with the HAM10000 dataset. Both approaches utilize mask-guided techniques, employing a specialized U2-Net segmentation module to generate masks. The CNN-based approach utilizes ResNet50, VGG16, and Xception with transfer learning. The training process is enhanced using a Bayesian hyperparameter tuner. Moreover, this study applies gradient-weighted class activation mapping (Grad-CAM) and Grad-CAM++ to generate heatmaps to explain the classification models. These visual heatmaps elucidate the contribution of each input region to the classification outcome. The CNN-based model approach achieved the highest accuracy at 98.37% in the Xception model with a sensitivity and specificity of 95.92% and 99.01%, respectively. The ViT-based model approach achieved high values for accuracy, sensitivity, and specificity, such as 92.79%, 91.09%, and 93.54%, respectively. Furthermore, the performance of the model was assessed through intersection over union (IOU) and other qualitative evaluations. Finally, we developed the proposed model as a web application that can be used as a support tool for medical practitioners in real-time. The system usability study score of 86.87% is reported, which shows the usefulness of the proposed solution

    Ensemble-based genetic algorithm explainer with automized image segmentation: A case study on melanoma detection dataset

    Get PDF
    Explainable Artificial Intelligence (XAI) makes AI understandable to the human user particularly when the model is complex and opaque. Local Interpretable Model-agnostic Explanations (LIME) has an image explainer package that is used to explain deep learning models. The image explainer of LIME needs some parameters to be manually tuned by the expert in advance, including the number of top features to be seen and the number of superpixels in the segmented input image. This parameter tuning is a time-consuming task. Hence, with the aim of developing an image explainer that automizes image segmentation, this paper proposes Ensemblebased Genetic Algorithm Explainer (EGAE) for melanoma cancer detection that automatically detects and presents the informative sections of the image to the user. EGAE has three phases. First, the sparsity of chromosomes in GAs is determined heuristically. Then, multiple GAs are executed consecutively. However, the difference between these GAs are in different number of superpixels in the input image that result in different chromosome lengths. Finally, the results of GAs are ensembled using consensus and majority votings. This paper also introduces how Euclidean distance can be used to calculate the distance between the actual explanation (delineated by experts) and the calculated explanation (computed by the explainer) for accuracy measurement. Experimental results on a melanoma dataset show that EGAE automatically detects informative lesions, and it also improves the accuracy of explanation in comparison with LIME efficiently. The python codes for EGAE, the ground truths delineated by clinicians, and the melanoma detection dataset are available at https://github.com/KhaosResearch/EGAEThis work has been partially funded by grant PID2020-112540RBC41 (funded by MCIN/AEI/10.13039/501100011033/, Spain), AETHERUMA, Spain (A smart data holistic approach for context-aware data analytics: semantics and context exploitation). Funding for open access charge: Universidad de Málaga/CBUA. Additionally, we thank Dr. Miguel Ángel Berciano Guerrero from Unidad de Oncología Intercentros, Hospitales Univesitarios Regional Virgen de la Victoria de Málaga, and Instituto de Investigaciones Biomédicas (IBIMA), Málaga, Spain, for his support in images selection and general medical orientation in the particular case of Melanoma

    Diagnóstico automático de melanoma mediante técnicas modernas de aprendizaje automático

    Get PDF
    The incidence and mortality rates of skin cancer remain a huge concern in many countries. According to the latest statistics about melanoma skin cancer, only in the Unites States, 7,650 deaths are expected in 2022, which represents 800 and 470 more deaths than 2020 and 2021, respectively. In 2022, melanoma is ranked as the fifth cause of new cases of cancer, with a total of 99,780 people. This illness is mainly diagnosed with a visual inspection of the skin, then, if doubts remain, a dermoscopic analysis is performed. The development of e_ective non-invasive diagnostic tools for the early stages of the illness should increase quality of life, and decrease the required economic resources. The early diagnosis of skin lesions remains a tough task even for expert dermatologists because of the complexity, variability, dubiousness of the symptoms, and similarities between the different categories among skin lesions. To achieve this goal, previous works have shown that early diagnosis from skin images can benefit greatly from using computational methods. Several studies have applied handcrafted-based methods on high quality dermoscopic and histological images, and on top of that, machine learning techniques, such as the k-nearest neighbors approach, support vector machines and random forest. However, one must bear in mind that although the previous extraction of handcrafted features incorporates an important knowledge base into the analysis, the quality of the extracted descriptors relies heavily on the contribution of experts. Lesion segmentation is also performed manually. The above procedures have a common issue: they are time-consuming manual processes prone to errors. Furthermore, an explicit definition of an intuitive and interpretable feature is hardly achievable, since it depends on pixel intensity space and, therefore, they are not invariant regarding the differences in the input images. On the other hand, the use of mobile devices has sharply increased, which offers an almost unlimited source of data. In the past few years, more and more attention has been paid to designing deep learning models for diagnosing melanoma, more specifically Convolutional Neural Networks. This type of model is able to extract and learn high-level features from raw images and/or other data without the intervention of experts. Several studies showed that deep learning models can overcome handcrafted-based methods, and even match the predictive performance of dermatologists. The International Skin Imaging Collaboration encourages the development of methods for digital skin imaging. Every year since 2016 to 2019, a challenge and a conference have been organized, in which more than 185 teams have participated. However, convolutional models present several issues for skin diagnosis. These models can fit on a wide diversity of non-linear data points, being prone to overfitting on datasets with small numbers of training examples per class and, therefore, attaining a poor generalization capacity. On the other hand, this type of model is sensitive to some characteristics in data, such as large inter-class similarities and intra-class variances, variations in viewpoints, changes in lighting conditions, occlusions, and background clutter, which can be mostly found in non-dermoscopic images. These issues represent challenges for the application of automatic diagnosis techniques in the early phases of the illness. As a consequence of the above, the aim of this Ph.D. thesis is to make significant contributions to the automatic diagnosis of melanoma. The proposals aim to avoid overfitting and improve the generalization capacity of deep models, as well as to achieve a more stable learning and better convergence. Bear in mind that research into deep learning commonly requires an overwhelming processing power in order to train complex architectures. For example, when developing NASNet architecture, researchers used 500 x NVidia P100s - each graphic unit cost from 5,899to5,899 to 7,374, which represents a total of 2,949,500.002,949,500.00 - 3,687,000.00. Unfortunately, the majority of research groups do not have access to such resources, including ours. In this Ph.D. thesis, the use of several techniques has been explored. First, an extensive experimental study was carried out, which included state-of-the-art models and methods to further increase the performance. Well-known techniques were applied, such as data augmentation and transfer learning. Data augmentation is performed in order to balance out the number of instances per category and act as a regularizer in preventing overfitting in neural networks. On the other hand, transfer learning uses weights of a pre-trained model from another task, as the initial condition for the learning of the target network. Results demonstrate that the automatic diagnosis of melanoma is a complex task. However, different techniques are able to mitigate such issues in some degree. Finally, suggestions are given about how to train convolutional models for melanoma diagnosis and future interesting research lines were presented. Next, the discovery of ensemble-based architectures is tackled by using genetic algorithms. The proposal is able to stabilize the training process. This is made possible by finding sub-optimal combinations of abstract features from the ensemble, which are used to train a convolutional block. Then, several predictive blocks are trained at the same time, and the final diagnosis is achieved by combining all individual predictions. We empirically investigate the benefits of the proposal, which shows better convergence, mitigates the overfitting of the model, and improves the generalization performance. On top of that, the proposed model is available online and can be consulted by experts. The next proposal is focused on designing an advanced architecture capable of fusing classical convolutional blocks and a novel model known as Dynamic Routing Between Capsules. This approach addresses the limitations of convolutional blocks by using a set of neurons instead of an individual neuron in order to represent objects. An implicit description of the objects is learned by each capsule, such as position, size, texture, deformation, and orientation. In addition, a hyper-tuning of the main parameters is carried out in order to ensure e_ective learning under limited training data. An extensive experimental study was conducted where the fusion of both methods outperformed six state-of-the-art models. On the other hand, a robust method for melanoma diagnosis, which is inspired on residual connections and Generative Adversarial Networks, is proposed. The architecture is able to produce plausible photorealistic synthetic 512 x 512 skin images, even with small dermoscopic and non-dermoscopic skin image datasets as problema domains. In this manner, the lack of data, the imbalance problems, and the overfitting issues are tackled. Finally, several convolutional modes are extensively trained and evaluated by using the synthetic images, illustrating its effectiveness in the diagnosis of melanoma. In addition, a framework, which is inspired on Active Learning, is proposed. The batch-based query strategy setting proposed in this work enables a more faster training process by learning about the complexity of the data. Such complexities allow us to adjust the training process after each epoch, which leads the model to achieve better performance in a lower number of iterations compared to random mini-batch sampling. Then, the training method is assessed by analyzing both the informativeness value of each image and the predictive performance of the models. An extensive experimental study is conducted, where models trained with the proposal attain significantly better results than the baseline models. The findings suggest that there is still space for improvement in the diagnosis of skin lesions. Structured laboratory data, unstructured narrative data, and in some cases, audio or observational data, are given by radiologists as key points during the interpretation of the prediction. This is particularly true in the diagnosis of melanoma, where substantial clinical context is often essential. For example, symptoms like itches and several shots of a skin lesion during a period of time proving that the lesion is growing, are very likely to suggest cancer. The use of different types of input data could help to improve the performance of medical predictive models. In this regard, a _rst evolutionary algorithm aimed at exploring multimodal multiclass data has been proposed, which surpassed a single-input model. Furthermore, the predictive features extracted by primary capsules could be used to train other models, such as Support Vector Machine

    Personalized medicine support system : resolving conflict in allocation to risk groups and predicting patient molecular response to targeted therapy

    Get PDF
    Treatment management in cancer patients is largely based on the use of a standardized set of predictive and prognostic factors. The former are used to evaluate specific clinical interventions, and they can be useful for selecting treatments because they directly predict the response to a treatment. The latter are used to evaluate a patient’s overall outcomes, and can be used to identify the risks or recurrence of a disease. Current intelligent systems can be a solution for transferring advancements in molecular biology into practice, especially for predicting the molecular response to molecular targeted therapy and the prognosis of risk groups in cancer medicine. This framework primarily focuses on the importance of integrating domain knowledge in predictive and prognostic models for personalized treatment. Our personalized medicine support system provides the needed support in complex decisions and can be incorporated into a treatment guide for selecting molecular targeted therapies.Haneen Banjar, David Adelson, Fred Brown, and Tamara Leclerc
    corecore