Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Patterns (N Y) ; 5(7): 100992, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39081575

RESUMEN

Prostate cancer diagnosis and treatment relies on precise MRI lesion segmentation, a challenge notably for small (<15 mm) and intermediate (15-30 mm) lesions. Our study introduces ProLesA-Net, a multi-channel 3D deep-learning architecture with multi-scale squeeze and excitation and attention gate mechanisms. Tested against six models across two datasets, ProLesA-Net significantly outperformed in key metrics: Dice score increased by 2.2%, and Hausdorff distance and average surface distance improved by 0.5 mm, with recall and precision also undergoing enhancements. Specifically, for lesions under 15 mm, our model showed a notable increase in five key metrics. In summary, ProLesA-Net consistently ranked at the top, demonstrating enhanced performance and stability. This advancement addresses crucial challenges in prostate lesion segmentation, enhancing clinical decision making and expediting treatment processes.

2.
Artículo en Inglés | MEDLINE | ID: mdl-38865229

RESUMEN

Developing AI models for digital pathology has traditionally relied on single-scale analysis of histopathology slides. However, a whole slide image is a rich digital representation of the tissue, captured at various magnification levels. Limiting our analysis to a single scale overlooks critical information, spanning from intricate high-resolution cellular details to broad low-resolution tissue structures. In this study, we propose a model-agnostic multiresolution feature aggregation framework tailored for the analysis of histopathology slides in the context of breast cancer, on a multicohort dataset of 2038 patient samples. We have adapted 9 state-of-the-art multiple instance learning models on our multi-scale methodology and evaluated their performance on grade prediction, TP53 mutation status prediction and survival prediction. The results prove the dominance of the multiresolution methodology, and specifically, concatenating or linearly transforming via a learnable layer the feature vectors of image patches from a high (20x) and low (10x) magnification factors achieve improved performance for all prediction tasks across domain-specific and imagenet-based features. On the contrary, the performance of uniresolution baseline models was not consistent across domain-specific and imagenet-based features. Moreover, we shed light on the inherent inconsistencies observed in models trained on whole-tissue-sections when validated against biopsy-based datasets. Despite these challenges, our findings underscore the superiority of multiresolution analysis over uniresolution methods. Finally, cross-scale analysis also benefits the explainability aspects of attention-based architectures, since one can extract attention maps at the tissue- and cell-levels, improving the interpretation of the model's decision. The code and results of this study can be found at github.com/tsikup/multiresolution_histopathology.

3.
J Imaging ; 10(5)2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38786569

RESUMEN

Image quality assessment of magnetic resonance imaging (MRI) data is an important factor not only for conventional diagnosis and protocol optimization but also for fairness, trustworthiness, and robustness of artificial intelligence (AI) applications, especially on large heterogeneous datasets. Information on image quality in multi-centric studies is important to complement the contribution profile from each data node along with quantity information, especially when large variability is expected, and certain acceptance criteria apply. The main goal of this work was to present a tool enabling users to assess image quality based on both subjective criteria as well as objective image quality metrics used to support the decision on image quality based on evidence. The evaluation can be performed on both conventional and dynamic MRI acquisition protocols, while the latter is also checked longitudinally across dynamic series. The assessment provides an overall image quality score and information on the types of artifacts and degrading factors as well as a number of objective metrics for automated evaluation across series (BRISQUE score, Total Variation, PSNR, SSIM, FSIM, MS-SSIM). Moreover, the user can define specific regions of interest (ROIs) to calculate the regional signal-to-noise ratio (SNR) and contrast-to-noise ratio (CNR), thus individualizing the quality output to specific use cases, such as tissue-specific contrast or regional noise quantification.

4.
Cancers (Basel) ; 16(10)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38791897

RESUMEN

To investigate the incidence and prognostically significant correlations and cooperations of LKB1 loss of expression in non-small cell lung cancer (NSCLC), surgical specimens from 188 metastatic and 60 non-metastatic operable stage I-IIIA NSCLC patients were analyzed to evaluate their expression of LKB1 and pAMPK proteins in relation to various processes. The investigated factors included antitumor immunity response regulators STING and PD-L1; pro-angiogenic, EMT and cell cycle targets, as well as metastasis-related (VEGFC, PDGFRα, PDGFRß, p53, p16, Cyclin D1, ZEB1, CD24) targets; and cell adhesion (ß-catenin) molecules. The protein expression levels were evaluated via immunohistochemistry; the RNA levels of LKB1 and NEDD9 were evaluated via PCR, while KRAS exon 2 and BRAFV600E mutations were evaluated by Sanger sequencing. Overall, loss of LKB1 protein expression was observed in 21% (51/248) patients and correlated significantly with histotype (p < 0.001), KRAS mutations (p < 0.001), KC status (concomitant KRAS mutation and p16 downregulation) (p < 0.001), STING loss (p < 0.001), and high CD24 expression (p < 0.001). STING loss also correlated significantly with loss of LKB1 expression in the metastatic setting both overall (p = 0.014) and in lung adenocarcinomas (LUACs) (p = 0.005). Additionally, LKB1 loss correlated significantly with a lack of or low ß-catenin membranous expression exclusively in LUACs, both independently of the metastatic status (p = 0.019) and in the metastatic setting (p = 0.007). Patients with tumors yielding LKB1 loss and concomitant nonexistent or low ß-catenin membrane expression experienced significantly inferior median overall survival of 20.50 vs. 52.99 months; p < 0.001 as well as significantly greater risk of death (HR: 3.32, 95% c.i.: 1.71-6.43; p <0.001). Our findings underscore the impact of the synergy of LKB1 with STING and ß-catenin in NSCLC, in prognosis.

5.
Insights Imaging ; 15(1): 130, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38816658

RESUMEN

Artificial intelligence (AI) is revolutionizing the field of medical imaging, holding the potential to shift medicine from a reactive "sick-care" approach to a proactive focus on healthcare and prevention. The successful development of AI in this domain relies on access to large, comprehensive, and standardized real-world datasets that accurately represent diverse populations and diseases. However, images and data are sensitive, and as such, before using them in any way the data needs to be modified to protect the privacy of the patients. This paper explores the approaches in the domain of five EU projects working on the creation of ethically compliant and GDPR-regulated European medical imaging platforms, focused on cancer-related data. It presents the individual approaches to the de-identification of imaging data, and describes the problems and the solutions adopted in each case. Further, lessons learned are provided, enabling future projects to optimally handle the problem of data de-identification. CRITICAL RELEVANCE STATEMENT: This paper presents key approaches from five flagship EU projects for the de-identification of imaging and clinical data offering valuable insights and guidelines in the domain. KEY POINTS: ΑΙ models for health imaging require access to large amounts of data. Access to large imaging datasets requires an appropriate de-identification process. This paper provides de-identification guidelines from the AI for health imaging (AI4HI) projects.

6.
Eur Radiol Exp ; 8(1): 42, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38589742

RESUMEN

BACKGROUND: Developing trustworthy artificial intelligence (AI) models for clinical applications requires access to clinical and imaging data cohorts. Reusing of publicly available datasets has the potential to fill this gap. Specifically in the domain of breast cancer, a large archive of publicly accessible medical images along with the corresponding clinical data is available at The Cancer Imaging Archive (TCIA). However, existing datasets cannot be directly used as they are heterogeneous and cannot be effectively filtered for selecting specific image types required to develop AI models. This work focuses on the development of a homogenized dataset in the domain of breast cancer including clinical and imaging data. METHODS: Five datasets were acquired from the TCIA and were harmonized. For the clinical data harmonization, a common data model was developed and a repeatable, documented "extract-transform-load" process was defined and executed for their homogenization. Further, Digital Imaging and COmmunications in Medicine (DICOM) information was extracted from magnetic resonance imaging (MRI) data and made accessible and searchable. RESULTS: The resulting harmonized dataset includes information about 2,035 subjects with breast cancer. Further, a platform named RV-Cherry-Picker enables search over both the clinical and diagnostic imaging datasets, providing unified access, facilitating the downloading of all study imaging that correspond to specific series' characteristics (e.g., dynamic contrast-enhanced series), and reducing the burden of acquiring the appropriate set of images for the respective AI model scenario. CONCLUSIONS: RV-Cherry-Picker provides access to the largest, publicly available, homogenized, imaging/clinical dataset for breast cancer to develop AI models on top. RELEVANCE STATEMENT: We present a solution for creating merged public datasets supporting AI model development, using as an example the breast cancer domain and magnetic resonance imaging images. KEY POINTS: • The proposed platform allows unified access to the largest, homogenized public imaging dataset for breast cancer. • A methodology for the semantically enriched homogenization of public clinical data is presented. • The platform is able to make a detailed selection of breast MRI data for the development of AI models.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/diagnóstico por imagen , Inteligencia Artificial , Mama
8.
Comput Biol Med ; 171: 108216, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38442555

RESUMEN

Despite being one of the most prevalent forms of cancer, prostate cancer (PCa) shows a significantly high survival rate, provided there is timely detection and treatment. Computational methods can help make this detection process considerably faster and more robust. However, some modern machine-learning approaches require accurate segmentation of the prostate gland and the index lesion. Since performing manual segmentations is a very time-consuming task, and highly prone to inter-observer variability, there is a need to develop robust semi-automatic segmentation models. In this work, we leverage the large and highly diverse ProstateNet dataset, which includes 638 whole gland and 461 lesion segmentation masks, from 3 different scanner manufacturers provided by 14 institutions, in addition to other 3 independent public datasets, to train accurate and robust segmentation models for the whole prostate gland, zones and lesions. We show that models trained on large amounts of diverse data are better at generalizing to data from other institutions and obtained with other manufacturers, outperforming models trained on single-institution single-manufacturer datasets in all segmentation tasks. Furthermore, we show that lesion segmentation models trained on ProstateNet can be reliably used as lesion detection models.


Asunto(s)
Próstata , Neoplasias de la Próstata , Masculino , Humanos , Próstata/diagnóstico por imagen , Imagenología Tridimensional/métodos , Estudios Retrospectivos , Algoritmos , Neoplasias de la Próstata/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos
9.
Clin Neurol Neurosurg ; 239: 108209, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38430649

RESUMEN

Elevated intracranial pressure (ICP) is a life-threatening condition that must be promptly diagnosed. However, the gold standard methods for ICP monitoring are invasive, time-consuming, and they involve certain risks. To address these risks, many noninvasive approaches have been proposed. This study undertakes a literature review of the existing noninvasive methods, which have reported promising results. The experimental base on which they are established, however, prevents their application in emergency conditions and thus none of them are capable of replacing the traditional invasive methods to date. On the other hand, contemporary methods leverage Machine Learning (ML) which has already shown unprecedented results in several medical research areas. That said, only a few publications exist on ML-based approaches for ICP estimation, which are not appropriate for emergency conditions due to their restricted capability of employing the medical imaging data available in intensive care units. The lack of such image-based ML models to estimate ICP is attributed to the scarcity of annotated datasets requiring directly measured ICP data. This ascertainment highlights an active and unexplored scientific frontier, calling for further research and development in the field of ICP estimation, particularly leveraging the untapped potential of ML techniques.


Asunto(s)
Hipertensión Intracraneal , Presión Intracraneal , Humanos , Monitoreo Fisiológico/métodos , Hipertensión Intracraneal/diagnóstico , Unidades de Cuidados Intensivos
10.
Eur Radiol ; 34(2): 1179-1186, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37581656

RESUMEN

OBJECTIVES: To develop a deep learning methodology that distinguishes early from late stages of avascular necrosis of the hip (AVN) to determine treatment decisions. METHODS: Three convolutional neural networks (CNNs) VGG-16, Inception ResnetV2, InceptionV3 were trained with transfer learning (ImageNet) and finetuned with a retrospectively collected cohort of (n = 104) MRI examinations of AVN patients, to differentiate between early (ARCO 1-2) and late (ARCO 3-4) stages. A consensus CNN ensemble decision was recorded as the agreement of at least two CNNs. CNN and ensemble performance was benchmarked on an independent cohort of 49 patients from another country and was compared to the performance of two MSK radiologists. CNN performance was expressed with areas under the curve (AUC), the respective 95% confidence intervals (CIs) and precision, and recall and f1-scores. AUCs were compared with DeLong's test. RESULTS: On internal testing, Inception-ResnetV2 achieved the highest individual performance with an AUC of 99.7% (95%CI 99-100%), followed by InceptionV3 and VGG-16 with AUCs of 99.3% (95%CI 98.4-100%) and 97.3% (95%CI 95.5-99.2%) respectively. The CNN ensemble the same AUCs Inception ResnetV2. On external validation, model performance dropped with VGG-16 achieving the highest individual AUC of 78.9% (95%CI 51.6-79.6%) The best external performance was achieved by the model ensemble with an AUC of 85.5% (95%CI 72.2-93.9%). No significant difference was found between the CNN ensemble and expert MSK radiologists (p = 0.22 and 0.092 respectively). CONCLUSION: An externally validated CNN ensemble accurately distinguishes between the early and late stages of AVN and has comparable performance to expert MSK radiologists. CLINICAL RELEVANCE STATEMENT: This paper introduces the use of deep learning for the differentiation between early and late avascular necrosis of the hip, assisting in a complex clinical decision that can determine the choice between conservative and surgical treatment. KEY POINTS: • A convolutional neural network ensemble achieved excellent performance in distinguishing between early and late avascular necrosis. • The performance of the deep learning method was similar to the performance of expert readers.


Asunto(s)
Aprendizaje Profundo , Osteonecrosis , Humanos , Estudios Retrospectivos , Redes Neurales de la Computación , Imagen por Resonancia Magnética/métodos
11.
Front Neurol ; 14: 1249452, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38046592

RESUMEN

Objective: This study aims to assess the value of biomarker based radiomics to predict IDH mutation in gliomas. The patient cohort consists of 160 patients histopathologicaly proven of primary glioma (WHO grades 2-4) from 3 different centers. Methods: To quantify the DSC perfusion signal two different mathematical modeling methods were used (Gamma fitting, leakage correction algorithms) considering the assumptions about the compartments contributing in the blood flow between the extra- and intra vascular space. Results: The Mean slope of increase (MSI) and the K1 parameter of the bidirectional exchange model exhibited the highest performance with (ACC 74.3% AUROC 74.2%) and (ACC 75% AUROC 70.5%) respectively. Conclusion: The proposed framework on DSC-MRI radiogenomics in gliomas has the potential of becoming a reliable diagnostic support tool exploiting the mathematical modeling of the DSC signal to characterize IDH mutation status through a more reproducible and standardized signal analysis scheme for facilitating clinical translation.

12.
Artículo en Inglés | MEDLINE | ID: mdl-38083519

RESUMEN

Digital histopathology image analysis of tumor tissue sections has seen great research interest for automating standard diagnostic tasks, but also for developing novel prognostic biomarkers. However, research has mainly been focused on developing uniresolution models, capturing either high-resolution cellular features or low-resolution tissue architectural features. In addition, in the patch-based weakly-supervised training of deep learning models, the features which represent the intratumoral heterogeneity are lost. In this study, we propose a multiresolution attention-based multiple instance learning framework that can capture cellular and contextual features from the whole tissue for predicting patient-level outcomes. Several basic mathematical operations were examined for integrating multiresolution features, i.e. addition, mean, multiplication and concatenation. The proposed multiplication-based multiresolution model performed the best (AUC=0.864), while all multiresolution models outperformed the uniresolution baseline models (AUC=0.669, 0.713) for breast-cancer grading. (Implementation: https://github.com/tsikup/multiresolution-clam).


Asunto(s)
Neoplasias de la Mama , Procesamiento de Imagen Asistido por Computador , Humanos , Femenino , Procesamiento de Imagen Asistido por Computador/métodos , Diagnóstico por Imagen , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/patología
13.
Biomedicines ; 11(12)2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38137400

RESUMEN

MRI is the modality of choice for a vast range of pathologies but also a sensitive probe into human physiology and tissue function. For this reason, several methodologies have been developed and continuously evolve in order to non-invasively monitor underlying phenomena in human adipose tissue that were difficult to assess in the past through visual inspection of standard imaging modalities. To this end, this work describes the imaging methodologies used in medical practice and lists the most important quantitative markers related to adipose tissue physiology and pathology that are currently supporting diagnosis, longitudinal evaluation and patient management decisions. The underlying physical principles and the resulting markers are presented and associated with frequently encountered pathologies in radiology in order to set the frame of the ability of MRI to reveal the complex role of adipose tissue, not as an inert tissue but as an active endocrine organ.

14.
Biomed Eng Online ; 22(1): 125, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38102586

RESUMEN

BACKGROUND: Multi-omics research has the potential to holistically capture intra-tumor variability, thereby improving therapeutic decisions by incorporating the key principles of precision medicine. The purpose of this study is to identify a robust method of integrating features from different sources, such as imaging, transcriptomics, and clinical data, to predict the survival and therapy response of non-small cell lung cancer patients. METHODS: 2996 radiomics, 5268 transcriptomics, and 8 clinical features were extracted from the NSCLC Radiogenomics dataset. Radiomics and deep features were calculated based on the volume of interest in pre-treatment, routine CT examinations, and then combined with RNA-seq and clinical data. Several machine learning classifiers were used to perform survival analysis and assess the patient's response to adjuvant chemotherapy. The proposed analysis was evaluated on an unseen testing set in a k-fold cross-validation scheme. Score- and concatenation-based multi-omics were used as feature integration techniques. RESULTS: Six radiomics (elongation, cluster shade, entropy, variance, gray-level non-uniformity, and maximal correlation coefficient), six deep features (NasNet-based activations), and three transcriptomics (OTUD3, SUCGL2, and RQCD1) were found to be significant for therapy response. The examined score-based multi-omic improved the AUC up to 0.10 on the unseen testing set (0.74 ± 0.06) and the balance between sensitivity and specificity for predicting therapy response for 106 patients, resulting in less biased models and improving upon the either highly sensitive or highly specific single-source models. Six radiomics (kurtosis, GLRLM- and GLSZM-based non-uniformity from images with no filtering, biorthogonal, and daubechies wavelets), seven deep features (ResNet-based activations), and seven transcriptomics (ELP3, ZZZ3, PGRMC2, TRAK1, ATIC, USP7, and PNPLA2) were found to be significant for the survival analysis. Accordingly, the survival analysis for 115 patients was also enhanced up to 0.20 by the proposed score-based multi-omics in terms of the C-index (0.79 ± 0.03). CONCLUSIONS: Compared to single-source models, multi-omics integration has the potential to improve prediction performance, increase model stability, and reduce bias for both treatment response and survival analysis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/genética , Entropía , Perfilación de la Expresión Génica , Aprendizaje Automático , Peptidasa Específica de Ubiquitina 7 , Proteasas Ubiquitina-Específicas
15.
Cancers (Basel) ; 15(13)2023 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-37444400

RESUMEN

Cardiotoxicity induced by breast cancer therapies is a potentially serious complication associated with the use of various breast cancer therapies. Prediction and better management of cardiotoxicity in patients receiving chemotherapy is of critical importance. However, the management of cancer therapy-related cardiac dysfunction (CTRCD) lacks clinical evidence and is based on limited clinical studies. AIM: To provide an overview of existing and potentially novel biomarkers that possess a promising predictive value for the early and late onset of CTRCD in the clinical setting. METHODS: A systematic review of published studies searching for promising biomarkers for the prediction of CTRCD in patients with breast cancer was undertaken according to PRISMA guidelines. A search strategy was performed using PubMed, Google Scholar, and Scopus for the period 2013-2023. All subjects were >18 years old, diagnosed with breast cancer, and received breast cancer therapies. RESULTS: The most promising biomarkers that can be used for the development of an alternative risk cardiac stratification plan for the prediction and/or early detection of CTRCD in patients with breast cancer were identified. CONCLUSIONS: We highlighted the new insights associated with the use of currently available biomarkers as a standard of care for the management of CTRCD and identified potentially novel clinical biomarkers that could be further investigated as promising predictors of CTRCD.

16.
J Med Internet Res ; 25: e43838, 2023 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-37307043

RESUMEN

BACKGROUND: Health professionals are often faced with the need to identify women at risk of manifesting poor psychological resilience following the diagnosis and treatment of breast cancer. Machine learning algorithms are increasingly used to support clinical decision support (CDS) tools in helping health professionals identify women who are at risk of adverse well-being outcomes and plan customized psychological interventions for women at risk. Clinical flexibility, cross-validated performance accuracy, and model explainability permitting person-specific identification of risk factors are highly desirable features of such tools. OBJECTIVE: This study aimed to develop and cross-validate machine learning models designed to identify breast cancer survivors at risk of poor overall mental health and global quality of life and identify potential targets of personalized psychological interventions according to an extensive set of clinical recommendations. METHODS: A set of 12 alternative models was developed to improve the clinical flexibility of the CDS tool. All models were validated using longitudinal data from a prospective, multicenter clinical pilot at 5 major oncology centers in 4 countries (Italy, Finland, Israel, and Portugal; the Predicting Effective Adaptation to Breast Cancer to Help Women to BOUNCE Back [BOUNCE] project). A total of 706 patients with highly treatable breast cancer were enrolled shortly after diagnosis and before the onset of oncological treatments and were followed up for 18 months. An extensive set of demographic, lifestyle, clinical, psychological, and biological variables measured within 3 months after enrollment served as predictors. Rigorous feature selection isolated key psychological resilience outcomes that could be incorporated into future clinical practice. RESULTS: Balanced random forest classifiers were successful at predicting well-being outcomes, with accuracies ranging between 78% and 82% (for 12-month end points after diagnosis) and between 74% and 83% (for 18-month end points after diagnosis). Explainability and interpretability analyses built on the best-performing models were used to identify potentially modifiable psychological and lifestyle characteristics that, if addressed systematically in the context of personalized psychological interventions, would be most likely to promote resilience for a given patient. CONCLUSIONS: Our results highlight the clinical utility of the BOUNCE modeling approach by focusing on resilience predictors that can be readily available to practicing clinicians at major oncology centers. The BOUNCE CDS tool paves the way for personalized risk assessment methods to identify patients at high risk of adverse well-being outcomes and direct valuable resources toward those most in need of specialized psychological interventions.


Asunto(s)
Neoplasias de la Mama , Sistemas de Apoyo a Decisiones Clínicas , Resiliencia Psicológica , Humanos , Femenino , Estudios Prospectivos , Calidad de Vida , Medición de Riesgo , Aprendizaje Automático
17.
Eur Radiol Exp ; 7(1): 20, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-37150779

RESUMEN

Artificial intelligence (AI) is transforming the field of medical imaging and has the potential to bring medicine from the era of 'sick-care' to the era of healthcare and prevention. The development of AI requires access to large, complete, and harmonized real-world datasets, representative of the population, and disease diversity. However, to date, efforts are fragmented, based on single-institution, size-limited, and annotation-limited datasets. Available public datasets (e.g., The Cancer Imaging Archive, TCIA, USA) are limited in scope, making model generalizability really difficult. In this direction, five European Union projects are currently working on the development of big data infrastructures that will enable European, ethically and General Data Protection Regulation-compliant, quality-controlled, cancer-related, medical imaging platforms, in which both large-scale data and AI algorithms will coexist. The vision is to create sustainable AI cloud-based platforms for the development, implementation, verification, and validation of trustable, usable, and reliable AI models for addressing specific unmet needs regarding cancer care provision. In this paper, we present an overview of the development efforts highlighting challenges and approaches selected providing valuable feedback to future attempts in the area.Key points• Artificial intelligence models for health imaging require access to large amounts of harmonized imaging data and metadata.• Main infrastructures adopted either collect centrally anonymized data or enable access to pseudonymized distributed data.• Developing a common data model for storing all relevant information is a challenge.• Trust of data providers in data sharing initiatives is essential.• An online European Union meta-tool-repository is a necessity minimizing effort duplication for the various projects in the area.


Asunto(s)
Inteligencia Artificial , Neoplasias , Humanos , Diagnóstico por Imagen , Predicción , Macrodatos
18.
Magn Reson Imaging ; 101: 1-12, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37004467

RESUMEN

Magnetic Resonance (MR) images suffer from spatial inhomogeneity, known as bias field corruption. The N4ITK filter is a state-of-the-art method used for correcting the bias field to optimize MR-based quantification. In this study, a novel approach is presented to quantitatively evaluate the performance of N4 bias field correction for pelvic prostate imaging. An exploratory analysis, regarding the different values of convergence threshold, shrink factor, fitting level, number of iterations and use of mask, is performed to quantify the performance of N4 filter in pelvic MR images. The performance of a total of 240 different N4 configurations is examined using the Full Width at Half Maximum (FWHM) of the segmented periprostatic fat distribution as evaluation metric. Phantom T2weighted images were used to assess the performance of N4 for a uniform test tissue mimicking material, excluding factors such as patient related susceptibility and anatomy heterogeneity. Moreover, 89 and 204 T2weighted patient images from two public datasets acquired by scanners with a combined surface and endorectal coil at 1.5 T and a surface coil at 3 T, respectively, were utilized and corrected with a variable set of N4 parameters. Furthermore, two external public datasets were used to validate the performance of the N4 filter in T2weighted patient images acquired by various scanning conditions with different magnetic field strengths and coils. The results show that the set of N4 parameters, converging to optimal representations of fat in the image, were: convergence threshold 0.001, shrink factor 2, fitting level 6, number of iterations 100 and the use of default mask for prostate images acquired by a combined surface and endorectal coil at both 1.5 T and 3 T. The corresponding optimal N4 configuration for MR prostate images acquired by a surface coil at 1.5 T or 3 T was: convergence threshold 0.001, shrink factor 2, fitting level 5, number of iterations 25 and the use of default mask. Hence, periprostatic fat segmentation can be used to define the optimal settings for achieving T2weighted prostate images free from bias field corruption to provide robust input for further analysis.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Próstata , Masculino , Humanos , Próstata/patología , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Sesgo , Fantasmas de Imagen
19.
Sci Rep ; 13(1): 7059, 2023 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-37120428

RESUMEN

Identifying individual patient characteristics that contribute to long-term mental health deterioration following diagnosis of breast cancer (BC) is critical in clinical practice. The present study employed a supervised machine learning pipeline to address this issue in a subset of data from a prospective, multinational cohort of women diagnosed with stage I-III BC with a curative treatment intention. Patients were classified as displaying stable HADS scores (Stable Group; n = 328) or reporting a significant increase in symptomatology between BC diagnosis and 12 months later (Deteriorated Group; n = 50). Sociodemographic, life-style, psychosocial, and medical variables collected on the first visit to their oncologist and three months later served as potential predictors of patient risk stratification. The flexible and comprehensive machine learning (ML) pipeline used entailed feature selection, model training, validation and testing. Model-agnostic analyses aided interpretation of model results at the variable- and patient-level. The two groups were discriminated with a high degree of accuracy (Area Under the Curve = 0.864) and a fair balance of sensitivity (0.85) and specificity (0.87). Both psychological (negative affect, certain coping with cancer reactions, lack of sense of control/positive expectations, and difficulties in regulating negative emotions) and biological variables (baseline percentage of neutrophils, thrombocyte count) emerged as important predictors of mental health deterioration in the long run. Personalized break-down profiles revealed the relative impact of specific variables toward successful model predictions for each patient. Identifying key risk factors for mental health deterioration is an essential first step toward prevention. Supervised ML models may guide clinical recommendations toward successful illness adaptation.


Asunto(s)
Neoplasias de la Mama , Salud Mental , Humanos , Femenino , Estudios Prospectivos , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/psicología , Algoritmos , Adaptación Psicológica
20.
Diagnostics (Basel) ; 13(4)2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36832225

RESUMEN

Radiotranscriptomics is an emerging field that aims to investigate the relationships between the radiomic features extracted from medical images and gene expression profiles that contribute in the diagnosis, treatment planning, and prognosis of cancer. This study proposes a methodological framework for the investigation of these associations with application on non-small-cell lung cancer (NSCLC). Six publicly available NSCLC datasets with transcriptomics data were used to derive and validate a transcriptomic signature for its ability to differentiate between cancer and non-malignant lung tissue. A publicly available dataset of 24 NSCLC-diagnosed patients, with both transcriptomic and imaging data, was used for the joint radiotranscriptomic analysis. For each patient, 749 Computed Tomography (CT) radiomic features were extracted and the corresponding transcriptomics data were provided through DNA microarrays. The radiomic features were clustered using the iterative K-means algorithm resulting in 77 homogeneous clusters, represented by meta-radiomic features. The most significant differentially expressed genes (DEGs) were selected by performing Significance Analysis of Microarrays (SAM) and 2-fold change. The interactions among the CT imaging features and the selected DEGs were investigated using SAM and a Spearman rank correlation test with a False Discovery Rate (FDR) of 5%, leading to the extraction of 73 DEGs significantly correlated with radiomic features. These genes were used to produce predictive models of the meta-radiomics features, defined as p-metaomics features, by performing Lasso regression. Of the 77 meta-radiomic features, 51 can be modeled in terms of the transcriptomic signature. These significant radiotranscriptomics relationships form a reliable basis to biologically justify the radiomics features extracted from anatomic imaging modalities. Thus, the biological value of these radiomic features was justified via enrichment analysis on their transcriptomics-based regression models, revealing closely associated biological processes and pathways. Overall, the proposed methodological framework provides joint radiotranscriptomics markers and models to support the connection and complementarities between the transcriptome and the phenotype in cancer, as demonstrated in the case of NSCLC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...