Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 220
Filtrar
1.
Radiol Artif Intell ; : e230348, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38900042

RESUMEN

"Just Accepted" papers have undergone full peer review and have been accepted for publication in Radiology: Artificial Intelligence. This article will undergo copyediting, layout, and proof review before it is published in its final version. Please note that during production of the final copyedited article, errors may be discovered which could affect the content. Purpose To determine whether time-dependent deep learning models can outperform single timepoint models in predicting preoperative upgrade of ductal carcinoma in situ (DCIS) to invasive malignancy on dynamic contrastenhanced (DCE) breast MRI without lesion segmentation prerequisite. Materials and Methods In this exploratory study, 154 cases of biopsy-proven DCIS (25 upgraded at surgery and 129 not upgraded) were selected consecutively from a retrospective cohort of preoperative DCE MRI in women with an average age of 58.6 years at time of diagnosis from 2012 to 2022. Binary classification was implemented with convolutional neural network-long short-term memory (CNN-LSTM) architectures benchmarked against traditional CNNs without manual segmentation of the lesions. Combinatorial performance analysis of ResNet50 versus VGG16-based models was performed with each contrast phase. Binary classification area under the receiver operating characteristic curve (AUC) was reported. Results VGG16-based models consistently provided better hold-out test AUCs than ResNet50 in CNN and CNNLSTM studies (multiphase test AUC: 0.67 versus 0.59, respectively, for CNN models; P = .04 and 0.73 versus 0.62 for CNN-LSTM models; P = .008). The time-dependent model (CNN-LSTM) provided a better multiphase test AUC over single-timepoint (CNN) models (0.73 versus 0.67, P = .04). Conclusion Compared with single-timepoint architectures, sequential deep learning algorithms using preoperative DCE MRI improved prediction of DCIS lesions upgraded to invasive malignancy without the need for lesion segmentation. ©RSNA, 2024.

2.
Radiother Oncol ; 197: 110345, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38838989

RESUMEN

BACKGROUND AND PURPOSE: Artificial Intelligence (AI) models in radiation therapy are being developed with increasing pace. Despite this, the radiation therapy community has not widely adopted these models in clinical practice. A cohesive guideline on how to develop, report and clinically validate AI algorithms might help bridge this gap. METHODS AND MATERIALS: A Delphi process with all co-authors was followed to determine which topics should be addressed in this comprehensive guideline. Separate sections of the guideline, including Statements, were written by subgroups of the authors and discussed with the whole group at several meetings. Statements were formulated and scored as highly recommended or recommended. RESULTS: The following topics were found most relevant: Decision making, image analysis, volume segmentation, treatment planning, patient specific quality assurance of treatment delivery, adaptive treatment, outcome prediction, training, validation and testing of AI model parameters, model availability for others to verify, model quality assurance/updates and upgrades, ethics. Key references were given together with an outlook on current hurdles and possibilities to overcome these. 19 Statements were formulated. CONCLUSION: A cohesive guideline has been written which addresses main topics regarding AI in radiation therapy. It will help to guide development, as well as transparent and consistent reporting and validation of new AI tools and facilitate adoption.

3.
medRxiv ; 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38746238

RESUMEN

Background: Adaptive treatment strategies that can dynamically react to individual cancer progression can provide effective personalized care. Longitudinal multi-omics information, paired with an artificially intelligent clinical decision support system (AI-CDSS) can assist clinicians in determining optimal therapeutic options and treatment adaptations. However, AI-CDSS is not perfectly accurate, as such, clinicians' over/under reliance on AI may lead to unintended consequences, ultimately failing to develop optimal strategies. To investigate such collaborative decision-making process, we conducted a Human-AI interaction case study on response-adaptive radiotherapy (RT). Methods: We designed and conducted a two-phase study for two disease sites and two treatment modalities-adaptive RT for non-small cell lung cancer (NSCLC) and adaptive stereotactic body RT for hepatocellular carcinoma (HCC)-in which clinicians were asked to consider mid-treatment modification of the dose per fraction for a number of retrospective cancer patients without AI-support (Unassisted Phase) and with AI-assistance (AI-assisted Phase). The AI-CDSS graphically presented trade-offs in tumor control and the likelihood of toxicity to organs at risk, provided an optimal recommendation, and associated model uncertainties. In addition, we asked for clinicians' decision confidence level and trust level in individual AI recommendations and encouraged them to provide written remarks. We enrolled 13 evaluators (radiation oncology physicians and residents) from two medical institutions located in two different states, out of which, 4 evaluators volunteered in both NSCLC and HCC studies, resulting in a total of 17 completed evaluations (9 NSCLC, and 8 HCC). To limit the evaluation time to under an hour, we selected 8 treated patients for NSCLC and 9 for HCC, resulting in a total of 144 sets of evaluations (72 from NSCLC and 72 from HCC). Evaluation for each patient consisted of 8 required inputs and 2 optional remarks, resulting in up to a total of 1440 data points. Results: AI-assistance did not homogeneously influence all experts and clinical decisions. From NSCLC cohort, 41 (57%) decisions and from HCC cohort, 34 (47%) decisions were adjusted after AI assistance. Two evaluations (12%) from the NSCLC cohort had zero decision adjustments, while the remaining 15 (88%) evaluations resulted in at least two decision adjustments. Decision adjustment level positively correlated with dissimilarity in decision-making with AI [NSCLC: ρ = 0.53 ( p < 0.001); HCC: ρ = 0.60 ( p < 0.001)] indicating that evaluators adjusted their decision closer towards AI recommendation. Agreement with AI-recommendation positively correlated with AI Trust Level [NSCLC: ρ = 0.59 ( p < 0.001); HCC: ρ = 0.7 ( p < 0.001)] indicating that evaluators followed AI's recommendation if they agreed with that recommendation. The correlation between decision confidence changes and decision adjustment level showed an opposite trend [NSCLC: ρ = -0.24 ( p = 0.045), HCC: ρ = 0.28 ( p = 0.017)] reflecting the difference in behavior due to underlying differences in disease type and treatment modality. Decision confidence positively correlated with the closeness of decisions to the standard of care (NSCLC: 2 Gy/fx; HCC: 10 Gy/fx) indicating that evaluators were generally more confident in prescribing dose fractionations more similar to those used in standard clinical practice. Inter-evaluator agreement increased with AI-assistance indicating that AI-assistance can decrease inter-physician variability. The majority of decisions were adjusted to achieve higher tumor control in NSCLC and lower normal tissue complications in HCC. Analysis of evaluators' remarks indicated concerns for organs at risk and RT outcome estimates as important decision-making factors. Conclusions: Human-AI interaction depends on the complex interrelationship between expert's prior knowledge and preferences, patient's state, disease site, treatment modality, model transparency, and AI's learned behavior and biases. The collaborative decision-making process can be summarized as follows: (i) some clinicians may not believe in an AI system, completely disregarding its recommendation, (ii) some clinicians may believe in the AI system but will critically analyze its recommendations on a case-by-case basis; (iii) when a clinician finds that the AI recommendation indicates the possibility for better outcomes they will adjust their decisions accordingly; and (iv) When a clinician finds that the AI recommendation indicate a worse possible outcome they will disregard it and seek their own alternative approach.

4.
iScience ; 27(4): 109614, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38632985

RESUMEN

Virtually all cells use energy-driven, ion-specific membrane pumps to maintain large transmembrane gradients of Na+, K+, Cl-, Mg++, and Ca++, but the corresponding evolutionary benefit remains unclear. We propose that these gradients enable a dynamic and versatile biological system that acquires, analyzes, and responds to environmental information. We hypothesize that environmental signals are transmitted into the cell by ion fluxes along pre-existing gradients through gated ion-specific membrane channels. The consequent changes in cytoplasmic ion concentration can generate a local response or orchestrate global/regional cellular dynamics through wire-like ion fluxes along pre-existing and self-assembling cytoskeleton to engage the endoplasmic reticulum, mitochondria, and nucleus.

5.
Cancers (Basel) ; 16(1)2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38201633

RESUMEN

BACKGROUND: While multiple cyst features are evaluated for stratifying pancreatic intraductal papillary mucinous neoplasms (IPMN), cyst size is an important factor that can influence treatment strategies. When magnetic resonance imaging (MRI) is used to evaluate IPMNs, no universally accepted sequence provides optimal size measurements. T2-weighted coronal/axial have been suggested as primary measurement sequences; however, it remains unknown how well these and maximum all-sequence diameter measurements correlate with pathology size. This study aims to compare agreement and bias between IPMN long-axis measurements on seven commonly obtained MRI sequences with pathologic size measurements. METHODS: This retrospective cohort included surgically resected IPMN cases with preoperative MRI exams. Long-axis diameter tumor measurements and the presence of worrisome features and/orhigh-risk stigmata were noted on all seven MRI sequences. MRI size and pathology agreement and MRI inter-observer agreement involved concordance correlation coefficient (CCC) and intraclass correlation coefficient (ICC), respectively. The presence of worrisome features and high-risk stigmata were compared to the tumor grade using kappa analysis. The Bland-Altman analysis assessed the systematic bias between MRI-size and pathology. RESULTS: In 52 patients (age 68 ± 13 years, 22 males), MRI sequences produced mean long-axis tumor measurements from 2.45-2.65 cm. The maximum MRI lesion size had a strong agreement with pathology (CCC = 0.82 (95% CI: 0.71-0.89)). The maximum IPMN size was typically observed on the axial T1 arterial post-contrast and MRCP coronal series and overestimated size versus pathology with bias +0.34 cm. The radiologist interobserver agreement reached ICCs 0.74 to 0.91 on the MRI sequences. CONCLUSION: The maximum MRI IPMN size strongly correlated with but tended to overestimate the length compared to the pathology, potentially related to formalin tissue shrinkage during tissue processing.

6.
Phys Imaging Radiat Oncol ; 28: 100505, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38045642

RESUMEN

Background and purpose: Diffusion weighted imaging (DWI) allows for the interrogation of tissue cellularity, which is a surrogate for cellular proliferation. Previous attempts to incorporate DWI into the workflow of a 0.35 T MR-linac (MRL) have lacked quantitative accuracy. In this study, accuracy, repeatability, and geometric precision of apparent diffusion coefficient (ADC) maps produced using an echo planar imaging (EPI)-based DWI protocol on the MRL system is illustrated, and in vivo potential for longitudinal patient imaging is demonstrated. Materials and methods: Accuracy and repeatability were assessed by measuring ADC values in a diffusion phantom at three timepoints and comparing to reference ADC values. System-dependent geometric distortion was quantified by measuring the distance between 93 pairs of phantom features on ADC maps acquired on a 0.35 T MRL and a 3.0 T diagnostic scanner and comparing to spatially precise CT images. Additionally, for five sarcoma patients receiving radiotherapy on the MRL, same-day in vivo ADC maps were acquired on both systems, one of which at multiple timepoints. Results: Phantom ADC quantification was accurate on the 0.35 T MRL with significant discrepancies only seen at high ADC. Average geometric distortions were 0.35 (±0.02) mm and 0.85 (±0.02) mm in the central slice and 0.66 (±0.04) mm and 2.14 (±0.07) mm at 5.4 cm off-center for the MRL and diagnostic system, respectively. In the sarcoma patients, a mean pretreatment ADC of 910x10-6 (±100x10-6) mm2/s was measured on the MRL. Conclusions: The acquisition of accurate, repeatable, and geometrically precise ADC maps is possible at 0.35 T with an EPI approach.

7.
Artículo en Inglés | MEDLINE | ID: mdl-38056778

RESUMEN

PURPOSE: Non-small cell lung cancer (NSCLC) stereotactic body radiation therapy with 50 Gy/5 fractions is sometimes considered controversial, as the nominal biologically effective dose (BED) of 100 Gy is felt by some to be insufficient for long-term local control of some lesions. In this study, we analyzed such patients using explainable deep learning techniques and consequently proposed appropriate treatment planning criteria. These novel criteria could help planners achieve optimized treatment plans for maximal local control. METHODS AND MATERIALS: A total of 535 patients treated with 50 Gy/5 fractions were used to develop a novel deep learning local response model. A multimodality approach, incorporating computed tomography images, 3-dimensional dose distribution, and patient demographics, combined with a discrete-time survival model, was applied to predict time to failure and the probability of local control. Subsequently, an integrated gradient-weighted class activation mapping method was used to identify the most significant dose-volume metrics predictive of local failure and their optimal cut-points. RESULTS: The model was cross-validated, showing an acceptable performance (c-index: 0.72, 95% CI, 0.68-0.75); the testing c-index was 0.69. The model's spatial attention was concentrated mostly in the tumors' periphery (planning target volume [PTV] - internal gross target volume [IGTV]) region. Statistically significant dose-volume metrics in improved local control were BED Dnear-min ≥ 103.8 Gy in IGTV (hazard ratio [HR], 0.31; 95% CI, 015-0.63), V104 ≥ 98% in IGTV (HR, 0.30; 95% CI, 0.15-0.60), gEUD ≥ 103.8 Gy in PTV-IGTV (HR, 0.25; 95% CI, 0.12-0.50), and Dmean ≥ 104.5 Gy in PTV-IGTV (HR, 0.25; 95% CI, 0.12-0.51). CONCLUSIONS: Deep learning-identified dose-volume metrics have shown significant prognostic power (log-rank, P = .003) and could be used as additional actionable criteria for treatment planning in NSCLC stereotactic body radiation therapy patients receiving 50 Gy in 5 fractions. Although our data do not confirm or refute that a significantly higher BED for the prescription dose is necessary for tumor control in NSCLC, it might be clinically effective to escalate the nominal prescribed dose from BED 100 to 105 Gy.

8.
BJR Open ; 5(1): 20220023, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37953865

RESUMEN

Novel and developing artificial intelligence (AI) systems can be integrated into healthcare settings in numerous ways. For example, in the case of automated image classification and natural language processing, AI systems are beginning to demonstrate near expert level performance in detecting abnormalities such as seizure activity. This paper, however, focuses on AI integration into clinical trials. During the clinical trial recruitment process, considerable labor and time is spent sifting through electronic health record and interviewing patients. With the advancement of deep learning techniques such as natural language processing, intricate electronic health record data can be efficiently processed. This provides utility to workflows such as recruitment for clinical trials. Studies are starting to show promise in shortening the time to recruitment and reducing workload for those involved in clinical trial design. Additionally, numerous guidelines are being constructed to encourage integration of AI into the healthcare setting with meaningful impact. The goal would be to improve the clinical trial process by reducing bias in patient composition, improving retention of participants, and lowering costs and labor.

9.
Cancers (Basel) ; 15(20)2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37894280

RESUMEN

BACKGROUND: We aimed to determine the prognostic value of an immunoscore reflecting CD3+ and CD8+ T cell density estimated from real-world transcriptomic data of a patient cohort with advanced malignancies treated with immune checkpoint inhibitors (ICIs) in an effort to validate a reference for future machine learning-based biomarker development. METHODS: Transcriptomic data was collected under the Total Cancer Care Protocol (NCT03977402) Avatar® project. The real-world immunoscore for each patient was calculated based on the estimated densities of tumor CD3+ and CD8+ T cells utilizing CIBERSORTx and the LM22 gene signature matrix. Then, the immunoscore association with overall survival (OS) was estimated using Cox regression and analyzed using Kaplan-Meier curves. The OS predictions were assessed using Harrell's concordance index (C-index). The Youden index was used to identify the optimal cut-off point. Statistical significance was assessed using the log-rank test. RESULTS: Our study encompassed 522 patients with four cancer types. The median duration to death was 10.5 months for the 275 participants who encountered an event. For the entire cohort, the results demonstrated that transcriptomics-based immunoscore could significantly predict patients at risk of death (p-value < 0.001). Notably, patients with an intermediate-high immunoscore achieved better OS than those with a low immunoscore. In subgroup analysis, the prediction of OS was significant for melanoma and head and neck cancer patients but did not reach significance in the non-small cell lung cancer or renal cell carcinoma cohorts. CONCLUSIONS: Calculating CD3+ and CD8+ T cell immunoscore using real-world transcriptomic data represents a promising signature for estimating OS with ICIs and can be used as a reference for future machine learning-based biomarker development.

10.
Br J Radiol ; 96(1150): 20230211, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37660402

RESUMEN

Multiomics data including imaging radiomics and various types of molecular biomarkers have been increasingly investigated for better diagnosis and therapy in the era of precision oncology. Artificial intelligence (AI) including machine learning (ML) and deep learning (DL) techniques combined with the exponential growth of multiomics data may have great potential to revolutionize cancer subtyping, risk stratification, prognostication, prediction and clinical decision-making. In this article, we first present different categories of multiomics data and their roles in diagnosis and therapy. Second, AI-based data fusion methods and modeling methods as well as different validation schemes are illustrated. Third, the applications and examples of multiomics research in oncology are demonstrated. Finally, the challenges regarding the heterogeneity data set, availability of omics data, and validation of the research are discussed. The transition of multiomics research to real clinics still requires consistent efforts in standardizing omics data collection and analysis, building computational infrastructure for data sharing and storing, developing advanced methods to improve data fusion and interpretability, and ultimately, conducting large-scale prospective clinical trials to fill the gap between study findings and clinical benefits.


Asunto(s)
Inteligencia Artificial , Neoplasias , Humanos , Neoplasias/diagnóstico por imagen , Neoplasias/genética , Multiómica , Estudios Prospectivos , Medicina de Precisión , Aprendizaje Automático
11.
Oncogene ; 42(42): 3089-3097, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37684407

RESUMEN

Artificial intelligence (AI) is a transformative technology that is capturing popular imagination and can revolutionize biomedicine. AI and machine learning (ML) algorithms have the potential to break through existing barriers in oncology research and practice such as automating workflow processes, personalizing care, and reducing healthcare disparities. Emerging applications of AI/ML in the literature include screening and early detection of cancer, disease diagnosis, response prediction, prognosis, and accelerated drug discovery. Despite this excitement, only few AI/ML models have been properly validated and fewer have become regulated products for routine clinical use. In this review, we highlight the main challenges impeding AI/ML clinical translation. We present different clinical use cases from the domains of radiology, radiation oncology, immunotherapy, and drug discovery in oncology. We dissect the unique challenges and opportunities associated with each of these cases. Finally, we summarize the general requirements for successful AI/ML implementation in the clinic, highlighting specific examples and points of emphasis including the importance of multidisciplinary collaboration of stakeholders, role of domain experts in AI augmentation, transparency of AI/ML models, and the establishment of a comprehensive quality assurance program to mitigate risks of training bias and data drifts, all culminating toward safer and more beneficial AI/ML applications in oncology labs and clinics.

12.
JNCI Cancer Spectr ; 7(6)2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37738580

RESUMEN

BACKGROUND: Randomized clinical trials of novel treatments for solid tumors normally measure disease progression using the Response Evaluation Criteria in Solid Tumors. However, novel, scalable approaches to estimate disease progression using real-world data are needed to advance cancer outcomes research. The purpose of this narrative review is to summarize examples from the existing literature on approaches to estimate real-world disease progression and their relative strengths and limitations, using lung cancer as a case study. METHODS: A narrative literature review was conducted in PubMed to identify articles that used approaches to estimate real-world disease progression in lung cancer patients. Data abstracted included data source, approach used to estimate real-world progression, and comparison to a selected gold standard (if applicable). RESULTS: A total of 40 articles were identified from 2008 to 2022. Five approaches to estimate real-world disease progression were identified including manual abstraction of medical records, natural language processing of clinical notes and/or radiology reports, treatment-based algorithms, changes in tumor volume, and delta radiomics-based approaches. The accuracy of these progression approaches were assessed using different methods, including correlations between real-world endpoints and overall survival for manual abstraction (Spearman rank ρ = 0.61-0.84) and area under the curve for natural language processing approaches (area under the curve = 0.86-0.96). CONCLUSIONS: Real-world disease progression has been measured in several observational studies of lung cancer. However, comparing the accuracy of methods across studies is challenging, in part, because of the lack of a gold standard and the different methods used to evaluate accuracy. Concerted efforts are needed to define a gold standard and quality metrics for real-world data.


Asunto(s)
Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/terapia , Evaluación de Resultado en la Atención de Salud , Progresión de la Enfermedad
13.
Lab Invest ; 103(11): 100255, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37757969

RESUMEN

Digital pathology has transformed the traditional pathology practice of analyzing tissue under a microscope into a computer vision workflow. Whole-slide imaging allows pathologists to view and analyze microscopic images on a computer monitor, enabling computational pathology. By leveraging artificial intelligence (AI) and machine learning (ML), computational pathology has emerged as a promising field in recent years. Recently, task-specific AI/ML (eg, convolutional neural networks) has risen to the forefront, achieving above-human performance in many image-processing and computer vision tasks. The performance of task-specific AI/ML models depends on the availability of many annotated training datasets, which presents a rate-limiting factor for AI/ML development in pathology. Task-specific AI/ML models cannot benefit from multimodal data and lack generalization, eg, the AI models often struggle to generalize to new datasets or unseen variations in image acquisition, staining techniques, or tissue types. The 2020s are witnessing the rise of foundation models and generative AI. A foundation model is a large AI model trained using sizable data, which is later adapted (or fine-tuned) to perform different tasks using a modest amount of task-specific annotated data. These AI models provide in-context learning, can self-correct mistakes, and promptly adjust to user feedback. In this review, we provide a brief overview of recent advances in computational pathology enabled by task-specific AI, their challenges and limitations, and then introduce various foundation models. We propose to create a pathology-specific generative AI based on multimodal foundation models and present its potentially transformative role in digital pathology. We describe different use cases, delineating how it could serve as an expert companion of pathologists and help them efficiently and objectively perform routine laboratory tasks, including quantifying image analysis, generating pathology reports, diagnosis, and prognosis. We also outline the potential role that foundation models and generative AI can play in standardizing the pathology laboratory workflow, education, and training.


Asunto(s)
Inteligencia Artificial , Aprendizaje Automático , Patología , Humanos , Procesamiento de Imagen Asistido por Computador , Redes Neurales de la Computación , Patólogos , Patología/tendencias
14.
Br J Radiol ; 96(1150): 20230142, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37493248

RESUMEN

Artificial intelligence has been introduced to clinical practice, especially radiology and radiation oncology, from image segmentation, diagnosis, treatment planning and prognosis. It is not only crucial to have an accurate artificial intelligence model, but also to understand the internal logic and gain the trust of the experts. This review is intended to provide some insights into core concepts of the interpretability, the state-of-the-art methods for understanding the machine learning models, the evaluation of these methods, identifying some challenges and limits of them, and gives some examples of medical applications.


Asunto(s)
Oncología por Radiación , Radiología , Humanos , Inteligencia Artificial , Radiología/métodos , Aprendizaje Automático , Radiografía
16.
Eur J Nucl Med Mol Imaging ; 50(10): 2984-2996, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37171633

RESUMEN

PURPOSE: Metastatic neuroendocrine tumors (NETs) overexpressing type 2 somatostatin receptors are the target for peptide receptor radionuclide therapy (PRRT) through the theragnostic pair of 68Ga/177Lu-DOTATATE. The main purpose of this study was to develop machine learning models to predict therapeutic tumor dose using pre therapy 68Ga -PET and clinicopathological biomarkers. METHODS: We retrospectively analyzed 90 segmented metastatic NETs from 25 patients (M14/F11, age 63.7 ± 9.5, range 38-76) treated by 177Lu-DOTATATE at our institute. Patients underwent both pretherapy [68Ga]Ga-DOTA-TATE PET/CT and four timepoints SPECT/CT at ~ 4, 24, 96, and 168 h post-177Lu-DOTATATE infusion. Tumors were segmented by a radiologist on baseline CT or MRI and transferred to co-registered PET/CT and SPECT/CT, and normal organs were segmented by deep learning-based method on CT of the PET and SPECT. The SUV metrics and tumor-to-normal tissue SUV ratios (SUV_TNRs) were calculated from 68Ga -PET at the contour-level. Posttherapy dosimetry was performed based on the co-registration of SPECT/CTs to generate time-integrated-activity, followed by an in-house Monte Carlo-based absorbed dose estimation. The correlation between delivered 177Lu Tumor absorbed dose and PET-derived metrics along with baseline clinicopathological biomarkers (such as Creatinine, Chromogranin A and prior therapies) were evaluated. Multiple interpretable machine-learning algorithms were developed to predict tumor dose using these pretherapy information. Model performance on a nested tenfold cross-validation was evaluated in terms of coefficient of determination (R2), mean-absolute-error (MAE), and mean-relative-absolute-error (MRAE). RESULTS: SUVmean showed a significant correlation (q-value < 0.05) with absorbed dose (Spearman ρ = 0.64), followed by TLSUVmean (SUVmean of total-lesion-burden) and SUVpeak (ρ = 0.45 and 0.41, respectively). The predictive value of PET-SUVmean in estimation of posttherapy absorbed dose was stronger compared to PET-SUVpeak, and SUV_TNRs in terms of univariate analysis (R2 = 0.28 vs. R2 ≤ 0.12). An optimal trivariate random forest model composed of SUVmean, TLSUVmean, and total liver SUVmean (normal and tumoral liver) provided the best performance in tumor dose prediction with R2 = 0.64, MAE = 0.73 Gy/GBq, and MRAE = 0.2. CONCLUSION: Our preliminary results demonstrate the feasibility of using baseline PET images for prediction of absorbed dose prior to 177Lu-PRRT. Machine learning models combining multiple PET-based metrics performed better than using a single SUV value and using other investigated clinicopathological biomarkers. Developing such quantitative models forms the groundwork for the role of 68Ga -PET not only for the implementation of personalized treatment planning but also for patient stratification in the era of precision medicine.


Asunto(s)
Tumores Neuroendocrinos , Compuestos Organometálicos , Humanos , Persona de Mediana Edad , Anciano , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Radioisótopos de Galio , Octreótido/uso terapéutico , Estudios Retrospectivos , Compuestos Organometálicos/uso terapéutico , Tumores Neuroendocrinos/diagnóstico por imagen , Tumores Neuroendocrinos/radioterapia , Tumores Neuroendocrinos/tratamiento farmacológico , Biomarcadores
17.
Int J Radiat Oncol Biol Phys ; 117(3): 581-593, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37150258

RESUMEN

PURPOSE: Brain radiation therapy can impair fine motor skills (FMS). Fine motor skills are essential for activities of daily living, enabling hand-eye coordination for manipulative movements. We developed normal tissue complication probability (NTCP) models for the decline in FMS after fractionated brain radiation therapy (RT). METHODS AND MATERIALS: On a prospective trial, 44 patients with primary brain tumors received fractioned RT; underwent high-resolution volumetric magnetic resonance imaging, diffusion tensor imaging, and comprehensive FMS assessments (Delis-Kaplan Executive Function System Trail Making Test Motor Speed [DKEFS-MS]; and Grooved Pegboard dominant/nondominant hands) at baseline and 6 months postRT. Regions of interest subserving motor function (including cortex, superficial white matter, thalamus, basal ganglia, cerebellum, and white matter tracts) were autosegmented using validated methods and manually verified. Dosimetric and clinical variables were included in multivariate NTCP models using automated bootstrapped logistic regression, least absolute shrinkage and selection operator logistic regression, and random forests with nested cross-validation. RESULTS: Half of the patients showed a decline on grooved pegboard test of nondominant hands, 17 of 42 (40.4%) on grooved pegboard test of -dominant hands, and 11 of 44 (25%) on DKEFS-MS. Automated bootstrapped logistic regression selected a 1-term model including maximum dose to dominant postcentral white matter. The least absolute shrinkage and selection operator logistic regression selected this term and steroid use. The top 5 variables in the random forest were all dosimetric: maximum dose to dominant thalamus, mean dose to dominant caudate, mean and maximum dose to the dominant corticospinal tract, and maximum dose to dominant postcentral white matter. This technique performed best with an area under the curve of 0.69 (95% CI, 0.68-0.70) on nested cross-validation. CONCLUSIONS: We present the first NTCP models for FMS impairment after brain RT. Dose to several supratentorial motor-associated regions of interest correlated with a decline in dominant-hand fine motor dexterity in patients with primary brain tumors in multivariate models, outperforming clinical variables. These data can guide prospective fine motor-sparing strategies for brain RT.


Asunto(s)
Neoplasias Encefálicas , Sustancia Blanca , Humanos , Imagen de Difusión Tensora/métodos , Destreza Motora , Estudios Prospectivos , Actividades Cotidianas , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/patología , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/patología , Probabilidad
18.
Br J Radiol ; 96(1150): 20220934, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37191072

RESUMEN

Artificial intelligence (AI) and its machine learning (ML) algorithms are offering new promise for personalized biomedicine and more cost-effective healthcare with impressive technical capability to mimic human cognitive capabilities. However, widespread application of this promising technology has been limited in the medical domain and expectations have been tampered by ethical challenges and concerns regarding patient privacy, legal responsibility, trustworthiness, and fairness. To balance technical innovation with ethical applications of AI/ML, developers must demonstrate the AI functions as intended and adopt strategies to minimize the risks for failure or bias. This review describes the new ethical challenges created by AI/ML for clinical care and identifies specific considerations for its practice in medicine. We provide an overview of regulatory and legal issues applicable in Europe and the United States, a description of technical aspects to consider, and present recommendations for trustworthy AI/ML that promote transparency, minimize risks of bias or error, and protect the patient well-being.


Asunto(s)
Inteligencia Artificial , Medicina , Humanos , Aprendizaje Automático , Algoritmos , Europa (Continente)
19.
Sci Rep ; 13(1): 5279, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-37002296

RESUMEN

Involvement of many variables, uncertainty in treatment response, and inter-patient heterogeneity challenge objective decision-making in dynamic treatment regime (DTR) in oncology. Advanced machine learning analytics in conjunction with information-rich dense multi-omics data have the ability to overcome such challenges. We have developed a comprehensive artificial intelligence (AI)-based optimal decision-making framework for assisting oncologists in DTR. In this work, we demonstrate the proposed framework to Knowledge Based Response-Adaptive Radiotherapy (KBR-ART) applications by developing an interactive software tool entitled Adaptive Radiotherapy Clinical Decision Support (ARCliDS). ARCliDS is composed of two main components: Artifcial RT Environment (ARTE) and Optimal Decision Maker (ODM). ARTE is designed as a Markov decision process and modeled via supervised learning. Given a patient's pre- and during-treatment information, ARTE can estimate treatment outcomes for a selected daily dosage value (radiation fraction size). ODM is formulated using reinforcement learning and is trained on ARTE. ODM can recommend optimal daily dosage adjustments to maximize the tumor local control probability and minimize the side effects. Graph Neural Networks (GNN) are applied to exploit the inter-feature relationships for improved modeling performance and a novel double GNN architecture is designed to avoid nonphysical treatment response. Datasets of size 117 and 292 were available from two clinical trials on adaptive RT in non-small cell lung cancer (NSCLC) patients and adaptive stereotactic body RT (SBRT) in hepatocellular carcinoma (HCC) patients, respectively. For training and validation, dense data with 297 features were available for 67 NSCLC patients and 110 features for 71 HCC patients. To increase the sample size for ODM training, we applied Generative Adversarial Networks to generate 10,000 synthetic patients. The ODM was trained on the synthetic patients and validated on the original dataset. We found that, Double GNN architecture was able to correct the nonphysical dose-response trend and improve ARCliDS recommendation. The average root mean squared difference (RMSD) between ARCliDS recommendation and reported clinical decisions using double GNNs were 0.61 [0.03] Gy/frac (mean [sem]) for adaptive RT in NSCLC patients and 2.96 [0.42] Gy/frac for adaptive SBRT HCC compared to the single GNN's RMSDs of 0.97 [0.12] Gy/frac and 4.75 [0.16] Gy/frac, respectively. Overall, For NSCLC and HCC, ARCliDS with double GNNs was able to reproduce 36% and 50% of the good clinical decisions (local control and no side effects) and improve 74% and 30% of the bad clinical decisions, respectively. In conclusion, ARCliDS is the first web-based software dedicated to assist KBR-ART with multi-omics data. ARCliDS can learn from the reported clinical decisions and facilitate AI-assisted clinical decision-making for improving the outcomes in DTR.


Asunto(s)
Carcinoma Hepatocelular , Carcinoma de Pulmón de Células no Pequeñas , Sistemas de Apoyo a Decisiones Clínicas , Neoplasias Hepáticas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Carcinoma de Pulmón de Células no Pequeñas/patología , Inteligencia Artificial , Neoplasias Pulmonares/patología , Neoplasias Hepáticas/radioterapia , Dosificación Radioterapéutica
20.
Nat Biotechnol ; 41(8): 1160-1167, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36593414

RESUMEN

Ionizing radiation acoustic imaging (iRAI) allows online monitoring of radiation's interactions with tissues during radiation therapy, providing real-time, adaptive feedback for cancer treatments. We describe an iRAI volumetric imaging system that enables mapping of the three-dimensional (3D) radiation dose distribution in a complex clinical radiotherapy treatment. The method relies on a two-dimensional matrix array transducer and a matching multi-channel preamplifier board. The feasibility of imaging temporal 3D dose accumulation was first validated in a tissue-mimicking phantom. Next, semiquantitative iRAI relative dose measurements were verified in vivo in a rabbit model. Finally, real-time visualization of the 3D radiation dose delivered to a patient with liver metastases was accomplished with a clinical linear accelerator. These studies demonstrate the potential of iRAI to monitor and quantify the 3D radiation dose deposition during treatment, potentially improving radiotherapy treatment efficacy using real-time adaptive treatment.


Asunto(s)
Neoplasias , Planificación de la Radioterapia Asistida por Computador , Conejos , Animales , Planificación de la Radioterapia Asistida por Computador/métodos , Diagnóstico por Imagen , Hígado/diagnóstico por imagen , Dosis de Radiación , Neoplasias/diagnóstico por imagen , Neoplasias/radioterapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...