Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 149(12): 917-931, 2024 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-38314583

RESUMEN

BACKGROUND: Artificial intelligence-enhanced ECG analysis shows promise to detect ventricular dysfunction and remodeling in adult populations. However, its application to pediatric populations remains underexplored. METHODS: A convolutional neural network was trained on paired ECG-echocardiograms (≤2 days apart) from patients ≤18 years of age without major congenital heart disease to detect human expert-classified greater than mild left ventricular (LV) dysfunction, hypertrophy, and dilation (individually and as a composite outcome). Model performance was evaluated on single ECG-echocardiogram pairs per patient at Boston Children's Hospital and externally at Mount Sinai Hospital using area under the receiver operating characteristic curve (AUROC) and area under the precision-recall curve (AUPRC). RESULTS: The training cohort comprised 92 377 ECG-echocardiogram pairs (46 261 patients; median age, 8.2 years). Test groups included internal testing (12 631 patients; median age, 8.8 years; 4.6% composite outcomes), emergency department (2830 patients; median age, 7.7 years; 10.0% composite outcomes), and external validation (5088 patients; median age, 4.3 years; 6.1% composite outcomes) cohorts. Model performance was similar on internal test and emergency department cohorts, with model predictions of LV hypertrophy outperforming the pediatric cardiologist expert benchmark. Adding age and sex to the model added no benefit to model performance. When using quantitative outcome cutoffs, model performance was similar between internal testing (composite outcome: AUROC, 0.88, AUPRC, 0.43; LV dysfunction: AUROC, 0.92, AUPRC, 0.23; LV hypertrophy: AUROC, 0.88, AUPRC, 0.28; LV dilation: AUROC, 0.91, AUPRC, 0.47) and external validation (composite outcome: AUROC, 0.86, AUPRC, 0.39; LV dysfunction: AUROC, 0.94, AUPRC, 0.32; LV hypertrophy: AUROC, 0.84, AUPRC, 0.25; LV dilation: AUROC, 0.87, AUPRC, 0.33), with composite outcome negative predictive values of 99.0% and 99.2%, respectively. Saliency mapping highlighted ECG components that influenced model predictions (precordial QRS complexes for all outcomes; T waves for LV dysfunction). High-risk ECG features include lateral T-wave inversion (LV dysfunction), deep S waves in V1 and V2 and tall R waves in V6 (LV hypertrophy), and tall R waves in V4 through V6 (LV dilation). CONCLUSIONS: This externally validated algorithm shows promise to inexpensively screen for LV dysfunction and remodeling in children, which may facilitate improved access to care by democratizing the expertise of pediatric cardiologists.


Asunto(s)
Aprendizaje Profundo , Disfunción Ventricular Izquierda , Adulto , Humanos , Niño , Preescolar , Electrocardiografía , Inteligencia Artificial , Disfunción Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/diagnóstico por imagen
2.
Ann Intern Med ; 176(10): 1358-1369, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37812781

RESUMEN

BACKGROUND: Substantial effort has been directed toward demonstrating uses of predictive models in health care. However, implementation of these models into clinical practice may influence patient outcomes, which in turn are captured in electronic health record data. As a result, deployed models may affect the predictive ability of current and future models. OBJECTIVE: To estimate changes in predictive model performance with use through 3 common scenarios: model retraining, sequentially implementing 1 model after another, and intervening in response to a model when 2 are simultaneously implemented. DESIGN: Simulation of model implementation and use in critical care settings at various levels of intervention effectiveness and clinician adherence. Models were either trained or retrained after simulated implementation. SETTING: Admissions to the intensive care unit (ICU) at Mount Sinai Health System (New York, New York) and Beth Israel Deaconess Medical Center (Boston, Massachusetts). PATIENTS: 130 000 critical care admissions across both health systems. INTERVENTION: Across 3 scenarios, interventions were simulated at varying levels of clinician adherence and effectiveness. MEASUREMENTS: Statistical measures of performance, including threshold-independent (area under the curve) and threshold-dependent measures. RESULTS: At fixed 90% sensitivity, in scenario 1 a mortality prediction model lost 9% to 39% specificity after retraining once and in scenario 2 a mortality prediction model lost 8% to 15% specificity when created after the implementation of an acute kidney injury (AKI) prediction model; in scenario 3, models for AKI and mortality prediction implemented simultaneously, each led to reduced effective accuracy of the other by 1% to 28%. LIMITATIONS: In real-world practice, the effectiveness of and adherence to model-based recommendations are rarely known in advance. Only binary classifiers for tabular ICU admissions data were simulated. CONCLUSION: In simulated ICU settings, a universally effective model-updating approach for maintaining model performance does not seem to exist. Model use may have to be recorded to maintain viability of predictive modeling. PRIMARY FUNDING SOURCE: National Center for Advancing Translational Sciences.


Asunto(s)
Lesión Renal Aguda , Inteligencia Artificial , Humanos , Unidades de Cuidados Intensivos , Cuidados Críticos , Atención a la Salud
3.
J Am Soc Nephrol ; 32(1): 151-160, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32883700

RESUMEN

BACKGROUND: Early reports indicate that AKI is common among patients with coronavirus disease 2019 (COVID-19) and associated with worse outcomes. However, AKI among hospitalized patients with COVID-19 in the United States is not well described. METHODS: This retrospective, observational study involved a review of data from electronic health records of patients aged ≥18 years with laboratory-confirmed COVID-19 admitted to the Mount Sinai Health System from February 27 to May 30, 2020. We describe the frequency of AKI and dialysis requirement, AKI recovery, and adjusted odds ratios (aORs) with mortality. RESULTS: Of 3993 hospitalized patients with COVID-19, AKI occurred in 1835 (46%) patients; 347 (19%) of the patients with AKI required dialysis. The proportions with stages 1, 2, or 3 AKI were 39%, 19%, and 42%, respectively. A total of 976 (24%) patients were admitted to intensive care, and 745 (76%) experienced AKI. Of the 435 patients with AKI and urine studies, 84% had proteinuria, 81% had hematuria, and 60% had leukocyturia. Independent predictors of severe AKI were CKD, men, and higher serum potassium at admission. In-hospital mortality was 50% among patients with AKI versus 8% among those without AKI (aOR, 9.2; 95% confidence interval, 7.5 to 11.3). Of survivors with AKI who were discharged, 35% had not recovered to baseline kidney function by the time of discharge. An additional 28 of 77 (36%) patients who had not recovered kidney function at discharge did so on posthospital follow-up. CONCLUSIONS: AKI is common among patients hospitalized with COVID-19 and is associated with high mortality. Of all patients with AKI, only 30% survived with recovery of kidney function by the time of discharge.


Asunto(s)
Lesión Renal Aguda/etiología , COVID-19/complicaciones , SARS-CoV-2 , Lesión Renal Aguda/epidemiología , Lesión Renal Aguda/terapia , Lesión Renal Aguda/orina , Anciano , Anciano de 80 o más Años , COVID-19/mortalidad , Femenino , Hematuria/etiología , Mortalidad Hospitalaria , Hospitales Privados/estadística & datos numéricos , Hospitales Urbanos/estadística & datos numéricos , Humanos , Incidencia , Pacientes Internos , Leucocitos , Masculino , Persona de Mediana Edad , Ciudad de Nueva York/epidemiología , Proteinuria/etiología , Diálisis Renal , Estudios Retrospectivos , Resultado del Tratamiento , Orina/citología
4.
Europace ; 23(8): 1179-1191, 2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-33564873

RESUMEN

In the recent decade, deep learning, a subset of artificial intelligence and machine learning, has been used to identify patterns in big healthcare datasets for disease phenotyping, event predictions, and complex decision making. Public datasets for electrocardiograms (ECGs) have existed since the 1980s and have been used for very specific tasks in cardiology, such as arrhythmia, ischemia, and cardiomyopathy detection. Recently, private institutions have begun curating large ECG databases that are orders of magnitude larger than the public databases for ingestion by deep learning models. These efforts have demonstrated not only improved performance and generalizability in these aforementioned tasks but also application to novel clinical scenarios. This review focuses on orienting the clinician towards fundamental tenets of deep learning, state-of-the-art prior to its use for ECG analysis, and current applications of deep learning on ECGs, as well as their limitations and future areas of improvement.


Asunto(s)
Cardiología , Aprendizaje Profundo , Inteligencia Artificial , Electrocardiografía , Humanos , Aprendizaje Automático
5.
Blood Purif ; 50(4-5): 621-627, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33631752

RESUMEN

BACKGROUND/AIMS: Acute kidney injury (AKI) in critically ill patients is common, and continuous renal replacement therapy (CRRT) is a preferred mode of renal replacement therapy (RRT) in hemodynamically unstable patients. Prediction of clinical outcomes in patients on CRRT is challenging. We utilized several approaches to predict RRT-free survival (RRTFS) in critically ill patients with AKI requiring CRRT. METHODS: We used the Medical Information Mart for Intensive Care (MIMIC-III) database to identify patients ≥18 years old with AKI on CRRT, after excluding patients who had ESRD on chronic dialysis, and kidney transplantation. We defined RRTFS as patients who were discharged alive and did not require RRT ≥7 days prior to hospital discharge. We utilized all available biomedical data up to CRRT initiation. We evaluated 7 approaches, including logistic regression (LR), random forest (RF), support vector machine (SVM), adaptive boosting (AdaBoost), extreme gradient boosting (XGBoost), multilayer perceptron (MLP), and MLP with long short-term memory (MLP + LSTM). We evaluated model performance by using area under the receiver operating characteristic (AUROC) curves. RESULTS: Out of 684 patients with AKI on CRRT, 205 (30%) patients had RRTFS. The median age of patients was 63 years and their median Simplified Acute Physiology Score (SAPS) II was 67 (interquartile range 52-84). The MLP + LSTM showed the highest AUROC (95% CI) of 0.70 (0.67-0.73), followed by MLP 0.59 (0.54-0.64), LR 0.57 (0.52-0.62), SVM 0.51 (0.46-0.56), AdaBoost 0.51 (0.46-0.55), RF 0.44 (0.39-0.48), and XGBoost 0.43 (CI 0.38-0.47). CONCLUSIONS: A MLP + LSTM model outperformed other approaches for predicting RRTFS. Performance could be further improved by incorporating other data types.


Asunto(s)
Lesión Renal Aguda/terapia , Terapia de Reemplazo Renal , Lesión Renal Aguda/diagnóstico , Factores de Edad , Anciano , Cuidados Críticos , Femenino , Humanos , Modelos Logísticos , Aprendizaje Automático , Masculino , Persona de Mediana Edad , Pronóstico
6.
Kidney Int ; 98(5): 1323-1330, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32540406

RESUMEN

Urinary tract stones have high heritability indicating a strong genetic component. However, genome-wide association studies (GWAS) have uncovered only a few genome wide significant single nucleotide polymorphisms (SNPs). Polygenic risk scores (PRS) sum cumulative effect of many SNPs and shed light on underlying genetic architecture. Using GWAS summary statistics from 361,141 participants in the United Kingdom Biobank, we generated a PRS and determined association with stone diagnosis in 28,877 participants in the Mount Sinai BioMe Biobank. In BioMe (1,071 cases and 27,806 controls), for every standard deviation increase, we observed a significant increment in adjusted odds ratio of a factor of 1.2 (95% confidence interval 1.13-1.26). In comparison, a risk score comprised of GWAS significant SNPs was not significantly associated with diagnosis. After stratifying individuals into low and high-risk categories on clinical risk factors, there was a significant increment in adjusted odds ratio of 1.3 (1.12-1.6) in the low- and 1.2 (1.1-1.2) in the high-risk group for every standard deviation increment in PRS. In a 14,348-participant validation cohort (Penn Medicine Biobank), every standard deviation increment was associated with a significant adjusted odds ratio of 1.1 (1.03 - 1.2). Thus, a genome-wide PRS is associated with urinary tract stones overall and in the absence of known clinical risk factors and illustrates their complex polygenic architecture.


Asunto(s)
Estudio de Asociación del Genoma Completo , Cálculos Urinarios , Predisposición Genética a la Enfermedad , Humanos , Herencia Multifactorial , Polimorfismo de Nucleótido Simple , Reino Unido/epidemiología
7.
Curr Opin Nephrol Hypertens ; 29(3): 319-326, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32235273

RESUMEN

PURPOSE OF REVIEW: The universal adoption of electronic health records, improvement in technology, and the availability of continuous monitoring has generated large quantities of healthcare data. Machine learning is increasingly adopted by nephrology researchers to analyze this data in order to improve the care of their patients. RECENT FINDINGS: In this review, we provide a broad overview of the different types of machine learning algorithms currently available and how researchers have applied these methods in nephrology research. Current applications have included prediction of acute kidney injury and chronic kidney disease along with progression of kidney disease. Researchers have demonstrated the ability of machine learning to read kidney biopsy samples, identify patient outcomes from unstructured data, and identify subtypes in complex diseases. We end with a discussion on the ethics and potential pitfalls of machine learning. SUMMARY: Machine learning provides researchers with the ability to analyze data that were previously inaccessible. While still burgeoning, several studies show promising results, which will enable researchers to perform larger scale studies and clinicians the ability to provide more personalized care. However, we must ensure that implementation aids providers and does not lead to harm to patients.


Asunto(s)
Enfermedades Renales/terapia , Aprendizaje Automático , Algoritmos , Humanos , Procesamiento de Lenguaje Natural , Investigación Biomédica Traslacional
8.
J Med Internet Res ; 22(11): e24018, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33027032

RESUMEN

BACKGROUND: COVID-19 has infected millions of people worldwide and is responsible for several hundred thousand fatalities. The COVID-19 pandemic has necessitated thoughtful resource allocation and early identification of high-risk patients. However, effective methods to meet these needs are lacking. OBJECTIVE: The aims of this study were to analyze the electronic health records (EHRs) of patients who tested positive for COVID-19 and were admitted to hospitals in the Mount Sinai Health System in New York City; to develop machine learning models for making predictions about the hospital course of the patients over clinically meaningful time horizons based on patient characteristics at admission; and to assess the performance of these models at multiple hospitals and time points. METHODS: We used Extreme Gradient Boosting (XGBoost) and baseline comparator models to predict in-hospital mortality and critical events at time windows of 3, 5, 7, and 10 days from admission. Our study population included harmonized EHR data from five hospitals in New York City for 4098 COVID-19-positive patients admitted from March 15 to May 22, 2020. The models were first trained on patients from a single hospital (n=1514) before or on May 1, externally validated on patients from four other hospitals (n=2201) before or on May 1, and prospectively validated on all patients after May 1 (n=383). Finally, we established model interpretability to identify and rank variables that drive model predictions. RESULTS: Upon cross-validation, the XGBoost classifier outperformed baseline models, with an area under the receiver operating characteristic curve (AUC-ROC) for mortality of 0.89 at 3 days, 0.85 at 5 and 7 days, and 0.84 at 10 days. XGBoost also performed well for critical event prediction, with an AUC-ROC of 0.80 at 3 days, 0.79 at 5 days, 0.80 at 7 days, and 0.81 at 10 days. In external validation, XGBoost achieved an AUC-ROC of 0.88 at 3 days, 0.86 at 5 days, 0.86 at 7 days, and 0.84 at 10 days for mortality prediction. Similarly, the unimputed XGBoost model achieved an AUC-ROC of 0.78 at 3 days, 0.79 at 5 days, 0.80 at 7 days, and 0.81 at 10 days. Trends in performance on prospective validation sets were similar. At 7 days, acute kidney injury on admission, elevated LDH, tachypnea, and hyperglycemia were the strongest drivers of critical event prediction, while higher age, anion gap, and C-reactive protein were the strongest drivers of mortality prediction. CONCLUSIONS: We externally and prospectively trained and validated machine learning models for mortality and critical events for patients with COVID-19 at different time horizons. These models identified at-risk patients and uncovered underlying relationships that predicted outcomes.


Asunto(s)
Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/mortalidad , Aprendizaje Automático/normas , Neumonía Viral/diagnóstico , Neumonía Viral/mortalidad , Lesión Renal Aguda/epidemiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Betacoronavirus , COVID-19 , Estudios de Cohortes , Registros Electrónicos de Salud , Femenino , Mortalidad Hospitalaria , Hospitalización/estadística & datos numéricos , Hospitales , Humanos , Masculino , Persona de Mediana Edad , Ciudad de Nueva York/epidemiología , Pandemias , Pronóstico , Curva ROC , Medición de Riesgo/métodos , Medición de Riesgo/normas , SARS-CoV-2 , Adulto Joven
9.
Artículo en Inglés | MEDLINE | ID: mdl-38687616

RESUMEN

OBJECTIVES: The study developed framework that leverages an open-source Large Language Model (LLM) to enable clinicians to ask plain-language questions about a patient's entire echocardiogram report history. This approach is intended to streamline the extraction of clinical insights from multiple echocardiogram reports, particularly in patients with complex cardiac diseases, thereby enhancing both patient care and research efficiency. MATERIALS AND METHODS: Data from over 10 years were collected, comprising echocardiogram reports from patients with more than 10 echocardiograms on file at the Mount Sinai Health System. These reports were converted into a single document per patient for analysis, broken down into snippets and relevant snippets were retrieved using text similarity measures. The LLaMA-2 70B model was employed for analyzing the text using a specially crafted prompt. The model's performance was evaluated against ground-truth answers created by faculty cardiologists. RESULTS: The study analyzed 432 reports from 37 patients for a total of 100 question-answer pairs. The LLM correctly answered 90% questions, with accuracies of 83% for temporality, 93% for severity assessment, 84% for intervention identification, and 100% for diagnosis retrieval. Errors mainly stemmed from the LLM's inherent limitations, such as misinterpreting numbers or hallucinations. CONCLUSION: The study demonstrates the feasibility and effectiveness of using a local, open-source LLM for querying and interpreting echocardiogram report data. This approach offers a significant improvement over traditional keyword-based searches, enabling more contextually relevant and semantically accurate responses; in turn showing promise in enhancing clinical decision-making and research by facilitating more efficient access to complex patient data.

10.
medRxiv ; 2024 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-38405776

RESUMEN

Timely and accurate assessment of electrocardiograms (ECGs) is crucial for diagnosing, triaging, and clinically managing patients. Current workflows rely on a computerized ECG interpretation using rule-based tools built into the ECG signal acquisition systems with limited accuracy and flexibility. In low-resource settings, specialists must review every single ECG for such decisions, as these computerized interpretations are not available. Additionally, high-quality interpretations are even more essential in such low-resource settings as there is a higher burden of accuracy for automated reads when access to experts is limited. Artificial Intelligence (AI)-based systems have the prospect of greater accuracy yet are frequently limited to a narrow range of conditions and do not replicate the full diagnostic range. Moreover, these models often require raw signal data, which are unavailable to physicians and necessitate costly technical integrations that are currently limited. To overcome these challenges, we developed and validated a format-independent vision encoder-decoder model - ECG-GPT - that can generate free-text, expert-level diagnosis statements directly from ECG images. The model shows robust performance, validated on 2.6 million ECGs across 6 geographically distinct health settings: (1) 2 large and diverse US health systems- Yale-New Haven and Mount Sinai Health Systems, (2) a consecutive ECG dataset from a central ECG repository from Minas Gerais, Brazil, (3) the prospective cohort study, UK Biobank, (4) a Germany-based, publicly available repository, PTB-XL, and (5) a community hospital in Missouri. The model demonstrated consistently high performance (AUROC≥0.81) across a wide range of rhythm and conduction disorders. This can be easily accessed via a web-based application capable of receiving ECG images and represents a scalable and accessible strategy for generating accurate, expert-level reports from images of ECGs, enabling accurate triage of patients globally, especially in low-resource settings.

11.
Artículo en Inglés | MEDLINE | ID: mdl-38771093

RESUMEN

BACKGROUND: Artificial intelligence (AI) and large language models (LLMs) can play a critical role in emergency room operations by augmenting decision-making about patient admission. However, there are no studies for LLMs using real-world data and scenarios, in comparison to and being informed by traditional supervised machine learning (ML) models. We evaluated the performance of GPT-4 for predicting patient admissions from emergency department (ED) visits. We compared performance to traditional ML models both naively and when informed by few-shot examples and/or numerical probabilities. METHODS: We conducted a retrospective study using electronic health records across 7 NYC hospitals. We trained Bio-Clinical-BERT and XGBoost (XGB) models on unstructured and structured data, respectively, and created an ensemble model reflecting ML performance. We then assessed GPT-4 capabilities in many scenarios: through Zero-shot, Few-shot with and without retrieval-augmented generation (RAG), and with and without ML numerical probabilities. RESULTS: The Ensemble ML model achieved an area under the receiver operating characteristic curve (AUC) of 0.88, an area under the precision-recall curve (AUPRC) of 0.72 and an accuracy of 82.9%. The naïve GPT-4's performance (0.79 AUC, 0.48 AUPRC, and 77.5% accuracy) showed substantial improvement when given limited, relevant data to learn from (ie, RAG) and underlying ML probabilities (0.87 AUC, 0.71 AUPRC, and 83.1% accuracy). Interestingly, RAG alone boosted performance to near peak levels (0.82 AUC, 0.56 AUPRC, and 81.3% accuracy). CONCLUSIONS: The naïve LLM had limited performance but showed significant improvement in predicting ED admissions when supplemented with real-world examples to learn from, particularly through RAG, and/or numerical probabilities from traditional ML models. Its peak performance, although slightly lower than the pure ML model, is noteworthy given its potential for providing reasoning behind predictions. Further refinement of LLMs with real-world data is necessary for successful integration as decision-support tools in care settings.

12.
J Am Heart Assoc ; 13(1): e031671, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38156471

RESUMEN

BACKGROUND: Right ventricular ejection fraction (RVEF) and end-diastolic volume (RVEDV) are not readily assessed through traditional modalities. Deep learning-enabled ECG analysis for estimation of right ventricular (RV) size or function is unexplored. METHODS AND RESULTS: We trained a deep learning-ECG model to predict RV dilation (RVEDV >120 mL/m2), RV dysfunction (RVEF ≤40%), and numerical RVEDV and RVEF from a 12-lead ECG paired with reference-standard cardiac magnetic resonance imaging volumetric measurements in UK Biobank (UKBB; n=42 938). We fine-tuned in a multicenter health system (MSHoriginal [Mount Sinai Hospital]; n=3019) with prospective validation over 4 months (MSHvalidation; n=115). We evaluated performance with area under the receiver operating characteristic curve for categorical and mean absolute error for continuous measures overall and in key subgroups. We assessed the association of RVEF prediction with transplant-free survival with Cox proportional hazards models. The prevalence of RV dysfunction for UKBB/MSHoriginal/MSHvalidation cohorts was 1.0%/18.0%/15.7%, respectively. RV dysfunction model area under the receiver operating characteristic curve for UKBB/MSHoriginal/MSHvalidation cohorts was 0.86/0.81/0.77, respectively. The prevalence of RV dilation for UKBB/MSHoriginal/MSHvalidation cohorts was 1.6%/10.6%/4.3%. RV dilation model area under the receiver operating characteristic curve for UKBB/MSHoriginal/MSHvalidation cohorts was 0.91/0.81/0.92, respectively. MSHoriginal mean absolute error was RVEF=7.8% and RVEDV=17.6 mL/m2. The performance of the RVEF model was similar in key subgroups including with and without left ventricular dysfunction. Over a median follow-up of 2.3 years, predicted RVEF was associated with adjusted transplant-free survival (hazard ratio, 1.40 for each 10% decrease; P=0.031). CONCLUSIONS: Deep learning-ECG analysis can identify significant cardiac magnetic resonance imaging RV dysfunction and dilation with good performance. Predicted RVEF is associated with clinical outcome.


Asunto(s)
Disfunción Ventricular Derecha , Función Ventricular Derecha , Humanos , Volumen Sistólico , Imagen por Resonancia Magnética/métodos , Corazón , Electrocardiografía
13.
medRxiv ; 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38559021

RESUMEN

Background: Point-of-care ultrasonography (POCUS) enables cardiac imaging at the bedside and in communities but is limited by abbreviated protocols and variation in quality. We developed and tested artificial intelligence (AI) models to automate the detection of underdiagnosed cardiomyopathies from cardiac POCUS. Methods: In a development set of 290,245 transthoracic echocardiographic videos across the Yale-New Haven Health System (YNHHS), we used augmentation approaches and a customized loss function weighted for view quality to derive a POCUS-adapted, multi-label, video-based convolutional neural network (CNN) that discriminates HCM (hypertrophic cardiomyopathy) and ATTR-CM (transthyretin amyloid cardiomyopathy) from controls without known disease. We evaluated the final model across independent, internal and external, retrospective cohorts of individuals who underwent cardiac POCUS across YNHHS and Mount Sinai Health System (MSHS) emergency departments (EDs) (2011-2024) to prioritize key views and validate the diagnostic and prognostic performance of single-view screening protocols. Findings: We identified 33,127 patients (median age 61 [IQR: 45-75] years, n=17,276 [52·2%] female) at YNHHS and 5,624 (57 [IQR: 39-71] years, n=1,953 [34·7%] female) at MSHS with 78,054 and 13,796 eligible cardiac POCUS videos, respectively. An AI-enabled single-view screening approach successfully discriminated HCM (AUROC of 0·90 [YNHHS] & 0·89 [MSHS]) and ATTR-CM (YNHHS: AUROC of 0·92 [YNHHS] & 0·99 [MSHS]). In YNHHS, 40 (58·0%) HCM and 23 (47·9%) ATTR-CM cases had a positive screen at median of 2·1 [IQR: 0·9-4·5] and 1·9 [IQR: 1·0-3·4] years before clinical diagnosis. Moreover, among 24,448 participants without known cardiomyopathy followed over 2·2 [IQR: 1·1-5·8] years, AI-POCUS probabilities in the highest (vs lowest) quintile for HCM and ATTR-CM conferred a 15% (adj.HR 1·15 [95%CI: 1·02-1·29]) and 39% (adj.HR 1·39 [95%CI: 1·22-1·59]) higher age- and sex-adjusted mortality risk, respectively. Interpretation: We developed and validated an AI framework that enables scalable, opportunistic screening of treatable cardiomyopathies wherever POCUS is used. Funding: National Heart, Lung and Blood Institute, Doris Duke Charitable Foundation, BridgeBio.

14.
medRxiv ; 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38352556

RESUMEN

Importance: Increased intracranial pressure (ICP) is associated with adverse neurological outcomes, but needs invasive monitoring. Objective: Development and validation of an AI approach for detecting increased ICP (aICP) using only non-invasive extracranial physiological waveform data. Design: Retrospective diagnostic study of AI-assisted detection of increased ICP. We developed an AI model using exclusively extracranial waveforms, externally validated it and assessed associations with clinical outcomes. Setting: MIMIC-III Waveform Database (2000-2013), a database derived from patients admitted to an ICU in an academic Boston hospital, was used for development of the aICP model, and to report association with neurologic outcomes. Data from Mount Sinai Hospital (2020-2022) in New York City was used for external validation. Participants: Patients were included if they were older than 18 years, and were monitored with electrocardiograms, arterial blood pressure, respiratory impedance plethysmography and pulse oximetry. Patients who additionally had intracranial pressure monitoring were used for development (N=157) and external validation (N=56). Patients without intracranial monitors were used for association with outcomes (N=1694). Exposures: Extracranial waveforms including electrocardiogram, arterial blood pressure, plethysmography and SpO2. Main Outcomes and Measures: Intracranial pressure > 15 mmHg. Measures were Area under receiver operating characteristic curves (AUROCs), sensitivity, specificity, and accuracy at threshold of 0.5. We calculated odds ratios and p-values for phenotype association. Results: The AUROC was 0.91 (95% CI, 0.90-0.91) on testing and 0.80 (95% CI, 0.80-0.80) on external validation. aICP had accuracy, sensitivity, and specificity of 73.8% (95% CI, 72.0%-75.6%), 99.5% (95% CI 99.3%-99.6%), and 76.9% (95% CI, 74.0-79.8%) on external validation. A ten-percentile increment was associated with stroke (OR=2.12; 95% CI, 1.27-3.13), brain malignancy (OR=1.68; 95% CI, 1.09-2.60), subdural hemorrhage (OR=1.66; 95% CI, 1.07-2.57), intracerebral hemorrhage (OR=1.18; 95% CI, 1.07-1.32), and procedures like percutaneous brain biopsy (OR=1.58; 95% CI, 1.15-2.18) and craniotomy (OR = 1.43; 95% CI, 1.12-1.84; P < 0.05 for all). Conclusions and Relevance: aICP provides accurate, non-invasive estimation of increased ICP, and is associated with neurological outcomes and neurosurgical procedures in patients without intracranial monitoring.

15.
Sci Rep ; 13(1): 16492, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37779171

RESUMEN

The United States Medical Licensing Examination (USMLE) has been a subject of performance study for artificial intelligence (AI) models. However, their performance on questions involving USMLE soft skills remains unexplored. This study aimed to evaluate ChatGPT and GPT-4 on USMLE questions involving communication skills, ethics, empathy, and professionalism. We used 80 USMLE-style questions involving soft skills, taken from the USMLE website and the AMBOSS question bank. A follow-up query was used to assess the models' consistency. The performance of the AI models was compared to that of previous AMBOSS users. GPT-4 outperformed ChatGPT, correctly answering 90% compared to ChatGPT's 62.5%. GPT-4 showed more confidence, not revising any responses, while ChatGPT modified its original answers 82.5% of the time. The performance of GPT-4 was higher than that of AMBOSS's past users. Both AI models, notably GPT-4, showed capacity for empathy, indicating AI's potential to meet the complex interpersonal, ethical, and professional demands intrinsic to the practice of medicine.


Asunto(s)
Inteligencia Artificial , Medicina , Empatía , Procesos Mentales
16.
Commun Med (Lond) ; 3(1): 24, 2023 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-36788316

RESUMEN

BACKGROUND: Aortic Stenosis and Mitral Regurgitation are common valvular conditions representing a hidden burden of disease within the population. The aim of this study was to develop and validate deep learning-based screening and diagnostic tools that can help guide clinical decision making. METHODS: In this multi-center retrospective cohort study, we acquired Transthoracic Echocardiogram reports from five Mount Sinai hospitals within New York City representing a demographically diverse cohort of patients. We developed a Natural Language Processing pipeline to extract ground-truth labels about valvular status and paired these to Electrocardiograms (ECGs). We developed and externally validated deep learning models capable of detecting valvular disease, in addition to considering scenarios of clinical deployment. RESULTS: We use 617,338 ECGs paired to transthoracic echocardiograms from 123,096 patients to develop a deep learning model for detection of Mitral Regurgitation. Area Under Receiver Operating Characteristic curve (AUROC) is 0.88 (95% CI:0.88-0.89) in internal testing, and 0.81 (95% CI:0.80-0.82) in external validation. To develop a model for detection of Aortic Stenosis, we use 617,338 Echo-ECG pairs for 128,628 patients. AUROC is 0.89 (95% CI: 0.88-0.89) in internal testing, going to 0.86 (95% CI: 0.85-0.87) in external validation. The model's performance increases leading up to the time of the diagnostic echo, and it performs well in validation against requirement of Transcatheter Aortic Valve Replacement procedures. CONCLUSIONS: Deep learning based tools can increase the amount of information extracted from ubiquitous investigations such as the ECG. Such tools are inexpensive, can help in earlier disease detection, and potentially improve prognosis.


The valves of the heart have flaps that open and close when the heart beats to maintain the flow of blood in the correct direction. Valvular disease, such as backflow or narrowing, puts additional strain upon heart muscles which can lead to heart failure. Usually, these conditions are diagnosed by doing an echocardiogram, an ultrasound scan of the heart and nearby blood vessels. The electrocardiogram (ECG) records the electrical signal generated by the heart and can be obtained more easily. We used deep learning neural networks, self-learning computer algorithms which excel at finding patterns within complex data. This enabled us to develop computer software able to diagnose valvular disease from ECGs. Earlier detection of such disease can help in improving overall outcome, while also reducing costs related to treatment.

17.
JACC Clin Electrophysiol ; 9(8 Pt 2): 1437-1451, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37480862

RESUMEN

BACKGROUND: Premature ventricular complexes (PVCs) are prevalent and, although often benign, they may lead to PVC-induced cardiomyopathy. We created a deep-learning algorithm to predict left ventricular ejection fraction (LVEF) reduction in patients with PVCs from a 12-lead electrocardiogram (ECG). OBJECTIVES: This study aims to assess a deep-learning model to predict cardiomyopathy among patients with PVCs. METHODS: We used electronic medical records from 5 hospitals and identified ECGs from adults with documented PVCs. Internal training and testing were performed at one hospital. External validation was performed with the others. The primary outcome was first diagnosis of LVEF ≤40% within 6 months. The dataset included 383,514 ECGs, of which 14,241 remained for analysis. We analyzed area under the receiver operating curves and explainability plots for representative patients, algorithm prediction, PVC burden, and demographics in a multivariable Cox model to assess independent predictors for cardiomyopathy. RESULTS: Among the 14,241-patient cohort (age 67.6 ± 14.8 years; female 43.8%; White 29.5%, Black 8.6%, Hispanic 6.5%, Asian 2.2%), 22.9% experienced reductions in LVEF to ≤40% within 6 months. The model predicted reductions in LVEF to ≤40% with area under the receiver operating curve of 0.79 (95% CI: 0.77-0.81). The gradient weighted class activation map explainability framework highlighted the sinus rhythm QRS complex-ST segment. In patients who underwent successful PVC ablation there was a post-ablation improvement in LVEF with resolution of cardiomyopathy in most (89%) patients. CONCLUSIONS: Deep-learning on the 12-lead ECG alone can accurately predict new-onset cardiomyopathy in patients with PVCs independent of PVC burden. Model prediction performed well across sex and race, relying on the QRS complex/ST-segment in sinus rhythm, not PVC morphology.


Asunto(s)
Cardiomiopatías , Aprendizaje Profundo , Complejos Prematuros Ventriculares , Adulto , Humanos , Femenino , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Volumen Sistólico , Función Ventricular Izquierda , Complejos Prematuros Ventriculares/diagnóstico , Complejos Prematuros Ventriculares/cirugía , Algoritmos , Cardiomiopatías/complicaciones , Cardiomiopatías/diagnóstico , Electrocardiografía
18.
NPJ Digit Med ; 6(1): 108, 2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-37280346

RESUMEN

The electrocardiogram (ECG) is a ubiquitous diagnostic modality. Convolutional neural networks (CNNs) applied towards ECG analysis require large sample sizes, and transfer learning approaches for biomedical problems may result in suboptimal performance when pre-training is done on natural images. We leveraged masked image modeling to create a vision-based transformer model, HeartBEiT, for electrocardiogram waveform analysis. We pre-trained this model on 8.5 million ECGs and then compared performance vs. standard CNN architectures for diagnosis of hypertrophic cardiomyopathy, low left ventricular ejection fraction and ST elevation myocardial infarction using differing training sample sizes and independent validation datasets. We find that HeartBEiT has significantly higher performance at lower sample sizes compared to other models. We also find that HeartBEiT improves explainability of diagnosis by highlighting biologically relevant regions of the EKG vs. standard CNNs. Domain specific pre-trained transformer models may exceed the classification performance of models trained on natural images especially in very low data regimes. The combination of the architecture and such pre-training allows for more accurate, granular explainability of model predictions.

19.
medRxiv ; 2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-37162979

RESUMEN

Background: Right ventricular ejection fraction (RVEF) and end-diastolic volume (RVEDV) are not readily assessed through traditional modalities. Deep-learning enabled 12-lead electrocardiogram analysis (DL-ECG) for estimation of RV size or function is unexplored. Methods: We trained a DL-ECG model to predict RV dilation (RVEDV>120 mL/m2), RV dysfunction (RVEF≤40%), and numerical RVEDV/RVEF from 12-lead ECG paired with reference-standard cardiac MRI (cMRI) volumetric measurements in UK biobank (UKBB; n=42,938). We fine-tuned in a multi-center health system (MSHoriginal; n=3,019) with prospective validation over 4 months (MSHvalidation; n=115). We evaluated performance using area under the receiver operating curve (AUROC) for categorical and mean absolute error (MAE) for continuous measures overall and in key subgroups. We assessed association of RVEF prediction with transplant-free survival with Cox proportional hazards models. Results: Prevalence of RV dysfunction for UKBB/MSHoriginal/MSHvalidation cohorts was 1.0%/18.0%/15.7%, respectively. RV dysfunction model AUROC for UKBB/MSHoriginal/MSHvalidation cohorts was 0.86/0.81/0.77, respectively. Prevalence of RV dilation for UKBB/MSHoriginal/MSHvalidation cohorts was 1.6%/10.6%/4.3%. RV dilation model AUROC for UKBB/MSHoriginal/MSHvalidation cohorts 0.91/0.81/0.92, respectively. MSHoriginal MAE was RVEF=7.8% and RVEDV=17.6 ml/m2. Performance was similar in key subgroups including with and without left ventricular dysfunction. Over median follow-up of 2.3 years, predicted RVEF was independently associated with composite outcome (HR 1.37 for each 10% decrease, p=0.046). Conclusions: DL-ECG analysis can accurately identify significant RV dysfunction and dilation both overall and in key subgroups. Predicted RVEF is independently associated with clinical outcome.

20.
JACC Clin Electrophysiol ; 9(8 Pt 3): 1804-1815, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37354170

RESUMEN

BACKGROUND: Interatrial block (IAB) is associated with thromboembolism and atrial arrhythmias. However, prior studies included small patient cohorts so it remains unclear whether IAB predicts adverse outcomes particularly in context of atrial fibrillation (AF)/atrial flutter (AFL). OBJECTIVES: This study sought to determine whether IAB portends increased stroke risk in a large cohort in the presence or absence of AFAF/AFL. METHODS: We performed a 5-center retrospective analysis of 4,837,989 electrocardiograms (ECGs) from 1,228,291 patients. IAB was defined as P-wave duration ≥120 ms in leads II, III, or aVF. Measurements were extracted as .XML files. After excluding patients with prior AF/AFL, 1,825,958 ECGs from 458,994 patients remained. Outcomes were analyzed using restricted mean survival time analysis and restricted mean time lost. RESULTS: There were 86,317 patients with IAB and 355,032 patients without IAB. IAB prevalence in the cohort was 19.6% and was most common in Black (26.1%), White (20.9%), and Hispanic (18.5%) patients and least prevalent in Native Americans (9.2%). IAB was independently associated with increased stroke probability (restricted mean time lost ratio coefficient [RMTLRC]: 1.43; 95% CI: 1.35-1.51; tau = 1,895), mortality (RMTLRC: 1.14; 95% CI: 1.07-1.21; tau = 1,924), heart failure (RMTLRC: 1.94; 95% CI: 1.83-2.04; tau = 1,921), systemic thromboembolism (RMTLRC: 1.62; 95% CI: 1.53-1.71; tau = 1,897), and incident AF/AFL (RMTLRC: 1.16; 95% CI: 1.10-1.22; tau = 1,888). IAB was not associated with stroke in patients with pre-existing AF/AFL. CONCLUSIONS: IAB is independently associated with stroke in patients with no history of AF/AFL even after adjustment for incident AF/AFL and CHA2DS2-VASc score. Patients are at increased risk of stroke even when AF/AFL is not identified.


Asunto(s)
Fibrilación Atrial , Aleteo Atrial , Accidente Cerebrovascular , Tromboembolia , Humanos , Fibrilación Atrial/complicaciones , Fibrilación Atrial/epidemiología , Bloqueo Interauricular/complicaciones , Bloqueo Interauricular/epidemiología , Estudios Retrospectivos , Electrocardiografía , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/etiología , Aleteo Atrial/complicaciones , Aleteo Atrial/epidemiología , Tromboembolia/epidemiología , Tromboembolia/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA