Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
2.
JACC Adv ; 3(9): 100890, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39372468

RESUMO

Background: Loneliness and social isolation are associated with poor health outcomes such as an increased risk of cardiovascular diseases. Objectives: The authors aimed to explore the association between social isolation with biological aging which was determined by artificial intelligence-enabled electrocardiography (AI-ECG) as well as the risk of all-cause mortality. Methods: The study included adults aged ≥18 years seen at Mayo Clinic from 2019 to 2022 who respond to a survey for social isolation assessment and had a 12-lead ECG within 1 year of completing the questionnaire. Biological age was determined from ECGs using a previously developed and validated convolutional neural network (AI-ECG age). Age-Gap was defined as AI-ECG age minus chronological age, where positive values reflect an older-than-expected age. The status of social isolation was measured by the previously validated multiple-choice questions based on Social Network Index (SNI) with score ranges between 0 (most isolated) and 4 (least isolated). Results: A total of 280,324 subjects were included (chronological age 59.8 ± 16.4 years, 50.9% female). The mean Age-Gap was -0.2 ± 9.16 years. A higher SNI was associated with a lower Age-Gap (ß of SNI = 4 was -0.11; 95% CI: -0.22 to -0.01; P < 0.001, adjusted to covariates). Cox proportional hazard analysis revealed the association between social connection and all-cause mortality (HR for SNI = 4, 0.47; 95% CI: 0.43-0.5; P < 0.001). Conclusions: Social isolation is associated with accelerating biological aging and all-cause mortality independent of conventional cardiovascular risk factors. This observation underscores the need to address social connection as a health care determinant.

3.
JACC Adv ; 3(9): 101169, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39372474

RESUMO

Background: Hyperkalemia has been associated with increased mortality in cardiac intensive care unit (CICU) patients. An artificial intelligence (AI) enhanced electrocardiogram (ECG) can predict hyperkalemia, and other AI-ECG algorithms have demonstrated mortality risk-stratification in CICU patients. Objectives: The authors hypothesized that the AI-ECG hyperkalemia algorithm could stratify mortality risk beyond laboratory serum potassium measurement alone. Methods: We included 11,234 unique Mayo Clinic CICU patients admitted from 2007 to 2018 with a 12-lead ECG and blood potassium (K) level obtained at admission with K ≥5 mEq/L defining hyperkalemia. ECGs underwent AI evaluation for the probability of hyperkalemia (probability >0.5 defined as positive). Hospital mortality was analyzed using logistic regression, and survival to 1 year was estimated using Kaplan-Meier and Cox analysis. Results: In the final cohort (n = 11,234), the mean age was 69.6 ± 10.5 years, 37.8% were females, and 92.4% were White. Chronic kidney disease was present in 20.2%. The mean laboratory potassium value for the cohort was 4.2 ± 0.3 mEq/L. The AI-ECG predicted hyperkalemia in 33.9% (n = 3,810) of CICU patients and 12.9% (n = 1,451) of patients had laboratory-confirmed hyperkalemia (K ≥5 mEq/L). In-hospital mortality increased in false-positive, false-negative, and true-positive patients, respectively (P < 0.001), and each of these patient groups had successively lower survival out to 1 year. Conclusions: AI-ECG-based prediction of hyperkalemia, even with a normal laboratory potassium value, was associated with higher in-hospital mortality and lower 1-year survival in CICU patients. This study demonstrated that AI-ECG probability of hyperkalemia may enable rapid individualized risk stratification in critically ill patients beyond laboratory value alone.

4.
JACC Adv ; 3(9): 101179, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39372476

RESUMO

Background: An artificial intelligence (AI)-based electrocardiogram (ECG) model identifies patients with a higher likelihood of low ejection fraction (EF). Patients with an abnormal AI-ECG score but normal EF (false positives; FP) more often developed future low EF. Objective: The purpose of this study was to evaluate echocardiographic characteristics and all-cause mortality risk in FP patients. Methods: Patients with transthoracic echocardiography and ECG were classified retrospectively into FP, true negatives (TN) (EF ≥50%, normal AI-ECG), true positives (TP) (EF <50%, abnormal AI-ECG), or false negatives (FN) (EF <50%, normal AI-ECG). Echocardiographic abnormalities included systolic and diastolic left ventricular function, valve disease, estimated pulmonary pressures, and right heart parameters. Cox regression was used to assess factors associated with all-cause mortality. Results: Of 100,586 patients (median age 63 years; 45.5% females), 79% were TN, 7% FP, 5% FN, and 8% TP. FPs had more echocardiographic abnormalities than TN but less than FN or TP patients. An echocardiographic abnormality was present in 97% of FPs. Over median 2.7 years, FPs had increased mortality risk (age and sex-adjusted HR: 1.64 [95% CI: 1.55-1.73]) vs TN. Age and sex-adjusted mortality was higher in FP with abnormal echocardiography than FP with normal echocardiography and to TN regardless of echocardiography result; FP with normal echocardiography had comparable mortality risk to TN with abnormal echocardiography. Conclusions: FP patients were more likely than TNs to have echocardiographic abnormalities with 97% of exams showing an abnormality. FP patients had higher mortality rates, especially when their echocardiograms also had an abnormality; the concomitant use of AI ECG and echocardiography helps in stratifying risk in patients with normal LVEF.

5.
Nat Med ; 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39223284

RESUMO

Nigeria has the highest reported incidence of peripartum cardiomyopathy worldwide. This open-label, pragmatic clinical trial randomized pregnant and postpartum women to usual care or artificial intelligence (AI)-guided screening to assess its impact on the diagnosis left ventricular systolic dysfunction (LVSD) in the perinatal period. The study intervention included digital stethoscope recordings with point of-care AI predictions and a 12-lead electrocardiogram with asynchronous AI predictions for LVSD. The primary end point was identification of LVSD during the study period. In the intervention arm, the primary end point was defined as the number of identified participants with LVSD as determined by a positive AI screen, confirmed by echocardiography. In the control arm, this was the number of participants with clinical recognition and documentation of LVSD on echocardiography in keeping with current standard of care. Participants in the intervention arm had a confirmatory echocardiogram at baseline for AI model validation. A total of 1,232 (616 in each arm) participants were randomized and 1,195 participants (587 intervention arm and 608 control arm) completed the baseline visit at 6 hospitals in Nigeria between August 2022 and September 2023 with follow-up through May 2024. Using the AI-enabled digital stethoscope, the primary study end point was met with detection of 24 out of 587 (4.1%) versus 12 out of 608 (2.0%) patients with LVSD (intervention versus control odds ratio 2.12, 95% CI 1.05-4.27; P = 0.032). With the 12-lead AI-electrocardiogram model, the primary end point was detected in 20 out of 587 (3.4%) versus 12 out of 608 (2.0%) patients (odds ratio 1.75, 95% CI 0.85-3.62; P = 0.125). A similar direction of effect was observed in prespecified subgroup analysis. There were no serious adverse events related to study participation. In pregnant and postpartum women, AI-guided screening using a digital stethoscope improved the diagnosis of pregnancy-related cardiomyopathy. ClinicalTrials.gov registration: NCT05438576.

7.
ESC Heart Fail ; 2024 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-39215684

RESUMO

AIMS: We aim to determine if our previously validated, diagnostic artificial intelligence (AI) electrocardiogram (ECG) model is prognostic for survival among patients with cardiac amyloidosis (CA). METHODS: A total of 2533 patients with CA (1834 with light chain amyloidosis (AL), 530 with wild-type transthyretin amyloid protein (ATTRwt) and 169 with hereditary transthyretin amyloid (ATTRv)] were included. An amyloid AI ECG (A2E) score was calculated for each patient reflecting the likelihood of CA. CA stage was calculated using the European modification of the Mayo 2004 criteria for AL and Mayo stage for transthyretin amyloid (ATTR). Risk of death was modelled using Cox proportional hazards, and Kaplan-Meier was used to estimate survival. RESULTS: Median age of the cohort was 67 [inter-quartile ratio (IQR) 59, 74], and 71.6% were male. The median overall survival for the cohort was 35.6 months [95% confidence interval (CI) 32.3, 39.5]. For AL, ATTRwt and ATTRv, respectively, median survival was 22.9 (95% CI 19.2, 28.2), 47.2 (95% CI 43.4, 52.3) and 61.4 (95% CI 48.7, 75.9) months. On univariate analysis, an increasing A2E score was associated with more than a two-fold risk of all-cause death. On multivariable analysis, the A2E score retained its importance with a risk ratio of 2.0 (95% CI 1.58, 2.55) in the AL group and 2.7 (95% CI 1.81, 4.24) in the ATTR group. CONCLUSIONS: Among patients with AL and ATTR amyloidosis, the A2E model helps to stratify risk of CA and adds another dimension of prognostication.

8.
Clin J Am Soc Nephrol ; 19(8): 952-958, 2024 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-39116276

RESUMO

Background: Artificial intelligence (AI) electrocardiogram (ECG) analysis can enable detection of hyperkalemia. In this validation, we assessed the algorithm's performance in two high acuity settings. Methods: An emergency department (ED) cohort (February to August 2021) and a mixed intensive care unit (ICU) cohort (August 2017 to February 2018) were identified and analyzed separately. For each group, pairs of laboratory-collected potassium and 12 lead ECGs obtained within 4 hours of each other were identified. The previously developed AI ECG algorithm was subsequently applied to leads 1 and 2 of the 12 lead ECGs to screen for hyperkalemia (potassium >6.0 mEq/L). Results: The ED cohort (N=40,128) had a mean age of 60 years, 48% were male, and 1% (N=351) had hyperkalemia. The area under the curve (AUC) of the AI-enhanced ECG (AI-ECG) to detect hyperkalemia was 0.88, with sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and positive likelihood ratio (LR+) of 80%, 80%, 3%, 99.8%, and 4.0, respectively, in the ED cohort. Low-eGFR (<30 ml/min) subanalysis yielded AUC, sensitivity, specificity, PPV, NPV, and LR+ of 0.83, 86%, 60%, 15%, 98%, and 2.2, respectively, in the ED cohort. The ICU cohort (N=2636) had a mean age of 65 years, 60% were male, and 3% (N=87) had hyperkalemia. The AUC for the AI-ECG was 0.88 and yielded sensitivity, specificity, PPV, NPV, and LR+ of 82%, 82%, 14%, 99%, and 4.6, respectively in the ICU cohort. Low-eGFR subanalysis yielded AUC, sensitivity, specificity, PPV, NPV, and LR+ of 0.85, 88%, 67%, 29%, 97%, and 2.7, respectively in the ICU cohort. Conclusions: The AI-ECG algorithm demonstrated a high NPV, suggesting that it is useful for ruling out hyperkalemia, but a low PPV, suggesting that it is insufficient for treating hyperkalemia.


Assuntos
Inteligência Artificial , Eletrocardiografia , Hiperpotassemia , Humanos , Hiperpotassemia/diagnóstico , Hiperpotassemia/sangue , Masculino , Feminino , Idoso , Pessoa de Meia-Idade , Valor Preditivo dos Testes
9.
Hypertension ; 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39011653

RESUMO

Hypertension is among the most important risk factors for cardiovascular disease, chronic kidney disease, and dementia. The artificial intelligence (AI) field is advancing quickly, and there has been little discussion on how AI could be leveraged for improving the diagnosis and management of hypertension. AI technologies, including machine learning tools, could alter the way we diagnose and manage hypertension, with potential impacts for improving individual and population health. The development of successful AI tools in public health and health care systems requires diverse types of expertise with collaborative relationships between clinicians, engineers, and data scientists. Unbiased data sources, management, and analyses remain a foundational challenge. From a diagnostic standpoint, machine learning tools may improve the measurement of blood pressure and be useful in the prediction of incident hypertension. To advance the management of hypertension, machine learning tools may be useful to find personalized treatments for patients using analytics to predict response to antihypertension medications and the risk for hypertension-related complications. However, there are real-world implementation challenges to using AI tools in hypertension. Herein, we summarize key findings from a diverse group of stakeholders who participated in a workshop held by the National Heart, Lung, and Blood Institute in March 2023. Workshop participants presented information on communication gaps between clinical medicine, data science, and engineering in health care; novel approaches to estimating BP, hypertension risk, and BP control; and real-world implementation challenges and issues.

10.
Eur Heart J Digit Health ; 5(4): 416-426, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39081936

RESUMO

Aims: Recently, deep learning artificial intelligence (AI) models have been trained to detect cardiovascular conditions, including hypertrophic cardiomyopathy (HCM), from the 12-lead electrocardiogram (ECG). In this external validation study, we sought to assess the performance of an AI-ECG algorithm for detecting HCM in diverse international cohorts. Methods and results: A convolutional neural network-based AI-ECG algorithm was developed previously in a single-centre North American HCM cohort (Mayo Clinic). This algorithm was applied to the raw 12-lead ECG data of patients with HCM and non-HCM controls from three external cohorts (Bern, Switzerland; Oxford, UK; and Seoul, South Korea). The algorithm's ability to distinguish HCM vs. non-HCM status from the ECG alone was examined. A total of 773 patients with HCM and 3867 non-HCM controls were included across three sites in the merged external validation cohort. The HCM study sample comprised 54.6% East Asian, 43.2% White, and 2.2% Black patients. Median AI-ECG probabilities of HCM were 85% for patients with HCM and 0.3% for controls (P < 0.001). Overall, the AI-ECG algorithm had an area under the receiver operating characteristic curve (AUC) of 0.922 [95% confidence interval (CI) 0.910-0.934], with diagnostic accuracy 86.9%, sensitivity 82.8%, and specificity 87.7% for HCM detection. In age- and sex-matched analysis (case-control ratio 1:2), the AUC was 0.921 (95% CI 0.909-0.934) with accuracy 88.5%, sensitivity 82.8%, and specificity 90.4%. Conclusion: The AI-ECG algorithm determined HCM status from the 12-lead ECG with high accuracy in diverse international cohorts, providing evidence for external validity. The value of this algorithm in improving HCM detection in clinical practice and screening settings requires prospective evaluation.

11.
Cardiovasc Digit Health J ; 5(3): 132-140, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38989045

RESUMO

Background: Cardiomyopathy is a leading cause of pregnancy-related mortality and the number one cause of death in the late postpartum period. Delay in diagnosis is associated with severe adverse outcomes. Objective: To evaluate the performance of an artificial intelligence-enhanced electrocardiogram (AI-ECG) and AI-enabled digital stethoscope to detect left ventricular systolic dysfunction in an obstetric population. Methods: We conducted a single-arm prospective study of pregnant and postpartum women enrolled at 3 sites between October 28, 2021, and October 27, 2022. Study participants completed a standard 12-lead ECG, digital stethoscope ECG and phonocardiogram recordings, and a transthoracic echocardiogram within 24 hours. Diagnostic performance was evaluated using the area under the curve (AUC). Results: One hundred women were included in the final analysis. The median age was 31 years (Q1: 27, Q3: 34). Thirty-eight percent identified as non-Hispanic White, 32% as non-Hispanic Black, and 21% as Hispanic. Five percent and 6% had left ventricular ejection fraction (LVEF) <45% and <50%, respectively. The AI-ECG model had near-perfect classification performance (AUC: 1.0, 100% sensitivity; 99%-100% specificity) for detection of cardiomyopathy at both LVEF categories. The AI-enabled digital stethoscope had an AUC of 0.98 (95% CI: 0.95, 1.00) and 0.97 (95% CI: 0.93, 1.00), for detection of LVEF <45% and <50%, respectively, with 100% sensitivity and 90% specificity. Conclusion: We demonstrate an AI-ECG and AI-enabled digital stethoscope were effective for detecting cardiac dysfunction in an obstetric population. Larger studies, including an evaluation of the impact of screening on clinical outcomes, are essential next steps.

12.
Eur Respir J ; 64(1)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38936966

RESUMO

BACKGROUND: Early diagnosis of pulmonary hypertension (PH) is critical for effective treatment and management. We aimed to develop and externally validate an artificial intelligence algorithm that could serve as a PH screening tool, based on analysis of a standard 12-lead ECG. METHODS: The PH Early Detection Algorithm (PH-EDA) is a convolutional neural network developed using retrospective ECG voltage-time data, with patients classified as "PH-likely" or "PH-unlikely" (controls) based on right heart catheterisation or echocardiography. In total, 39 823 PH-likely patients and 219 404 control patients from Mayo Clinic were randomly split into training (48%), validation (12%) and test (40%) sets. ECGs taken within 1 month of PH diagnosis (diagnostic dataset) were used to train the PH-EDA at Mayo Clinic. Performance was tested on diagnostic ECGs within the test sets from Mayo Clinic (n=16 175/87 998 PH-likely/controls) and Vanderbilt University Medical Center (VUMC; n=6045/24 256 PH-likely/controls). In addition, performance was tested on ECGs taken 6-18 months (pre-emptive dataset), and up to 5 years prior to a PH diagnosis at both sites. RESULTS: Performance testing yielded an area under the receiver operating characteristic curve (AUC) of 0.92 and 0.88 in the diagnostic test sets at Mayo Clinic and VUMC, respectively, and 0.86 and 0.81, respectively, in the pre-emptive test sets. The AUC remained a minimum of 0.79 at Mayo Clinic and 0.73 at VUMC up to 5 years before diagnosis. CONCLUSION: The PH-EDA can detect PH at diagnosis and 6-18 months prior, demonstrating the potential to accelerate diagnosis and management of this debilitating disease.


Assuntos
Algoritmos , Diagnóstico Precoce , Eletrocardiografia , Hipertensão Pulmonar , Humanos , Hipertensão Pulmonar/diagnóstico , Eletrocardiografia/métodos , Feminino , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Idoso , Inteligência Artificial , Curva ROC , Ecocardiografia , Adulto , Redes Neurais de Computação , Cateterismo Cardíaco
14.
Eur Heart J Digit Health ; 5(3): 295-302, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38774378

RESUMO

Aims: Cardiac amyloidosis (CA) is common in patients with severe aortic stenosis (AS) undergoing transcatheter aortic valve replacement (TAVR). Cardiac amyloidosis has poor outcomes, and its assessment in all TAVR patients is costly and challenging. Electrocardiogram (ECG) artificial intelligence (AI) algorithms that screen for CA may be useful to identify at-risk patients. Methods and results: In this retrospective analysis of our institutional National Cardiovascular Disease Registry (NCDR)-TAVR database, patients undergoing TAVR between January 2012 and December 2018 were included. Pre-TAVR CA probability was analysed by an ECG AI predictive model, with >50% risk defined as high probability for CA. Univariable and propensity score covariate adjustment analyses using Cox regression were performed to compare clinical outcomes between patients with high CA probability vs. those with low probability at 1-year follow-up after TAVR. Of 1426 patients who underwent TAVR (mean age 81.0 ± 8.5 years, 57.6% male), 349 (24.4%) had high CA probability on pre-procedure ECG. Only 17 (1.2%) had a clinical diagnosis of CA. After multivariable adjustment, high probability of CA by ECG AI algorithm was significantly associated with increased all-cause mortality [hazard ratio (HR) 1.40, 95% confidence interval (CI) 1.01-1.96, P = 0.046] and higher rates of major adverse cardiovascular events (transient ischaemic attack (TIA)/stroke, myocardial infarction, and heart failure hospitalizations] (HR 1.36, 95% CI 1.01-1.82, P = 0.041), driven primarily by heart failure hospitalizations (HR 1.58, 95% CI 1.13-2.20, P = 0.008) at 1-year follow-up. There were no significant differences in TIA/stroke or myocardial infarction. Conclusion: Artificial intelligence applied to pre-TAVR ECGs identifies a subgroup at higher risk of clinical events. These targeted patients may benefit from further diagnostic evaluation for CA.

15.
JACC CardioOncol ; 6(2): 251-263, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38774001

RESUMO

Background: The use of an artificial intelligence electrocardiography (AI-ECG) algorithm has demonstrated its reliability in predicting the risk of atrial fibrillation (AF) within the general population. Objectives: This study aimed to determine the effectiveness of the AI-ECG score in identifying patients with chronic lymphocytic leukemia (CLL) who are at high risk of developing AF. Methods: We estimated the probability of AF based on AI-ECG among patients with CLL extracted from the Mayo Clinic CLL database. Additionally, we computed the Mayo Clinic CLL AF risk score and determined its ability to predict AF. Results: Among 754 newly diagnosed patients with CLL, 71.4% were male (median age = 69 years). The median baseline AI-ECG score was 0.02 (range = 0-0.93), with a value ≥0.1 indicating high risk. Over a median follow-up of 5.8 years, the estimated 10-year cumulative risk of AF was 26.1%. Patients with an AI-ECG score of ≥0.1 had a significantly higher risk of AF (HR: 3.9; 95% CI: 2.6-5.7; P < 0.001). This heightened risk remained significant (HR: 2.5; 95% CI: 1.6-3.9; P < 0.001) even after adjusting for the Mayo CLL AF risk score, heart failure, chronic kidney disease, and CLL therapy. In a second cohort of CLL patients treated with a Bruton tyrosine kinase inhibitor (n = 220), a pretreatment AI-ECG score ≥0.1 showed a nonsignificant increase in the risk of AF (HR: 1.7; 95% CI: 0.8-3.6; P = 0.19). Conclusions: An AI-ECG algorithm, in conjunction with the Mayo CLL AF risk score, can predict the risk of AF in patients with newly diagnosed CLL. Additional studies are needed to determine the role of AI-ECG in predicting AF risk in CLL patients treated with a Bruton tyrosine kinase inhibitor.

16.
Eur Heart J Digit Health ; 5(3): 314-323, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38774362

RESUMO

Aims: Mobile devices such as smartphones and watches can now record single-lead electrocardiograms (ECGs), making wearables a potential screening tool for cardiac and wellness monitoring outside of healthcare settings. Because friends and family often share their smart phones and devices, confirmation that a sample is from a given patient is important before it is added to the electronic health record. Methods and results: We sought to determine whether the application of Siamese neural network would permit the diagnostic ECG sample to serve as both a medical test and biometric identifier. When using similarity scores to discriminate whether a pair of ECGs came from the same patient or different patients, inputs of single-lead and 12-lead medians produced an area under the curve of 0.94 and 0.97, respectively. Conclusion: The similar performance of the single-lead and 12-lead configurations underscores the potential use of mobile devices to monitor cardiac health.

17.
Eur Heart J Digit Health ; 5(3): 260-269, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38774376

RESUMO

Aims: Augmenting echocardiography with artificial intelligence would allow for automated assessment of routine parameters and identification of disease patterns not easily recognized otherwise. View classification is an essential first step before deep learning can be applied to the echocardiogram. Methods and results: We trained two- and three-dimensional convolutional neural networks (CNNs) using transthoracic echocardiographic (TTE) studies obtained from 909 patients to classify nine view categories (10 269 videos). Transthoracic echocardiographic studies from 229 patients were used in internal validation (2582 videos). Convolutional neural networks were tested on 100 patients with comprehensive TTE studies (where the two examples chosen by CNNs as most likely to represent a view were evaluated) and 408 patients with five view categories obtained via point-of-care ultrasound (POCUS). The overall accuracy of the two-dimensional CNN was 96.8%, and the averaged area under the curve (AUC) was 0.997 on the comprehensive TTE testing set; these numbers were 98.4% and 0.998, respectively, on the POCUS set. For the three-dimensional CNN, the accuracy and AUC were 96.3% and 0.998 for full TTE studies and 95.0% and 0.996 on POCUS videos, respectively. The positive predictive value, which defined correctly identified predicted views, was higher with two-dimensional rather than three-dimensional networks, exceeding 93% in apical, short-axis aortic valve, and parasternal long-axis left ventricle views. Conclusion: An automated view classifier utilizing CNNs was able to classify cardiac views obtained using TTE and POCUS with high accuracy. The view classifier will facilitate the application of deep learning to echocardiography.

18.
Transplantation ; 108(9): 1976-1985, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38557657

RESUMO

BACKGROUND: Predicting long-term mortality postkidney transplantation (KT) using baseline clinical data presents significant challenges. This study aims to evaluate the predictive power of artificial intelligence (AI)-enabled analysis of preoperative electrocardiograms (ECGs) in forecasting long-term mortality following KT. METHODS: We analyzed preoperative ECGs from KT recipients at three Mayo Clinic sites (Minnesota, Florida, and Arizona) between January 1, 2006, and July 30, 2021. The study involved 6 validated AI algorithms, each trained to predict future development of atrial fibrillation, aortic stenosis, low ejection fraction, hypertrophic cardiomyopathy, amyloid heart disease, and biological age. These algorithms' outputs based on a single preoperative ECG were correlated with patient mortality data. RESULTS: Among 6504 KT recipients included in the study, 1764 (27.1%) died within a median follow-up of 5.7 y (interquartile range: 3.00-9.29 y). All AI-ECG algorithms were independently associated with long-term all-cause mortality ( P < 0.001). Notably, few patients had a clinical cardiac diagnosis at the time of transplant, indicating that AI-ECG scores were predictive even in asymptomatic patients. When adjusted for multiple clinical factors such as recipient age, diabetes, and pretransplant dialysis, AI algorithms for atrial fibrillation and aortic stenosis remained independently associated with long-term mortality. These algorithms also improved the C-statistic for predicting overall (C = 0.74) and cardiac-related deaths (C = 0.751). CONCLUSIONS: The findings suggest that AI-enabled preoperative ECG analysis can be a valuable tool in predicting long-term mortality following KT and could aid in identifying patients who may benefit from enhanced cardiac monitoring because of increased risk.


Assuntos
Algoritmos , Inteligência Artificial , Eletrocardiografia , Transplante de Rim , Valor Preditivo dos Testes , Humanos , Transplante de Rim/efeitos adversos , Transplante de Rim/mortalidade , Feminino , Masculino , Pessoa de Meia-Idade , Adulto , Fatores de Risco , Fatores de Tempo , Medição de Risco , Idoso , Estudos Retrospectivos , Resultado do Tratamento
19.
JACC Clin Electrophysiol ; 10(4): 775-789, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38597855

RESUMO

Biological age may be a more valuable predictor of morbidity and mortality than a person's chronological age. Mathematical models have been used for decades to predict biological age, but recent developments in artificial intelligence (AI) have led to new capabilities in age estimation. Using deep learning methods to train AI models on hundreds of thousands of electrocardiograms (ECGs) to predict age results in a good, but imperfect, age prediction. The error predicting age using ECG, or the difference between AI-ECG-derived age and chronological age (delta age), may be a surrogate measurement of biological age, as the delta age relates to survival, even after adjusting for chronological age and other covariates associated with total and cardiovascular mortality. The relative affordability, noninvasiveness, and ubiquity of ECGs, combined with ease of access and potential to be integrated with smartphone or wearable technology, presents a potential paradigm shift in assessment of biological age.


Assuntos
Envelhecimento , Inteligência Artificial , Eletrocardiografia , Idoso , Humanos , Envelhecimento/fisiologia , Aprendizado Profundo
20.
Eur Heart J Digit Health ; 5(2): 192-194, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38505482

RESUMO

Aims: ECG abnormalities are often the first signs of arrhythmogenic right ventricular cardiomyopathy (ARVC) and we hypothesized that an artificial intelligence (AI)-enhanced ECG could help identify patients with ARVC and serve as a valuable disease-detection tool. Methods and results: We created a convolutional neural network to detect ARVC using a 12-lead ECG. All patients with ARVC who met the 2010 task force criteria and had disease-causative genetic variants were included. All case ECGs were randomly assigned in an 8:1:1 ratio into training, validation, and testing groups. The case ECGs were age- and sex-matched with control ECGs at our institution in a 1:100 ratio. Seventy-seven patients (51% male; mean age 47.2 ± 19.9), including 56 patients with PKP2, 7 with DSG2, 6 with DSC2, 6 with DSP, and 2 with JUP were included. The model was trained using 61 case ECGs and 5009 control ECGs; validated with 7 case ECGs and 678 control ECGs and tested in 22 case ECGs and 1256 control ECGs. The sensitivity, specificity, positive and negative predictive values of the model were 77.3, 62.9, 3.32, and 99.4%, respectively. The area under the curve for rhythm ECG and median beat ECG was 0.75 and 0.76, respectively. Conclusion: Our study found that the model performed well in excluding ARVC and supports the concept that the AI ECG can serve as a biomarker for ARVC if a larger cohort were available for network training. A multicentre study including patients with ARVC from other centres would be the next step in refining, testing, and validating this algorithm.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA