Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
medRxiv ; 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38633808

RESUMEN

Background: Current risk stratification strategies for heart failure (HF) risk require either specific blood-based biomarkers or comprehensive clinical evaluation. In this study, we evaluated the use of artificial intelligence (AI) applied to images of electrocardiograms (ECGs) to predict HF risk. Methods: Across multinational longitudinal cohorts in the integrated Yale New Haven Health System (YNHHS) and in population-based UK Biobank (UKB) and Brazilian Longitudinal Study of Adult Health (ELSA-Brasil), we identified individuals without HF at baseline. Incident HF was defined based on the first occurrence of an HF hospitalization. We evaluated an AI-ECG model that defines the cross-sectional probability of left ventricular dysfunction from a single image of a 12-lead ECG and its association with incident HF. We accounted for the competing risk of death using the Fine-Gray subdistribution model and evaluated the discrimination using Harrel's c-statistic. The pooled cohort equations to prevent HF (PCP-HF) were used as a comparator for estimating incident HF risk. Results: Among 231,285 individuals at YNHHS, 4472 had a primary HF hospitalization over 4.5 years (IQR 2.5-6.6) of follow-up. In UKB and ELSA-Brasil, among 42,741 and 13,454 people, 46 and 31 developed HF over a follow-up of 3.1 (2.1-4.5) and 4.2 (3.7-4.5) years, respectively. A positive AI-ECG screen portended a 4-fold higher risk of incident HF among YNHHS patients (age-, sex-adjusted HR [aHR] 3.88 [95% CI, 3.63-4.14]). In UKB and ELSA-Brasil, a positive-screen ECG portended 13- and 24-fold higher hazard of incident HF, respectively (aHR: UKBB, 12.85 [6.87-24.02]; ELSA-Brasil, 23.50 [11.09-49.81]). The association was consistent after accounting for comorbidities and the competing risk of death. Higher model output probabilities were progressively associated with a higher risk for HF. The model's discrimination for incident HF was 0.718 in YNHHS, 0.769 in UKB, and 0.810 in ELSA-Brasil. Across cohorts, incorporating model probability with PCP-HF yielded a significant improvement in discrimination over PCP-HF alone. Conclusions: An AI model applied to images of 12-lead ECGs can identify those at elevated risk of HF across multinational cohorts. As a digital biomarker of HF risk that requires just an ECG image, this AI-ECG approach can enable scalable and efficient screening for HF risk.

2.
medRxiv ; 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38559021

RESUMEN

Background: Point-of-care ultrasonography (POCUS) enables access to cardiac imaging directly at the bedside but is limited by brief acquisition, variation in acquisition quality, and lack of advanced protocols. Objective: To develop and validate deep learning models for detecting underdiagnosed cardiomyopathies on cardiac POCUS, leveraging a novel acquisition quality-adapted modeling strategy. Methods: To develop the models, we identified transthoracic echocardiograms (TTEs) of patients across five hospitals in a large U.S. health system with transthyretin amyloid cardiomyopathy (ATTR-CM, confirmed by Tc99m-pyrophosphate imaging), hypertrophic cardiomyopathy (HCM, confirmed by cardiac magnetic resonance), and controls enriched for the presence of severe AS. In a sample of 290,245 TTE videos, we used novel augmentation approaches and a customized loss function to weigh image and view quality to train a multi-label, view agnostic video-based convolutional neural network (CNN) to discriminate the presence of ATTR-CM, HCM, and/or AS. Models were tested across 3,758 real-world POCUS videos from 1,879 studies in 1,330 independent emergency department (ED) patients from 2011 through 2023. Results: Our multi-label, view-agnostic classifier demonstrated state-of-the-art performance in discriminating ATTR-CM (AUROC 0.98 [95%CI: 0.96-0.99]) and HCM (AUROC 0.95 [95% CI: 0.94-0.96]) on standard TTE studies. Automated metrics of anatomical view correctness confirmed significantly lower quality in POCUS vs TTE videos (median view classifier confidence of 0.63 [IQR: 0.44-0.88] vs 0.93 [IQR: 0.69-1.00], p<0.001). When deployed to POCUS videos, our algorithm effectively discriminated ATTR-CM and HCM with AUROC of up to 0.94 (parasternal long-axis (PLAX)), and 0.85 (apical 4 chamber), corresponding to positive diagnostic odds ratios of 46.7 and 25.5, respectively. In total, 18/35 (51.4%) of ATTR-CM and 32/57 (41.1%) of HCM patients in the POCUS cohort had an AI-positive screen in the year before their eventual confirmatory imaging. Conclusions: We define and validate an AI framework that enables scalable, opportunistic screening of under-diagnosed cardiomyopathies using POCUS.

3.
medRxiv ; 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38562867

RESUMEN

Introduction: Portable devices capable of electrocardiogram (ECG) acquisition have the potential to enhance structural heart disease (SHD) management by enabling early detection through artificial intelligence-ECG (AI-ECG) algorithms. However, the performance of these AI algorithms for identifying SHD in a real-world screening setting is unknown. To address this gap, we aim to evaluate the validity of our wearable-adapted AI algorithm, which has been previously developed and validated for detecting SHD from single-lead portable ECGs in patients undergoing routine echocardiograms in the Yale New Haven Hospital (YNHH). Research Methods and Analysis: This is the protocol for a cross-sectional study in the echocardiographic laboratories of YNHH. The study will enroll 585 patients referred for outpatient transthoracic echocardiogram (TTE) as part of their routine clinical care. Patients expressing interest in participating in the study will undergo a screening interview, followed by enrollment upon meeting eligibility criteria and providing informed consent. During their routine visit, patients will undergo a 1-lead ECG with two devices - one with an Apple Watch and the second with another portable 1-lead ECG device. With participant consent, these 1-lead ECG data will be linked to participant demographic and clinical data recorded in the YNHH electronic health records (EHR). The study will assess the performance of the AI-ECG algorithm in identifying SHD, including left ventricular systolic dysfunction (LVSD), valvular disease and severe left ventricular hypertrophy (LVH), by comparing the algorithm's results with data obtained from TTE, which is the established gold standard for diagnosing SHD. Ethics and Dissemination: All patient EHR data required for assessing eligibility and conducting the AI-ECG will be accessed through secure servers approved for protected health information. Data will be maintained on secure, encrypted servers for a minimum of five years after the publication of our findings in a peer-reviewed journal, and any unanticipated adverse events or risks will be reported by the principal investigator to the Yale Institutional Review Board, which has reviewed and approved this protocol (Protocol Number: 2000035532).

4.
medRxiv ; 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38562897

RESUMEN

Background: Risk stratification strategies for cancer therapeutics-related cardiac dysfunction (CTRCD) rely on serial monitoring by specialized imaging, limiting their scalability. Objectives: To examine an artificial intelligence (AI)-enhanced electrocardiographic (AI-ECG) surrogate for imaging risk biomarkers, and its association with CTRCD. Methods: Across a five-hospital U.S.-based health system (2013-2023), we identified patients with breast cancer or non-Hodgkin lymphoma (NHL) who received anthracyclines (AC) and/or trastuzumab (TZM), and a control cohort receiving immune checkpoint inhibitors (ICI). We deployed a validated AI model of left ventricular systolic dysfunction (LVSD) to ECG images (≥0.1, positive screen) and explored its association with i) global longitudinal strain (GLS) measured within 15 days (n=7,271 pairs); ii) future CTRCD (new cardiomyopathy, heart failure, or left ventricular ejection fraction [LVEF]<50%), and LVEF<40%. In the ICI cohort we correlated baseline AI-ECG-LVSD predictions with downstream myocarditis. Results: Higher AI-ECG LVSD predictions were associated with worse GLS (-18% [IQR:-20 to -17%] for predictions<0.1, to -12% [IQR:-15 to -9%] for ≥0.5 (p<0.001)). In 1,308 patients receiving AC/TZM (age 59 [IQR:49-67] years, 999 [76.4%] women, 80 [IQR:42-115] follow-up months) a positive baseline AI-ECG LVSD screen was associated with ~2-fold and ~4.8-fold increase in the incidence of the composite CTRCD endpoint (adj.HR 2.22 [95%CI:1.63-3.02]), and LVEF<40% (adj.HR 4.76 [95%CI:2.62-8.66]), respectively. Among 2,056 patients receiving ICI (age 65 [IQR:57-73] years, 913 [44.4%] women, follow-up 63 [IQR:28-99] months) AI-ECG predictions were not associated with ICI myocarditis (adj.HR 1.36 [95%CI:0.47-3.93]). Conclusion: AI applied to baseline ECG images can stratify the risk of CTRCD associated with anthracycline or trastuzumab exposure.

5.
medRxiv ; 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38585929

RESUMEN

Randomized clinical trials (RCTs) are essential to guide medical practice; however, their generalizability to a given population is often uncertain. We developed a statistically informed Generative Adversarial Network (GAN) model, RCT-Twin-GAN, that leverages relationships between covariates and outcomes and generates a digital twin of an RCT (RCT-Twin) conditioned on covariate distributions from a second patient population. We used RCT-Twin-GAN to reproduce treatment effect outcomes of the Systolic Blood Pressure Intervention Trial (SPRINT) and the Action to Control Cardiovascular Risk in Diabetes (ACCORD) Blood Pressure Trial, which tested the same intervention but had different treatment effect results. To demonstrate treatment effect estimates of each RCT conditioned on the other RCT patient population, we evaluated the cardiovascular event-free survival of SPRINT digital twins conditioned on the ACCORD cohort and vice versa (SPRINT-conditioned ACCORD twins). The conditioned digital twins were balanced by the intervention arm (mean absolute standardized mean difference (MASMD) of covariates between treatment arms 0.019 (SD 0.018), and the conditioned covariates of the SPRINT-Twin on ACCORD were more similar to ACCORD than a sprint (MASMD 0.0082 SD 0.016 vs. 0.46 SD 0.20). Most importantly, across iterations, SPRINT conditioned ACCORD-Twin datasets reproduced the overall non-significant effect size seen in ACCORD (5-year cardiovascular outcome hazard ratio (95% confidence interval) of 0.88 (0.73-1.06) in ACCORD vs median 0.87 (0.68-1.13) in the SPRINT conditioned ACCORD-Twin), while the ACCORD conditioned SPRINT-Twins reproduced the significant effect size seen in SPRINT (0.75 (0.64-0.89) vs median 0.79 (0.72-0.86)) in ACCORD conditioned SPRINT-Twin). Finally, we describe the translation of this approach to real-world populations by conditioning the trials on an electronic health record population. Therefore, RCT-Twin-GAN simulates the direct translation of RCT-derived treatment effects across various patient populations with varying covariate distributions.

6.
JAMA Cardiol ; 2024 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-38581644

RESUMEN

Importance: Aortic stenosis (AS) is a major public health challenge with a growing therapeutic landscape, but current biomarkers do not inform personalized screening and follow-up. A video-based artificial intelligence (AI) biomarker (Digital AS Severity index [DASSi]) can detect severe AS using single-view long-axis echocardiography without Doppler characterization. Objective: To deploy DASSi to patients with no AS or with mild or moderate AS at baseline to identify AS development and progression. Design, Setting, and Participants: This is a cohort study that examined 2 cohorts of patients without severe AS undergoing echocardiography in the Yale New Haven Health System (YNHHS; 2015-2021) and Cedars-Sinai Medical Center (CSMC; 2018-2019). A novel computational pipeline for the cross-modal translation of DASSi into cardiac magnetic resonance (CMR) imaging was further developed in the UK Biobank. Analyses were performed between August 2023 and February 2024. Exposure: DASSi (range, 0-1) derived from AI applied to echocardiography and CMR videos. Main Outcomes and Measures: Annualized change in peak aortic valve velocity (AV-Vmax) and late (>6 months) aortic valve replacement (AVR). Results: A total of 12 599 participants were included in the echocardiographic study (YNHHS: n = 8798; median [IQR] age, 71 [60-80] years; 4250 [48.3%] women; median [IQR] follow-up, 4.1 [2.4-5.4] years; and CSMC: n = 3801; median [IQR] age, 67 [54-78] years; 1685 [44.3%] women; median [IQR] follow-up, 3.4 [2.8-3.9] years). Higher baseline DASSi was associated with faster progression in AV-Vmax (per 0.1 DASSi increment: YNHHS, 0.033 m/s per year [95% CI, 0.028-0.038] among 5483 participants; CSMC, 0.082 m/s per year [95% CI, 0.053-0.111] among 1292 participants), with values of 0.2 or greater associated with a 4- to 5-fold higher AVR risk than values less than 0.2 (YNHHS: 715 events; adjusted hazard ratio [HR], 4.97 [95% CI, 2.71-5.82]; CSMC: 56 events; adjusted HR, 4.04 [95% CI, 0.92-17.70]), independent of age, sex, race, ethnicity, ejection fraction, and AV-Vmax. This was reproduced across 45 474 participants (median [IQR] age, 65 [59-71] years; 23 559 [51.8%] women; median [IQR] follow-up, 2.5 [1.6-3.9] years) undergoing CMR imaging in the UK Biobank (for participants with DASSi ≥0.2 vs those with DASSi <.02, adjusted HR, 11.38 [95% CI, 2.56-50.57]). Saliency maps and phenome-wide association studies supported associations with cardiac structure and function and traditional cardiovascular risk factors. Conclusions and Relevance: In this cohort study of patients without severe AS undergoing echocardiography or CMR imaging, a new AI-based video biomarker was independently associated with AS development and progression, enabling opportunistic risk stratification across cardiovascular imaging modalities as well as potential application on handheld devices.

8.
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.

9.
medRxiv ; 2024 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-38293023

RESUMEN

Background: Artificial intelligence-enhanced electrocardiography (AI-ECG) can identify hypertrophic cardiomyopathy (HCM) on 12-lead ECGs and offers a novel way to monitor treatment response. While the surgical or percutaneous reduction of the interventricular septum (SRT) represented initial HCM therapies, mavacamten offers an oral alternative. Objective: To evaluate biological response to SRT and mavacamten. Methods: We applied an AI-ECG model for HCM detection to ECG images from patients who underwent SRT across three sites: Yale New Haven Health System (YNHHS), Cleveland Clinic Foundation (CCF), and Atlantic Health System (AHS); and to ECG images from patients receiving mavacamten at YNHHS. Results: A total of 70 patients underwent SRT at YNHHS, 100 at CCF, and 145 at AHS. At YNHHS, there was no significant change in the AI-ECG HCM score before versus after SRT (pre-SRT: median 0.55 [IQR 0.24-0.77] vs post-SRT: 0.59 [0.40-0.75]). The AI-ECG HCM scores also did not improve post SRT at CCF (0.61 [0.32-0.79] vs 0.69 [0.52-0.79]) and AHS (0.52 [0.35-0.69] vs 0.61 [0.49-0.70]). Among 36 YNHHS patients on mavacamten therapy, the median AI-ECG score before starting mavacamten was 0.41 (0.22-0.77), which decreased significantly to 0.28 (0.11-0.50, p <0.001 by Wilcoxon signed-rank test) at the end of a median follow-up period of 237 days. Conclusions: The lack of improvement in AI-based HCM score with SRT, in contrast to a significant decrease with mavacamten, suggests the potential role of AI-ECG for serial monitoring of pathophysiological improvement in HCM at the point-of-care using ECG images.

10.
J Am Med Inform Assoc ; 31(4): 855-865, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38269618

RESUMEN

OBJECTIVE: Artificial intelligence (AI) detects heart disease from images of electrocardiograms (ECGs). However, traditional supervised learning is limited by the need for large amounts of labeled data. We report the development of Biometric Contrastive Learning (BCL), a self-supervised pretraining approach for label-efficient deep learning on ECG images. MATERIALS AND METHODS: Using pairs of ECGs from 78 288 individuals from Yale (2000-2015), we trained a convolutional neural network to identify temporally separated ECG pairs that varied in layouts from the same patient. We fine-tuned BCL-pretrained models to detect atrial fibrillation (AF), gender, and LVEF < 40%, using ECGs from 2015 to 2021. We externally tested the models in cohorts from Germany and the United States. We compared BCL with ImageNet initialization and general-purpose self-supervised contrastive learning for images (simCLR). RESULTS: While with 100% labeled training data, BCL performed similarly to other approaches for detecting AF/Gender/LVEF < 40% with an AUROC of 0.98/0.90/0.90 in the held-out test sets, it consistently outperformed other methods with smaller proportions of labeled data, reaching equivalent performance at 50% of data. With 0.1% data, BCL achieved AUROC of 0.88/0.79/0.75, compared with 0.51/0.52/0.60 (ImageNet) and 0.61/0.53/0.49 (simCLR). In external validation, BCL outperformed other methods even at 100% labeled training data, with an AUROC of 0.88/0.88 for Gender and LVEF < 40% compared with 0.83/0.83 (ImageNet) and 0.84/0.83 (simCLR). DISCUSSION AND CONCLUSION: A pretraining strategy that leverages biometric signatures of different ECGs from the same patient enhances the efficiency of developing AI models for ECG images. This represents a major advance in detecting disorders from ECG images with limited labeled data.


Asunto(s)
Fibrilación Atrial , Aprendizaje Profundo , Humanos , Inteligencia Artificial , Electrocardiografía , Biometría
11.
medRxiv ; 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-37808685

RESUMEN

Importance: Aortic stenosis (AS) is a major public health challenge with a growing therapeutic landscape, but current biomarkers do not inform personalized screening and follow-up. Objective: A video-based artificial intelligence (AI) biomarker (Digital AS Severity index [DASSi]) can detect severe AS using single-view long-axis echocardiography without Doppler. Here, we deploy DASSi to patients with no or mild/moderate AS at baseline to identify AS development and progression. Design Setting and Participants: We defined two cohorts of patients without severe AS undergoing echocardiography in the Yale-New Haven Health System (YNHHS) (2015-2021, 4.1[IQR:2.4-5.4] follow-up years) and Cedars-Sinai Medical Center (CSMC) (2018-2019, 3.4[IQR:2.8-3.9] follow-up years). We further developed a novel computational pipeline for the cross-modality translation of DASSi into cardiac magnetic resonance (CMR) imaging in the UK Biobank (2.5[IQR:1.6-3.9] follow-up years). Analyses were performed between August 2023-February 2024. Exposure: DASSi (range: 0-1) derived from AI applied to echocardiography and CMR videos. Main Outcomes and Measures: Annualized change in peak aortic valve velocity (AV-V max ) and late (>6 months) aortic valve replacement (AVR). Results: A total of 12,599 participants were included in the echocardiographic study (YNHHS: n =8,798, median age of 71 [IQR (interquartile range):60-80] years, 4250 [48.3%] women, and CSMC: n =3,801, 67 [IQR:54-78] years, 1685 [44.3%] women). Higher baseline DASSi was associated with faster progression in AV-V max (per 0.1 DASSi increments: YNHHS: +0.033 m/s/year [95%CI:0.028-0.038], n=5,483, and CSMC: +0.082 m/s/year [0.053-0.111], n=1,292), with levels ≥ vs <0.2 linked to a 4-to-5-fold higher AVR risk (715 events in YNHHS; adj.HR 4.97 [95%CI: 2.71-5.82], 56 events in CSMC: 4.04 [0.92-17.7]), independent of age, sex, ethnicity/race, ejection fraction and AV-V max . This was reproduced across 45,474 participants (median age 65 [IQR:59-71] years, 23,559 [51.8%] women) undergoing CMR in the UK Biobank (adj.HR 11.4 [95%CI:2.56-50.60] for DASSi ≥vs<0.2). Saliency maps and phenome-wide association studies supported links with traditional cardiovascular risk factors and diastolic dysfunction. Conclusions and Relevance: In this cohort study of patients without severe AS undergoing echocardiography or CMR imaging, a new AI-based video biomarker is independently associated with AS development and progression, enabling opportunistic risk stratification across cardiovascular imaging modalities as well as potential application on handheld devices.

12.
JACC Adv ; 2(7)2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38094515

RESUMEN

BACKGROUND: Smartphone-based health applications are increasingly popular, but their real-world use for cardiovascular risk management remains poorly understood. OBJECTIVES: The purpose of this study was to investigate the patterns of tracking health goals using smart devices, including smartphones and/or tablets, in the United States. METHODS: Using the nationally representative Health Information National Trends Survey for 2017 to 2020, we examined self-reported tracking of health-related goals (optimizing body weight, increasing physical activity, and/or quitting smoking) using smart devices among those with cardiovascular disease (CVD) or cardiovascular risk factors of hypertension, diabetes, obesity, and/or smoking. Survey analyses were used to obtain national estimates of use patterns and identify features associated with the use of these devices for tracking health goals. RESULTS: Of 16,092 Health Information National Trends Survey participants, 10,660 had CVD or cardiovascular risk factors, representing 154.2 million (95% CI: 149.2-159.3 million) U.S. adults. Among the general U.S. adult population, 46% (95% CI: 44%-47%) tracked their health goals using their smart devices, compared with 42% (95% CI: 40%-43%) of those with or at risk of CVD. Younger age, female, Black race, higher educational attainment, and greater income were independently associated with tracking of health goals using smart devices. CONCLUSIONS: Two in 5 U.S. adults with or at risk of CVD use their smart devices to track health goals. While representing a potential avenue to improve care, the lower use of smart devices among older and low-income individuals, who are at higher risk of adverse cardiovascular outcomes, requires that digital health interventions are designed so as not to exacerbate existing disparities.

13.
medRxiv ; 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38106089

RESUMEN

Background: Randomized clinical trials (RCTs) are designed to produce evidence in selected populations. Assessing their effects in the real-world is essential to change medical practice, however, key populations are historically underrepresented in the RCTs. We define an approach to simulate RCT-based effects in real-world settings using RCT digital twins reflecting the covariate patterns in an electronic health record (EHR). Methods: We developed a Generative Adversarial Network (GAN) model, RCT-Twin-GAN, which generates a digital twin of an RCT (RCT-Twin) conditioned on covariate distributions from an EHR cohort. We improved upon a traditional tabular conditional GAN, CTGAN, with a loss function adapted for data distributions and by conditioning on multiple discrete and continuous covariates simultaneously. We assessed the similarity between a Heart Failure with preserved Ejection Fraction (HFpEF) RCT (TOPCAT), a Yale HFpEF EHR cohort, and RCT-Twin. We also evaluated cardiovascular event-free survival stratified by Spironolactone (treatment) use. Results: By applying RCT-Twin-GAN to 3445 TOPCAT participants and conditioning on 3445 Yale EHR HFpEF patients, we generated RCT-Twin datasets between 1141-3445 patients in size, depending on covariate conditioning and model parameters. RCT-Twin randomly allocated spironolactone (S)/ placebo (P) arms like an RCT, was similar to RCT by a multi-dimensional distance metric, and balanced covariates (median absolute standardized mean difference (MASMD) 0.017, IQR 0.0034-0.030). The 5 EHR-conditioned covariates in RCT-Twin were closer to the EHR compared with the RCT (MASMD 0.008 vs 0.63, IQR 0.005-0.018 vs 0.59-1.11). RCT-Twin reproduced the overall effect size seen in TOPCAT (5-year cardiovascular composite outcome odds ratio (95% confidence interval) of 0.89 (0.75-1.06) in RCT vs 0.85 (0.69-1.04) in RCT-Twin). Conclusions: RCT-Twin-GAN simulates RCT-derived effects in real-world patients by translating these effects to the covariate distributions of EHR patients. This key methodological advance may enable the direct translation of RCT-derived effects into real-world patient populations and may enable causal inference in real-world settings.

14.
medRxiv ; 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37961715

RESUMEN

Randomized controlled trials (RCT) represent the cornerstone of evidence-based medicine but are resource-intensive. We propose and evaluate a machine learning (ML) strategy of adaptive predictive enrichment through computational trial phenomaps to optimize RCT enrollment. In simulated group sequential analyses of two large cardiovascular outcomes RCTs of (1) a therapeutic drug (pioglitazone versus placebo; Insulin Resistance Intervention after Stroke (IRIS) trial), and (2) a disease management strategy (intensive versus standard systolic blood pressure reduction in the Systolic Blood Pressure Intervention Trial (SPRINT)), we constructed dynamic phenotypic representations to infer response profiles during interim analyses and examined their association with study outcomes. Across three interim timepoints, our strategy learned dynamic phenotypic signatures predictive of individualized cardiovascular benefit. By conditioning a prospective candidate's probability of enrollment on their predicted benefit, we estimate that our approach would have enabled a reduction in the final trial size across ten simulations (IRIS: -14.8% ± 3.1%, pone-sample t-test=0.001; SPRINT: -17.6% ± 3.6%, pone-sample t-test<0.001), while preserving the original average treatment effect (IRIS: hazard ratio of 0.73 ± 0.01 for pioglitazone vs placebo, vs 0.76 in the original trial; SPRINT: hazard ratio of 0.72 ± 0.01 for intensive vs standard systolic blood pressure, vs 0.75 in the original trial; all with pone-sample t-test<0.01). This adaptive framework has the potential to maximize RCT enrollment efficiency.

15.
NPJ Digit Med ; 6(1): 217, 2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-38001154

RESUMEN

Randomized clinical trials (RCT) represent the cornerstone of evidence-based medicine but are resource-intensive. We propose and evaluate a machine learning (ML) strategy of adaptive predictive enrichment through computational trial phenomaps to optimize RCT enrollment. In simulated group sequential analyses of two large cardiovascular outcomes RCTs of (1) a therapeutic drug (pioglitazone versus placebo; Insulin Resistance Intervention after Stroke (IRIS) trial), and (2) a disease management strategy (intensive versus standard systolic blood pressure reduction in the Systolic Blood Pressure Intervention Trial (SPRINT)), we constructed dynamic phenotypic representations to infer response profiles during interim analyses and examined their association with study outcomes. Across three interim timepoints, our strategy learned dynamic phenotypic signatures predictive of individualized cardiovascular benefit. By conditioning a prospective candidate's probability of enrollment on their predicted benefit, we estimate that our approach would have enabled a reduction in the final trial size across ten simulations (IRIS: -14.8% ± 3.1%, pone-sample t-test = 0.001; SPRINT: -17.6% ± 3.6%, pone-sample t-test < 0.001), while preserving the original average treatment effect (IRIS: hazard ratio of 0.73 ± 0.01 for pioglitazone vs placebo, vs 0.76 in the original trial; SPRINT: hazard ratio of 0.72 ± 0.01 for intensive vs standard systolic blood pressure, vs 0.75 in the original trial; all simulations with Cox regression-derived p value of < 0.01 for the effect of the intervention on the respective primary outcome). This adaptive framework has the potential to maximize RCT enrollment efficiency.

16.
medRxiv ; 2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37873174

RESUMEN

In the rapidly evolving landscape of modern healthcare, the integration of wearable and portable technology provides a unique opportunity for personalized health monitoring in the community. Devices like the Apple Watch, FitBit, and AliveCor KardiaMobile have revolutionized the acquisition and processing of intricate health data streams that were previously accessible only through devices only available to healthcare providers. Amidst the variety of data collected by these gadgets, single-lead electrocardiogram (ECG) recordings have emerged as a crucial source of information for monitoring cardiovascular health. Notably, there has been significant advances in artificial intelligence capable of interpreting these 1-lead ECGs, facilitating clinical diagnosis as well as the detection of rare cardiac disorders. This design study describes the development of an innovative multi-platform system aimed at the rapid deployment of AI-based ECG solutions for clinical investigation and care delivery. The study examines various design considerations, aligning them with specific applications, and develops data flows to maximize efficiency for research and clinical use. This process encompasses the reception of single-lead ECGs from diverse wearable devices, channeling this data into a centralized data lake, and facilitating real-time inference through AI models for ECG interpretation. An evaluation of the platform demonstrates a mean duration from acquisition to reporting of results of 33.0 to 35.7 seconds, after a standard 30 second acquisition, allowing the complete process to be completed in 63.0 to 65.7 seconds. There were no substantial differences in acquisition to reporting across two commercially available devices (Apple Watch and KardiaMobile). These results demonstrate the succcessful translation of design principles into a fully integrated and efficient strategy for leveraging 1-lead ECGs across platforms and interpretation by AI-ECG algorithms. Such a platform is critical to translating AI discoveries for wearable and portable ECG devices to clinical impact through rapid deployment.

17.
medRxiv ; 2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37790355

RESUMEN

Importance: Elevated lipoprotein(a) [Lp(a)] is associated with atherosclerotic cardiovascular disease (ASCVD) and major adverse cardiovascular events (MACE). However, fewer than 0.5% of patients undergo Lp(a) testing, limiting the evaluation and use of novel targeted therapeutics currently under development. Objective: We developed and validated a machine learning model to enable targeted screening for elevated Lp(a). Design: Cross-sectional. Setting: 4 multinational population-based cohorts. Participants: We included 456,815 participants from the UK Biobank (UKB), the largest cohort with protocolized Lp(a) testing for model development. The model's external validity was assessed in Atherosclerosis Risk in Communities (ARIC) (N=14,484), Coronary Artery Risk Development in Young Adults (CARDIA) (N=4,124), and Multi-Ethnic Study of Atherosclerosis (MESA) (N=4,672) cohorts. Exposures: Demographics, medications, diagnoses, procedures, vitals, and laboratory measurements from UKB and linked electronic health records (EHR) were candidate input features to predict high Lp(a). We used the pooled cohort equations (PCE), an ASCVD risk marker, as a comparator to identify elevated Lp(a). Main Outcomes and Measures: The main outcome was elevated Lp(a) (≥150 nmol/L), and the number-needed-to-test (NNT) to find one case with elevated Lp(a). We explored the association of the model's prediction probabilities with all-cause and cardiovascular mortality, and MACE. Results: The Algorithmic Risk Inspection for Screening Elevated Lp(a) (ARISE) used low-density lipoprotein cholesterol, statin use, triglycerides, high-density lipoprotein cholesterol, history of ASCVD, and anti-hypertensive medication use as input features. ARISE outperformed cardiovascular risk stratification through PCE for predicting elevated Lp(a) with a significantly lower NNT (4.0 versus 8.0 [with or without PCE], P<0.001). ARISE performed comparably across external validation cohorts and subgroups, reducing the NNT by up to 67.3%, depending on the probability threshold. Over a median follow-up of 4.2 years, a high ARISE probability was also associated with a greater hazard of all-cause death and MACE (age/sex-adjusted hazard ratio [aHR], 1.35, and 1.38, respectively, P<0.001), with a greater increase in cardiovascular mortality (aHR, 2.17, P<0.001). Conclusions and Relevance: ARISE optimizes screening for elevated Lp(a) using commonly available clinical features. ARISE can be deployed in EHR and other settings to encourage greater Lp(a) testing and to improve identifying cases eligible for novel targeted therapeutics in trials. KEY POINTS: Question: How can we optimize the identification of individuals with elevated lipoprotein(a) [Lp(a)] who may be eligible for novel targeted therapeutics?Findings: Using 4 multinational population-based cohorts, we developed and validated a machine learning model, Algorithmic Risk Inspection for Screening Elevated Lp(a) (ARISE), to enable targeted screening for elevated Lp(a). In contrast to the pooled cohort equations that do not identify those with elevated Lp(a), ARISE reduces the "number-needed-to-test" to find one case with elevated Lp(a) by up to 67.3%.Meaning: ARISE can be deployed in electronic health records and other settings to enable greater yield of Lp(a) testing, thereby improving the identification of individuals with elevated Lp(a).

18.
Cardiovasc Diabetol ; 22(1): 259, 2023 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-37749579

RESUMEN

Artificial intelligence and machine learning are driving a paradigm shift in medicine, promising data-driven, personalized solutions for managing diabetes and the excess cardiovascular risk it poses. In this comprehensive review of machine learning applications in the care of patients with diabetes at increased cardiovascular risk, we offer a broad overview of various data-driven methods and how they may be leveraged in developing predictive models for personalized care. We review existing as well as expected artificial intelligence solutions in the context of diagnosis, prognostication, phenotyping, and treatment of diabetes and its cardiovascular complications. In addition to discussing the key properties of such models that enable their successful application in complex risk prediction, we define challenges that arise from their misuse and the role of methodological standards in overcoming these limitations. We also identify key issues in equity and bias mitigation in healthcare and discuss how the current regulatory framework should ensure the efficacy and safety of medical artificial intelligence products in transforming cardiovascular care and outcomes in diabetes.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus , Humanos , Inteligencia Artificial , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/prevención & control , Factores de Riesgo , Aprendizaje Automático , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/epidemiología , Diabetes Mellitus/terapia , Factores de Riesgo de Enfermedad Cardiaca
19.
medRxiv ; 2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37745527

RESUMEN

Objective: Artificial intelligence (AI) detects heart disease from images of electrocardiograms (ECGs), however traditional supervised learning is limited by the need for large amounts of labeled data. We report the development of Biometric Contrastive Learning (BCL), a self-supervised pretraining approach for label-efficient deep learning on ECG images. Materials and Methods: Using pairs of ECGs from 78,288 individuals from Yale (2000-2015), we trained a convolutional neural network to identify temporally-separated ECG pairs that varied in layouts from the same patient. We fine-tuned BCL-pretrained models to detect atrial fibrillation (AF), gender, and LVEF<40%, using ECGs from 2015-2021. We externally tested the models in cohorts from Germany and the US. We compared BCL with random initialization and general-purpose self-supervised contrastive learning for images (simCLR). Results: While with 100% labeled training data, BCL performed similarly to other approaches for detecting AF/Gender/LVEF<40% with AUROC of 0.98/0.90/0.90 in the held-out test sets, it consistently outperformed other methods with smaller proportions of labeled data, reaching equivalent performance at 50% of data. With 0.1% data, BCL achieved AUROC of 0.88/0.79/0.75, compared with 0.51/0.52/0.60 (random) and 0.61/0.53/0.49 (simCLR). In external validation, BCL outperformed other methods even at 100% labeled training data, with AUROC of 0.88/0.88 for Gender and LVEF<40% compared with 0.83/0.83 (random) and 0.84/0.83 (simCLR). Discussion and Conclusion: A pretraining strategy that leverages biometric signatures of different ECGs from the same patient enhances the efficiency of developing AI models for ECG images. This represents a major advance in detecting disorders from ECG images with limited labeled data.

20.
Eur Heart J ; 44(43): 4592-4604, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37611002

RESUMEN

BACKGROUND AND AIMS: Early diagnosis of aortic stenosis (AS) is critical to prevent morbidity and mortality but requires skilled examination with Doppler imaging. This study reports the development and validation of a novel deep learning model that relies on two-dimensional (2D) parasternal long axis videos from transthoracic echocardiography without Doppler imaging to identify severe AS, suitable for point-of-care ultrasonography. METHODS AND RESULTS: In a training set of 5257 studies (17 570 videos) from 2016 to 2020 [Yale-New Haven Hospital (YNHH), Connecticut], an ensemble of three-dimensional convolutional neural networks was developed to detect severe AS, leveraging self-supervised contrastive pretraining for label-efficient model development. This deep learning model was validated in a temporally distinct set of 2040 consecutive studies from 2021 from YNHH as well as two geographically distinct cohorts of 4226 and 3072 studies, from California and other hospitals in New England, respectively. The deep learning model achieved an area under the receiver operating characteristic curve (AUROC) of 0.978 (95% CI: 0.966, 0.988) for detecting severe AS in the temporally distinct test set, maintaining its diagnostic performance in geographically distinct cohorts [0.952 AUROC (95% CI: 0.941, 0.963) in California and 0.942 AUROC (95% CI: 0.909, 0.966) in New England]. The model was interpretable with saliency maps identifying the aortic valve, mitral annulus, and left atrium as the predictive regions. Among non-severe AS cases, predicted probabilities were associated with worse quantitative metrics of AS suggesting an association with various stages of AS severity. CONCLUSION: This study developed and externally validated an automated approach for severe AS detection using single-view 2D echocardiography, with potential utility for point-of-care screening.


Asunto(s)
Estenosis de la Válvula Aórtica , Aprendizaje Profundo , Humanos , Ecocardiografía , Estenosis de la Válvula Aórtica/diagnóstico por imagen , Estenosis de la Válvula Aórtica/complicaciones , Válvula Aórtica/diagnóstico por imagen , Ultrasonografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...