Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Can J Cardiol ; 2024 May 10.
Article in English | MEDLINE | ID: mdl-38735528

ABSTRACT

In the dynamic field of medical artificial intelligence (AI), cardiology stands out as a key area for its technological advancements and clinical application. This review explores the complex issue of data bias, specifically addressing those encountered during the development and implementation of AI tools in cardiology. We dissect the origins and impacts of these biases, which challenge their reliability and widespread applicability in healthcare. Using a case study, we highlight the complexities involved in addressing these biases from a clinical viewpoint. The goal of this review is to equip researchers and clinicians with the practical knowledge needed to identify, understand, and mitigate these biases, advocating for the creation of AI solutions that are not just technologically sound, but also fair and effective for all patient demographics.

2.
Eur Heart J Digit Health ; 5(3): 389-393, 2024 May.
Article in English | MEDLINE | ID: mdl-38774370

ABSTRACT

Aims: The accuracy of voice-assisted technologies, such as Amazon Alexa, to collect data in patients who are older or have heart failure (HF) is unknown. The aim of this study is to analyse the impact of increasing age and comorbid HF, when compared with younger participants and caregivers, and how these different subgroups classify their experience using a voice-assistant device, for screening purposes. Methods and results: Subgroup analysis (HF vs. caregivers and younger vs. older participants) of the VOICE-COVID-II trial, a randomized controlled study where participants were assigned with subsequent crossover to receive a SARS-CoV2 screening questionnaire by Amazon Alexa or a healthcare personnel. Overall concordance between the two methods was compared using unweighted kappa scores and percentage of agreement. From the 52 participants included, the median age was 51 (34-65) years and 21 (40%) were HF patients. The HF subgroup showed a significantly lower percentage of agreement compared with caregivers (95% vs. 99%, P = 0.03), and both the HF and older subgroups tended to have lower unweighted kappa scores than their counterparts. In a post-screening survey, both the HF and older subgroups were less acquainted and found the voice-assistant device more difficult to use compared with caregivers and younger individuals. Conclusion: This subgroup analysis highlights important differences in the performance of a voice-assistant-based technology in an older and comorbid HF population. Younger individuals and caregivers, serving as facilitators, have the potential to bridge the gap and enhance the integration of these technologies into clinical practice. Study Registration: ClinicalTrials.gov Identifier: NCT04508972.

4.
J Am Coll Cardiol ; 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38593945

ABSTRACT

Recent Artificial Intelligence (AI) advancements in cardiovascular care offer potential enhancements in effective diagnosis, treatment, and outcomes. Over 600 Food and Drug Administration (FDA)-approved clinical AI algorithms now exist, with 10% focusing on cardiovascular applications, highlighting the growing opportunities for AI to augment care. This review discusses the latest advancements in the field of AI, with a particular focus on the utilization of multimodal inputs and the field of generative AI. Further discussions in this review involve an approach to understanding the larger context in which AI-augmented care may exist, and include a discussion of the need for rigorous evaluation, appropriate infrastructure for deployment, ethics and equity assessments, regulatory oversight, and viable business cases for deployment. Embracing this rapidly evolving technology while setting an appropriately high evaluation benchmark with careful and patient-centered implementation will be crucial for cardiology to leverage AI to enhance patient care and the provider experience.

5.
J Am Coll Cardiol ; 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38593946

ABSTRACT

Recent AI advancements in cardiovascular care offer potential enhancements in diagnosis, treatment, and outcomes. Innovations to date focus on automating measurements, enhancing image quality, and detecting diseases using novel methods. Applications span wearables, electrocardiograms, echocardiography, angiography, genetics, and more. AI models detect diseases from electrocardiograms at accuracy not previously achieved by technology or human experts, including reduced ejection fraction, valvular heart disease, and other cardiomyopathies. However, AI's unique characteristics necessitates rigorous validation by addressing training methods, real-world efficacy, equity concerns, and long-term reliability. Despite an exponentially growing number of studies in cardiovascular AI, trials showing improvement in outcomes remain lacking. A number are currently underway. Embracing this rapidly evolving technology while setting a high evaluation benchmark will be crucial for cardiology to leverage AI to enhance patient care and the provider experience.

6.
Can J Cardiol ; 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38670456

ABSTRACT

Inherited arrhythmia disorders account for a significant proportion of sudden cardiac death, particularly among young individuals. Recent advances in our understanding of these syndromes have improved patient diagnosis and care, yet certain clinical gaps remain, particularly within case ascertainment, access to genetic testing and risk stratification. Artificial intelligence (AI), specifically machine learning and its subset deep learning, present promising solutions to these challenges. The capacity of AI to process vast amounts of patient data and identify disease patterns differentiates them from traditional methods, which are time and resource intensive. To date, AI models have demonstrated immense potential in condition detection (including asymptomatic/concealed disease) and genotype and phenotype identification, exceeding expert cardiologists in these tasks. Additionally, they have exhibited applicability for general population screening, improving case ascertainment in a set of conditions that are often asymptomatic such as left ventricular dysfunction. Third, models have displayed ability to improve testing protocols, as through model identification of disease and genotype, specific clinical testing (e.g. drug challenges or further diagnostic imaging) can be avoided, reducing health care expenses, speeding diagnosis, and possibly allowing for more incremental or targeted genetic testing approaches. These significant benefits warrant continued investigation of the field, particularly regarding the development and implementation of clinically applicable screening tools. This review summarizes key developments in the field, including studies in Long QT Syndrome, Brugada Syndrome, Hypertrophic Cardiomyopathy, and Arrhythmogenic Cardiomyopathies, and provides direction for effective future AI implementation in clinical practice.

7.
JAMA Cardiol ; 9(4): 377-384, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38446445

ABSTRACT

Importance: Congenital long QT syndrome (LQTS) is associated with syncope, ventricular arrhythmias, and sudden death. Half of patients with LQTS have a normal or borderline-normal QT interval despite LQTS often being detected by QT prolongation on resting electrocardiography (ECG). Objective: To develop a deep learning-based neural network for identification of LQTS and differentiation of genotypes (LQTS1 and LQTS2) using 12-lead ECG. Design, Setting, and Participants: This diagnostic accuracy study used ECGs from patients with suspected inherited arrhythmia enrolled in the Hearts in Rhythm Organization Registry (HiRO) from August 2012 to December 2021. The internal dataset was derived at 2 sites and an external validation dataset at 4 sites within the HiRO Registry; an additional cross-sectional validation dataset was from the Montreal Heart Institute. The cohort with LQTS included probands and relatives with pathogenic or likely pathogenic variants in KCNQ1 or KCNH2 genes with normal or prolonged corrected QT (QTc) intervals. Exposures: Convolutional neural network (CNN) discrimination between LQTS1, LQTS2, and negative genetic test results. Main Outcomes and Measures: The main outcomes were area under the curve (AUC), F1 scores, and sensitivity for detecting LQTS and differentiating genotypes using a CNN method compared with QTc-based detection. Results: A total of 4521 ECGs from 990 patients (mean [SD] age, 42 [18] years; 589 [59.5%] female) were analyzed. External validation within the national registry (101 patients) demonstrated the CNN's high diagnostic capacity for LQTS detection (AUC, 0.93; 95% CI, 0.89-0.96) and genotype differentiation (AUC, 0.91; 95% CI, 0.86-0.96). This surpassed expert-measured QTc intervals in detecting LQTS (F1 score, 0.84 [95% CI, 0.78-0.90] vs 0.22 [95% CI, 0.13-0.31]; sensitivity, 0.90 [95% CI, 0.86-0.94] vs 0.36 [95% CI, 0.23-0.47]), including in patients with normal or borderline QTc intervals (F1 score, 0.70 [95% CI, 0.40-1.00]; sensitivity, 0.78 [95% CI, 0.53-0.95]). In further validation in a cross-sectional cohort (406 patients) of high-risk patients and genotype-negative controls, the CNN detected LQTS with an AUC of 0.81 (95% CI, 0.80-0.85), which was better than QTc interval-based detection (AUC, 0.74; 95% CI, 0.69-0.78). Conclusions and Relevance: The deep learning model improved detection of congenital LQTS from resting ECGs and allowed for differentiation between the 2 most common genetic subtypes. Broader validation over an unselected general population may support application of this model to patients with suspected LQTS.


Subject(s)
Deep Learning , Long QT Syndrome , Humans , Female , Adult , Male , Cross-Sectional Studies , Long QT Syndrome/diagnosis , Long QT Syndrome/genetics , Electrocardiography , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/complications , Genotype
8.
JACC Clin Electrophysiol ; 10(2): 334-345, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38340117

ABSTRACT

BACKGROUND: Continuous monitoring for atrial fibrillation (AF) using photoplethysmography (PPG) from smartwatches or other wearables is challenging due to periods of poor signal quality during motion or suboptimal wearing. As a result, many consumer wearables sample infrequently and only analyze when the user is at rest, which limits the ability to perform continuous monitoring or to quantify AF. OBJECTIVES: This study aimed to compare 2 methods of continuous monitoring for AF in free-living patients: a well-validated signal processing (SP) heuristic and a convolutional deep neural network (DNN) trained on raw signal. METHODS: We collected 4 weeks of continuous PPG and electrocardiography signals in 204 free-living patients. Both SP and DNN models were developed and validated both on holdout patients and an external validation set. RESULTS: The results show that the SP model demonstrated receiver-operating characteristic area under the curve (AUC) of 0.972 (sensitivity 99.6%, specificity: 94.4%), which was similar to the DNN receiver-operating characteristic AUC of 0.973 (sensitivity 92.2, specificity: 95.5%); however, the DNN classified significantly more data (95% vs 62%), revealing its superior tolerance of tracings prone to motion artifact. Explainability analysis revealed that the DNN automatically suppresses motion artifacts, evaluates irregularity, and learns natural AF interbeat variability. The DNN performed better and analyzed more signal in the external validation cohort using a different population and PPG sensor (AUC, 0.994; 97% analyzed vs AUC, 0.989; 88% analyzed). CONCLUSIONS: DNNs perform at least as well as SP models, classify more data, and thus may be better for continuous PPG monitoring.


Subject(s)
Atrial Fibrillation , Deep Learning , Humans , Atrial Fibrillation/diagnosis , Photoplethysmography/methods , Heuristics , Monitoring, Physiologic
10.
J Med Internet Res ; 25: e47475, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37948098

ABSTRACT

BACKGROUND: Accurate, timely ascertainment of clinical end points, particularly hospitalizations, is crucial for clinical trials. The Tailored Antiplatelet Initiation to Lessen Outcomes Due to Decreased Clopidogrel Response after Percutaneous Coronary Intervention (TAILOR-PCI) Digital Study extended the main TAILOR-PCI trial's follow-up to 2 years, using a smartphone-based research app featuring geofencing-triggered surveys and routine monthly mobile phone surveys to detect cardiovascular (CV) hospitalizations. This pilot study compared these digital tools to conventional site-coordinator ascertainment of CV hospitalizations. OBJECTIVE: The objectives were to evaluate geofencing-triggered notifications and routine monthly mobile phone surveys' performance in detecting CV hospitalizations compared to telephone visits and health record reviews by study coordinators at each site. METHODS: US and Canadian participants from the TAILOR-PCI Digital Follow-Up Study were invited to download the Eureka Research Platform mobile app, opting in for location tracking using geofencing, triggering a smartphone-based survey if near a hospital for ≥4 hours. Participants were sent monthly notifications for CV hospitalization surveys. RESULTS: From 85 participants who consented to the Digital Study, downloaded the mobile app, and had not previously completed their final follow-up visit, 73 (85.8%) initially opted in and consented to geofencing. There were 9 CV hospitalizations ascertained by study coordinators among 5 patients, whereas 8 out of 9 (88.9%) were detected by routine monthly hospitalization surveys. One CV hospitalization went undetected by the survey as it occurred within two weeks of the previous event, and the survey only allowed reporting of a single hospitalization. Among these, 3 were also detected by the geofencing algorithm, but 6 out of 9 (66.7%) were missed by geofencing: 1 occurred in a participant who never consented to geofencing, while 5 hospitalizations occurred among participants who had subsequently turned off geofencing prior to their hospitalization. Geofencing-detected hospitalizations were ascertained within a median of 2 (IQR 1-3) days, monthly surveys within 11 (IQR 6.5-25) days, and site coordinator methods within 38 (IQR 9-105) days. The geofencing algorithm triggered 245 notifications among 39 participants, with 128 (52.2%) from true hospital presence and 117 (47.8%) from nonhospital health care facility visits. Additional geofencing iterative improvements to reduce hospital misidentification were made to the algorithm at months 7 and 12, elevating the rate of true alerts from 35.4% (55 true alerts/155 total alerts before month 7) to 78.7% (59 true alerts/75 total alerts in months 7-12) and ultimately to 93.3% (14 true alerts/5 total alerts in months 13-21), respectively. CONCLUSIONS: The monthly digital survey detected most CV hospitalizations, while the geofencing survey enabled earlier detection but did not offer incremental value beyond traditional tools. Digital tools could potentially reduce the burden on study coordinators in ascertaining CV hospitalizations. The advantages of timely reporting via geofencing should be weighed against the issue of false notifications, which can be mitigated through algorithmic refinements.


Subject(s)
Percutaneous Coronary Intervention , Humans , Clopidogrel/therapeutic use , Follow-Up Studies , Pilot Projects , Canada , Hospitalization
11.
NPJ Digit Med ; 6(1): 142, 2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37568050

ABSTRACT

Coronary angiography is the primary procedure for diagnosis and management decisions in coronary artery disease (CAD), but ad-hoc visual assessment of angiograms has high variability. Here we report a fully automated approach to interpret angiographic coronary artery stenosis from standard coronary angiograms. Using 13,843 angiographic studies from 11,972 adult patients at University of California, San Francisco (UCSF), between April 1, 2008 and December 31, 2019, we train neural networks to accomplish four sequential necessary tasks for automatic coronary artery stenosis localization and estimation. Algorithms are internally validated against criterion-standard labels for each task in hold-out test datasets. Algorithms are then externally validated in real-world angiograms from the University of Ottawa Heart Institute (UOHI) and also retrained using quantitative coronary angiography (QCA) data from the Montreal Heart Institute (MHI) core lab. The CathAI system achieves state-of-the-art performance across all tasks on unselected, real-world angiograms. Positive predictive value, sensitivity and F1 score are all ≥90% to identify projection angle and ≥93% for left/right coronary artery angiogram detection. To predict obstructive CAD stenosis (≥70%), CathAI exhibits an AUC of 0.862 (95% CI: 0.843-0.880). In UOHI external validation, CathAI achieves AUC 0.869 (95% CI: 0.830-0.907) to predict obstructive CAD. In the MHI QCA dataset, CathAI achieves an AUC of 0.775 (95%. CI: 0.594-0.955) after retraining. In conclusion, multiple purpose-built neural networks can function in sequence to accomplish automated analysis of real-world angiograms, which could increase standardization and reproducibility in angiographic coronary stenosis assessment.

12.
JAMA Cardiol ; 8(6): 586-594, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37163297

ABSTRACT

Importance: Understanding left ventricular ejection fraction (LVEF) during coronary angiography can assist in disease management. Objective: To develop an automated approach to predict LVEF from left coronary angiograms. Design, Setting, and Participants: This was a cross-sectional study with external validation using patient data from December 12, 2012, to December 31, 2019, from the University of California, San Francisco (UCSF). Data were randomly split into training, development, and test data sets. External validation data were obtained from the University of Ottawa Heart Institute. Included in the analysis were all patients 18 years or older who received a coronary angiogram and transthoracic echocardiogram (TTE) within 3 months before or 1 month after the angiogram. Exposure: A video-based deep neural network (DNN) called CathEF was used to discriminate (binary) reduced LVEF (≤40%) and to predict (continuous) LVEF percentage from standard angiogram videos of the left coronary artery. Guided class-discriminative gradient class activation mapping (GradCAM) was applied to visualize pixels in angiograms that contributed most to DNN LVEF prediction. Results: A total of 4042 adult angiograms with corresponding TTE LVEF from 3679 UCSF patients were included in the analysis. Mean (SD) patient age was 64.3 (13.3) years, and 2212 patients were male (65%). In the UCSF test data set (n = 813), the video-based DNN discriminated (binary) reduced LVEF (≤40%) with an area under the receiver operating characteristic curve (AUROC) of 0.911 (95% CI, 0.887-0.934); diagnostic odds ratio for reduced LVEF was 22.7 (95% CI, 14.0-37.0). DNN-predicted continuous LVEF had a mean absolute error (MAE) of 8.5% (95% CI, 8.1%-9.0%) compared with TTE LVEF. Although DNN-predicted continuous LVEF differed 5% or less compared with TTE LVEF in 38.0% (309 of 813) of test data set studies, differences greater than 15% were observed in 15.2% (124 of 813). In external validation (n = 776), video-based DNN discriminated (binary) reduced LVEF (≤40%) with an AUROC of 0.906 (95% CI, 0.881-0.931), and DNN-predicted continuous LVEF had an MAE of 7.0% (95% CI, 6.6%-7.4%). Video-based DNN tended to overestimate low LVEFs and underestimate high LVEFs. Video-based DNN performance was consistent across sex, body mass index, low estimated glomerular filtration rate (≤45), presence of acute coronary syndromes, obstructive coronary artery disease, and left ventricular hypertrophy. Conclusion and relevance: This cross-sectional study represents an early demonstration of estimating LVEF from standard angiogram videos of the left coronary artery using video-based DNNs. Further research can improve accuracy and reduce the variability of DNNs to maximize their clinical utility.


Subject(s)
Ventricular Dysfunction, Left , Ventricular Function, Left , Adult , Humans , Male , Middle Aged , Female , Ventricular Function, Left/physiology , Coronary Angiography , Stroke Volume/physiology , Artificial Intelligence , Ventricular Dysfunction, Left/diagnostic imaging , Cross-Sectional Studies , Algorithms
13.
J Card Fail ; 29(10): 1456-1460, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37224994

ABSTRACT

BACKGROUND: Voice-assisted artificial intelligence-based systems may streamline clinical care among patients with heart failure (HF) and caregivers; however, randomized clinical trials are needed. We evaluated the potential for Amazon Alexa (Alexa), a voice-assisted artificial intelligence-based system, to conduct screening for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in a HF clinic. METHODS AND RESULTS: We enrolled 52 participants (patients and caregivers) from a HF clinic who were randomly assigned with a subsequent cross-over to receive a SARS-CoV-2 screening questionnaire via Alexa or health care personnel. The primary outcome was overall response concordance, as measured by the percentage of agreement and unweighted kappa scores between groups. A postscreening survey evaluated comfort with using the artificial intelligence-based device. In total, 36 participants (69%) were male, the median age was 51 years (range 34-65 years) years and 36 (69%) were English speaking. Twenty-one participants (40%) were patients with HF. For the primary outcome, there were no statistical differences between the groups: Alexa-research coordinator group 96.9% agreement and unweighted kappa score of 0.92 (95% confidence interval 0.84-1.00) vs research coordinator-Alexa group 98.5% agreement and unweighted kappa score of 0.95 (95% confidence interval 0.88-1.00) (P value for all comparisons > .05). Overall, 87% of participants rated their screening experience as good or outstanding. CONCLUSIONS: Alexa demonstrated comparable performance to a health care professional for SARS-CoV-2 screening in a group of patients with HF and caregivers and may represent an attractive approach to symptom screening in this population. Future studies evaluating such technologies for other uses among patients with HF and caregivers are warranted. NCT04508972.

16.
Sci Rep ; 13(1): 3364, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36849487

ABSTRACT

Chest pain is a common clinical complaint for which myocardial injury is the primary concern and is associated with significant morbidity and mortality. To aid providers' decision-making, we aimed to analyze the electrocardiogram (ECG) using a deep convolutional neural network (CNN) to predict serum troponin I (TnI) from ECGs. We developed a CNN using 64,728 ECGs from 32,479 patients who underwent ECG within 2 h prior to a serum TnI laboratory result at the University of California, San Francisco (UCSF). In our primary analysis, we classified patients into groups of TnI < 0.02 or ≥ 0.02 µg/L using 12-lead ECGs. This was repeated with an alternative threshold of 1.0 µg/L and with single-lead ECG inputs. We also performed multiclass prediction for a set of serum troponin ranges. Finally, we tested the CNN in a cohort of patients selected for coronary angiography, including 3038 ECGs from 672 patients. Cohort patients were 49.0% female, 42.8% white, and 59.3% (19,283) never had a positive TnI value (≥ 0.02 µg/L). CNNs accurately predicted elevated TnI, both at a threshold of 0.02 µg/L (AUC = 0.783, 95% CI 0.780-0.786) and at a threshold of 1.0 µg/L (AUC = 0.802, 0.795-0.809). Models using single-lead ECG data achieved significantly lower accuracy, with AUCs ranging from 0.740 to 0.773 with variation by lead. Accuracy of the multi-class model was lower for intermediate TnI value-ranges. Our models performed similarly on the cohort of patients who underwent coronary angiography. Biomarker-defined myocardial injury can be predicted by CNNs from 12-lead and single-lead ECGs.


Subject(s)
Deep Learning , Heart Injuries , Humans , Female , Male , Troponin I , Area Under Curve , Biomarkers , Electrocardiography , Heart Injuries/diagnosis
17.
J Cardiovasc Transl Res ; 16(3): 541-545, 2023 06.
Article in English | MEDLINE | ID: mdl-36749563

ABSTRACT

The acceptability of artificially intelligent interactive voice response (AI-IVR) systems in cardiovascular research settings is unclear. As a result, we evaluated peoples' attitudes regarding the Amazon Echo Show 8 device when used for electronic data capture in cardiovascular clinics. Participants were recruited following the Voice-Based Screening for SARS-CoV-2 Exposure in Cardiovascular clinics study. Overall, 215 people enrolled and underwent screening (mean age 46.1; 55% females) in the VOICE-COVID study and 58 people consented to participate in a post-screening survey. Following thematic analysis, four key themes affecting AI-IVR acceptability were identified. These were difficulties with communication (44.8%), limitations with available interaction modalities (41.4%), barriers with the development of therapeutic relationships (25.9%), and concerns with universality and accessibility (8.6%). While there are potential concerns with the use of AI-IVR technologies, these systems appeared to be well accepted in cardiovascular clinics. Increased development of these technologies could significantly improve healthcare access and efficiency.


Subject(s)
COVID-19 , Female , Humans , Middle Aged , Male , SARS-CoV-2 , Attitude
18.
JMIR Res Protoc ; 12: e41209, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36719720

ABSTRACT

BACKGROUND: The COVID-19 pandemic has disrupted the health care system, limiting health care resources such as the availability of health care professionals, patient monitoring, contact tracing, and continuous surveillance. As a result of this significant burden, digital tools have become an important asset in increasing the efficiency of patient care delivery. Digital tools can help support health care institutions by tracking transmission of the virus, aiding in the screening process, and providing telemedicine support. However, digital health tools face challenges associated with barriers to accessibility, efficiency, and privacy-related ethical issues. OBJECTIVE: This paper describes the study design of an open-label, noninterventional, crossover, randomized controlled trial aimed at assessing whether interactive voice response systems can screen for SARS-CoV-2 in patients as accurately as standard screening done by people. The study aims to assess the concordance and interrater reliability of symptom screening done by Amazon Alexa compared to manual screening done by research coordinators. The perceived level of comfort of patients when interacting with voice response systems and their personal experience will also be evaluated. METHODS: A total of 52 patients visiting the heart failure clinic at the Royal Victoria Hospital of the McGill University Health Center, in Montreal, Quebec, will be recruited. Patients will be randomly assigned to first be screened for symptoms of SARS-CoV-2 either digitally, by Amazon Alexa, or manually, by the research coordinator. Participants will subsequently be crossed over and screened either digitally or manually. The clinical setup includes an Amazon Echo Show, a tablet, and an uninterrupted power supply mounted on a mobile cart. The primary end point will be the interrater reliability on the accuracy of randomized screening data performed by Amazon Alexa versus research coordinators. The secondary end point will be the perceived level of comfort and app engagement of patients as assessed using 5-point Likert scales and binary mode responses. RESULTS: Data collection started in May 2021 and is expected to be completed in fall 2022. Data analysis is expected to be completed in early 2023. CONCLUSIONS: The use of voice-based assistants could improve the provision of health services and reduce the burden on health care personnel. Demonstrating a high interrater reliability between Amazon Alexa and health care coordinators may serve future digital tools to streamline the screening and delivery of care in the context of other conditions and clinical settings. The COVID-19 pandemic occurs during the first digital era using digital tools such as Amazon Alexa for disease screening, and it represents an opportunity to implement such technology in health care institutions in the long term. TRIAL REGISTRATION: ClinicalTrials.gov NCT04508972; https://clinicaltrials.gov/ct2/show/NCT04508972. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): DERR1-10.2196/41209.

19.
J Cardiovasc Transl Res ; 16(3): 513-525, 2023 06.
Article in English | MEDLINE | ID: mdl-35460017

ABSTRACT

Cardiovascular diseases are the leading cause of death globally and contribute significantly to the cost of healthcare. Artificial intelligence (AI) is poised to reshape cardiology. Using supervised and unsupervised learning, the two main branches of AI, several applications have been developed in recent years to improve risk prediction, allow large-scale analysis of medical data, and phenotype patients for personalized medicine. In this review, we examine the key advances in AI in cardiology and its limitations regarding bias in the data, standardization in reporting, data access, and model trust and accountability in cases of error. Finally, we discuss implementation methods to unleash AI's potential in making healthcare more accurate and efficient. Several steps need to be followed and challenges overcome in order to successfully integrate AI in clinical practice and ensure its longevity.


Subject(s)
Cardiology , Cardiovascular Diseases , Humans , Artificial Intelligence , Algorithms , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/therapy , Precision Medicine
20.
CJC Open ; 4(11): 913-920, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36444364

ABSTRACT

Background: Peripartum cardiomyopathy (PPCM) is associated with severe morbidity and mortality, and the significance of right ventricular (RV) involvement is unclear. We sought to determine whether RV systolic dysfunction or dilatation is associated with adverse clinical outcomes in women with PPCM. Methods: We conducted a multicentre retrospective cohort study examining the association between echocardiographic RV systolic dysfunction or dilatation at the time of PPCM diagnosis and clinical outcomes. Clinical endpoints of interest were the need for mechanical support, recovery of left ventricular ejection fraction at follow-up, and a combined endpoint of hospitalization for heart failure, cardiac transplant, or death. Results: A total of 67 women, median age 30 years (interquartile range: 7), were diagnosed with PPCM between 1994 and 2015 in 17 participating centres. Twin pregnancies occurred in 11%; 62% of women were multiparous; and 24% had preeclampsia. RV systolic function was impaired in 18 (27%) and dilated in 8 (12%). Seven women required ventricular assistance, and 8 experienced the composite outcome during follow-up (25 [interquartile range 61] months). RV dysfunction was associated with the need for mechanical support (odds ratio 10.10 (95% confidence interval: 1.86-54.81), P = 0.007), but neither RV dysfunction nor dilatation was associated with left ventricular ejection fraction recovery, the need for cardiac transplant, heart failure hospitalization, or death. Conclusions: RV dysfunction is associated with the need for mechanical support in women with PPCM. These findings may improve risk stratification of complications and clinical management.


Introduction: La cardiomyopathie du péripartum (CMP-PP) est associée à la morbidité grave et à la mortalité, mais on ignore l'importance de l'atteinte ventriculaire droite (VD). Nous avons cherché à déterminer si la dysfonction systolique ou la dilatation VD sont associées aux résultats cliniques défavorables chez les femmes atteintes de CMP-PP. Méthodes: Nous avons mené une étude de cohorte rétrospective multicentrique sur l'association entre la dysfonction systolique ou la dilatation VD à l'échographie au moment du diagnostic de CMP-PP et les résultats cliniques. Les critères cliniques d'intérêt étaient la nécessité d'une assistance mécanique, la récupération de la fraction d'éjection ventriculaire gauche (FEVG) au suivi et un critère combiné d'hospitalisation liée à l'insuffisance cardiaque (IC), la transplantation cardiaque ou la mort. Résultats: Un total de 67 femmes, dont l'âge médian était de 30 ans (écart interquartile [EI] : 7), ont reçu un diagnostic de CMP-PP entre 1994 et 2015 dans 17 centres participants. Les grossesses gémellaires sont survenues chez 11 % ; 62 % de femmes étaient multipares ; et 24 % souffraient de prééclampsie. La fonction systolique VD était compromise chez 18 (27 %) femmes et le VD, dilaté, chez huit (12 %) femmes. Sept femmes ont eu besoin d'une assistance ventriculaire, et huit ont subi le critère composite durant le suivi (25 [EI : 61] mois). La dysfonction VD a été associée à la nécessité d'une assistance mécanique (rapport de cotes 10,10 [intervalle de confiance à 95 % : 1,86-54,81], P = 0,007), mais ni la dysfonction ni la dilatation VD n'ont été associées à la récupération de la FEVG, à la nécessité d'une transplantation cardiaque, à une hospitalisation liée à l'IC ou à la mort. Conclusions: La dysfonction VD est associée à la nécessité d'une assistance mécanique chez les femmes atteintes de CMP-PP. Ces conclusions peuvent permettre d'améliorer la stratification des risques de complications et la prise en charge clinique.

SELECTION OF CITATIONS
SEARCH DETAIL
...