Next Article in Journal
Novel Biomarkers in Hepatocellular Carcinoma from Embryogenic Antigens to cfDNA
Previous Article in Journal
Modelling Population Genetic Screening in Rare Neurodegenerative Diseases
Previous Article in Special Issue
A Novel Bradycardia-Associated Variant in HCN4 as a Candidate Modifier in Type 3 Long QT Syndrome: Case Report and Deep In Silico Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hearts, Data, and Artificial Intelligence Wizardry: From Imitation to Innovation in Cardiovascular Care

by
Panteleimon Pantelidis
1,2,*,
Polychronis Dilaveris
3,
Samuel Ruipérez-Campillo
4,5,
Athina Goliopoulou
1,
Alexios Giannakodimos
1,
Panagiotis Theofilis
3,
Raffaele De Lucia
6,
Ourania Katsarou
1,
Konstantinos Zisimos
1,
Konstantinos Kalogeras
1,
Evangelos Oikonomou
1 and
Gerasimos Siasos
1
1
3rd Department of Cardiology, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Department of Computer and Systems Sciences, Stockholm University, 16455 Stockholm, Sweden
3
1st Department of Cardiology, National and Kapodistrian University of Athens, 11527 Athens, Greece
4
Department of Computer Science, ETH Zurich, 8092 Zurich, Switzerland
5
Department of Medicine, Stanford University, Stanford, CA 94305, USA
6
2nd Division of Cardiology, Cardiac Thoracic and Vascular Department, Azienda Ospedaliero Universitaria Pisana, 56124 Pisa, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(5), 1019; https://doi.org/10.3390/biomedicines13051019
Submission received: 16 March 2025 / Revised: 14 April 2025 / Accepted: 21 April 2025 / Published: 23 April 2025
(This article belongs to the Special Issue Cardiovascular Diseases in the Era of Precision Medicine)

Abstract

:
Artificial intelligence (AI) is transforming cardiovascular medicine by enabling the analysis of high-dimensional biomedical data with unprecedented precision. Initially employed to automate human tasks such as electrocardiogram (ECG) interpretation and imaging segmentation, AI’s true potential lies in uncovering hidden disease data patterns, predicting long-term cardiovascular risk, and personalizing treatments. Unlike human cognition, which excels in certain tasks but is limited by memory and processing constraints, AI integrates multimodal data sources—including ECG, echocardiography, cardiac magnetic resonance (CMR) imaging, genomics, and wearable sensor data—to generate novel clinical insights. AI models have demonstrated remarkable success in early dis-ease detection, such as predicting heart failure from standard ECGs before symptom on-set, distinguishing genetic cardiomyopathies, and forecasting arrhythmic events. However, several challenges persist, including AI’s lack of contextual understanding in most of these tasks, its “black-box” nature, and biases in training datasets that may contribute to disparities in healthcare delivery. Ethical considerations and regulatory frameworks are evolving, with governing bodies establishing guidelines for AI-driven medical applications. To fully harness the potential of AI, interdisciplinary collaboration among clinicians, data scientists, and engineers is essential, alongside open science initiatives to promote data accessibility and reproducibility. Future AI models must go beyond task automation, focusing instead on augmenting human expertise to enable proactive, precision-driven cardiovascular care. By embracing AI’s computational strengths while addressing its limitations, cardiology is poised to enter an era of transformative innovation beyond traditional diagnostic and therapeutic paradigms.

Graphical Abstract

1. Introduction

Artificial Intelligence (AI) has emerged as a game-changer in cardiovascular medicine, pushing the boundaries of what was once thought possible. Over the past decade, rapid advances in computational power and algorithmic sophistication have facilitated the transition from traditional machine learning (ML) techniques to deep learning (DL) models. These models have significantly enhanced diagnostic precision, prognosis assessment, treatment guidance, and risk stratification in cardiovascular care [1].
Despite AI’s potential, its current applications in cardiology and vascular diseases remain largely focused on automating human tasks, such as detecting arrhythmias in electrocardiograms (ECGs) or segmenting cardiac and vascular structures in imaging. While these developments might improve workflow efficiency, they primarily emphasize automation rather than true innovation in leveraging AI for novel discoveries and clinical breakthroughs [2,3]. Human cognition excels in pattern recognition and the application of experiential knowledge but remains inherently limited in tasks requiring large-scale data integration, complex temporal analysis, and parallel processing [4]. AI, on the other hand, is well suited for such challenges, as it can be trained on high-dimensional datasets from multiple sources—including ECG, echocardiography, cardiac magnetic resonance imaging (CMR), genomics, and clinical and wearable sensor data—to extract clinically relevant insights that may not be immediately apparent through conventional analysis (Figure 1). By shifting from task replication to augmentation and discovery, AI holds the potential to advance precision medicine in cardiology, enabling more accurate diagnosis, refined risk stratification, and personalized treatment strategies [5].
This work combines a review of the evolving role of AI in cardiovascular medicine, with a perspective-driven narrative and a forward-looking discussion on its transformative potential and future directions in clinical practice. It synthesizes evidence from recent studies, highlights AI’s strengths, and discusses key challenges in integrating AI-driven tools into routine clinical practice.

2. Human Cognition vs. Artificial Intelligence

2.1. Literature Search Strategy

We performed a structured literature search across PubMed, Scopus, and IEEE Xplore, covering publications from inception through March 2025. Our aim was to identify high-impact and clinically relevant studies on artificial intelligence in cardiovascular medicine. Search terms included combinations of the following keywords: (“artificial intelligence” or “machine learning” or “deep learning” or “neural networks”) and (“cardiology” or “cardiovascular” or “ECG” or “electrocardiogram” or “echocardiography” or “CMR” or “cardiac imaging” or “arrhythmia” or “heart failure” or “precision medicine” or “multi-omics”).
We included original research articles that focused on the development or implementation of AI methods for diagnosis, prognosis, risk stratification, or treatment of cardiovascular diseases, particularly those involving novel paradigms beyond traditional clinical tasks. We excluded non-English publications, non-original research (e.g., reviews or editorials), and studies unrelated to cardiovascular applications.
We also manually screened the references of the selected papers, through “snowballing” [6], to identify additional relevant studies not captured through database queries.

2.2. Human Expertise in Context

The way clinicians and AI process information is fundamentally distinct [7]. Clinicians integrate a combination of logical reasoning, experiential learning, subconscious pattern recognition, and contextual reasoning. When formulating a diagnosis, they synthesize multiple sources of information, including patient history, physical examination findings, laboratory results, imaging studies, and even non-verbal cues such as stress levels, lifestyle factors, or subtle hesitations in speech [8]. This ability to integrate complex, often unstructured data allows for intuitive, adaptable, and highly personalized decision-making.
In contrast, AI systems operate through mathematical optimization, identifying statistical and non-linear correlations across large datasets to mimic clinical workflows and established diagnostic pathways. However, this imitative approach is inherently limited [2,9]. AI often struggles to generalize to unseen data, often exhibiting poor performance in rare disease presentations or when confronted with patients with uncommon risk factors.
Furthermore, while AI excels in pattern recognition, it lacks the ability to incorporate real-world context, such as a patient’s social determinants of health or emotional state. This limitation reduces its adaptability in clinical settings, where dynamic decision-making is needed. Unlike human clinicians, AI does not possess the ability to question its own conclusions, apply ethical reasoning, or account for unseen variables. These constraints highlight why, despite AI’s capability to process vast amounts of data at unprecedented speeds, its real-world diagnostic accuracy often remains comparable, or sometimes inferior to that of experienced physicians [3,9]. For AI to make meaningful contributions to cardiovascular medicine, its role must evolve beyond naïve mimicry toward complementing human expertise by addressing challenges that are difficult or impossible for human cognition alone to resolve.

2.3. AI’s True Strength: Tackling Intractable Problems

Unlike human clinicians, who rely on experiential learning and intuition, AI operates through mathematical reasoning, enabling it to process immense amounts of complex data at speeds far beyond human cognition (Figure 2). AI can analyze thousands of ECGs, echocardiograms, or genomic datasets within seconds, uncovering patterns that even the most skilled cardiologists might overlook [10,11]. Its ability to integrate high-dimensional, multimodal data—including imaging, genomics, biomarkers, and continuous monitoring—facilitates a level of analysis that exceeds traditional diagnostic methods.
Moreover, AI’s consistency eliminates cognitive fatigue, variability, and biases inherent in human decision-making. In high-risk clinical settings, where continuous real-time monitoring is essential, AI can provide uninterrupted assessment, detecting subtle physiological changes that might otherwise go unnoticed [12]. Perhaps AI’s most revolutionary potential lies in its ability to uncover hidden disease markers. Traditional diagnostic approaches rely on well-defined clinical features, such as ST-segment elevations in ECGs [13] or morphological abnormalities in echocardiograms [14]. In contrast, AI has demonstrated the capability to detect preclinical disease states by identifying subtle, latent patterns that would be imperceptible through conventional methods [15].
For instance, AI models have successfully predicted heart failure from ECGs long be-fore the onset of overt symptoms [16] and distinguished genetic cardiomyopathies using echocardiographic [17], ECG [18], and multimodal data. These insights represent a paradigm shift in cardiovascular diagnostics, moving from feature-based assessment to a data-driven approach capable of recognizing novel, previously undetectable disease markers.

3. From Data to Innovation: AI’s Transformative Applications

AI is no longer confined to detecting abnormalities; it is now uncovering subtle, pre-clinical markers of disease progression. Its applications extend from precision medicine to real-time monitoring from predictive analytics, reshaping cardiovascular diagnostics and risk assessment (Table 1). Next, we present some of the transformative applications of AI through three lenses: electrocardiography, cardiovascular imaging, and precision medicine and omics.

3.1. Electrocardiogram

AI models have demonstrated remarkable performance in extracting predictive signatures from standard 12-lead ECGs, facilitating early detection of left ventricular systolic dysfunction and atrial fibrillation even in sinus rhythm [16,19]. Beyond conventional diagnostics, AI-driven ECG analysis has proven effective in detecting hypertrophic cardiomyopathy [18], dynamically predicting malignant ventricular arrhythmias in patients with an implantable cardioverter-defibrillator (ICD) [20], and it has also succeeded in identifying patients with concealed long QT syndrome and even differentiating specific genetic subtypes [21]. These advancements underscore AI’s ability to recognize complex patterns imperceptible to human experts, often surpassing traditional biomarkers such as QTc measurements.
AI is also revolutionizing cardiovascular outcome prediction, particularly for ventricular arrhythmias and sudden cardiac death. For instance, a recent AI-driven model trained on 24-h ambulatory ECG recordings demonstrated superior performance in stratifying patients at risk for ventricular tachycardia compared to conventional risk assessment tools [22]. Moreover, integrating AI with wearable biosensors has enabled real-time prediction of cardiac events, such as atrial fibrillation and ventricular tachycardia [23]. A recent study introduced a DL model that uses smartwatch-derived R-to-R intervals to predict atrial fibrillation onset up to 30 min in advance [24], while another applied machine learning to Holter ECG recordings, significantly improving short-term arrhythmia risk prediction [25].

3.2. Cardiovascular Imaging

In cardiovascular imaging, advanced DL models have made significant strides in detecting complex conditions such as cardiac amyloidosis using multimodal data or even ECG alone [26,27], or improving pipelines by allowing automatic segmentation of cardiac chambers in computed tomography (CT) images [28]. AI-assisted echocardiography has further enhanced diagnostic precision by powering lesion localization and disease phenotyping across multiple cardiac conditions, including atrial septal defects, dilated cardiomyopathy, hypertrophic cardiomyopathy, and prior myocardial infarction [17].
Additionally, DL techniques have been successfully leveraged to predict late gadolinium enhancement (LGE) from echocardiographic data, an achievement that could revolutionize fibrosis detection by providing a widely accessible, ultrasound-based alternative to cardiac magnetic resonance imaging (MRI) [29]. AI models have also demonstrated high accuracy in differentiating ischemic from dilated cardiomyopathy [30], while the recent application of DL in echocardiographic imaging has allowed the genetic profiling of patients with hypertrophic cardiomyopathy [31]. Other modalities like MRI have also been used in multimodal settings for various tasks, including the identification of patients with non-ischemic cardiomyopathy at risk of lethal ventricular arrhythmias [32].

3.3. Precision Medicine and Omics

AI is also reshaping treatment strategies, particularly in the realm of personalized medicine. AI-driven phenomapping has been employed for guiding chest pain assessment [33], while multimodal imaging integration has optimized patient selection for ICD [34] or to identify phenotypes for acute and long-term response to atrial fibrillation ablation [35]. Moreover, AI is now increasingly integrated into interventional workflows, guiding precision-driven procedural planning across both cardiology and vascular domains. Fields of application include lesion assessment and stent sizing in percutaneous coronary interventions [36,37] or substrate localization and catheter guidance in electrophysiology procedures [38,39]. In vascular surgery and endovascular interventions, AI is adopted to support procedural planning and risk stratification in peripheral artery disease interventions [40].
Furthermore, AI’s fusion with genomics, transcriptomics, and metabolomics is revolutionizing biomarker discovery. Recent studies illustrate AI’s ability to integrate multi-omic datasets, uncovering complex molecular associations that would remain undetectable using traditional statistical methodologies [41,42]. By merging demographic, clinical, imaging, and multi-omics data, AI is ushering in a new era of precision medicine, one that moves beyond population-based guidelines to tailor therapeutic strategies to individual patients.

3.4. Preventive Cardiology

Artificial intelligence is redefining preventive cardiology by enhancing traditional risk stratification tools and enabling individualized interventions. Historically, cardiovascular prevention has relied heavily on population-derived risk scores such as the Framingham Risk Score [43,44], which, while useful at a population level, often fail to account for the nuances of individual risk profiles. AI overcomes these limitations by integrating diverse data streams—ranging from electronic health records and imaging to genomics, metabolomics, and social determinants of health—to generate dynamic, personalized risk predictions and outperform conventional scores [45].
An emerging paradigm in this realm is the identification of subclinical disease states or high-risk inflammatory phenotypes. For instance, deep learning applied to coronary computed tomography angiography (CCTA) has enabled automated quantification of pericoronary fat inflammation, which is an early driver of atherosclerotic plaque destabilization [46]. This is particularly relevant in prediabetic patients, where chronic low-grade inflammation is both a hallmark and a prognostic driver of disease progression. In this context, the studies have shown that in certain prediabetic populations, elevated levels of sodium-glucose cotransporter 2 (SGLT2) and leptin, alongside altered sirtuin 6 (SIRT6) expression and microRNA levels, were associated with higher rates of major adverse cardiac events [47]. AI could leverage such molecular and imaging biomarkers to detect inflammatory signatures in at-risk individuals and optimize therapeutic decisions, such as the early administration of metformin and other drugs, in such populations [48]. Another promising avenue is the integration of AI into wearable devices and remote monitoring systems to detect real-time physiologic perturbations indicative of increased cardiovascular risk. Coupled with digital twins or personalized health avatars, these systems could deliver tailored lifestyle, pharmacologic, or behavioral recommendations, thus translating molecular insights into everyday preventive care [49]. Finally, broader efforts like the CardioSight platform from Singapore represent cutting-edge AI-enabled surveillance systems that can visualize cardiovascular risk factor data geospatially and in real time. CardioSight maps parameters like HbA1c and lipid levels, not only to identify individuals at risk, but also expose population-level care gaps [50].
Table 1. Selection of key studies on advanced AI applications in cardiovascular medicine.
Table 1. Selection of key studies on advanced AI applications in cardiovascular medicine.
StudyTaskAI ApproachInput ModalitiesOutcomes and Explainability *
Agibetov et al., 2021 [51]Automatic diagnosis of CA using CMRPretrained VGG16 CNN (transfer learning)CMR imagingAUC: 0.96, SEN: 94%, SPE: 90%; similar performance to expert radiologists
Akita et al., 2024 [29]Predict myocardial fibrosis from TTECNNTTE, CMR (LGE presence) for ground truthAUC: 0.86; outperformed reference model based on clinical parameters
Attia et al., 2019 [16]Screen and predict future LVSD using ECGCNN (6 layers with Relu activation, batch normalization, max pooling, and spatial fusion)12-lead ECGAUC: 0.93, SEN: 86.3%, SPE: 85.7%, ACC: 85.7%; superior AUC compared to traditional screening
Attia et al., 2019 [19]Identify AF from normal sinus rhythm ECGsCNN12-lead ECGAUC: 0.87, SEN: 79.0%, SPE: 79.5%; outperformed standard risk scores
Bos et al., 2021 [52]Detect LQTS from 12-lead ECGsCNNs (10 blocks of convolutional, batch normalization, Relu, and max pooling layers)12-lead ECGAUC: 0.90; improved performance over QTc interval measurement-based diagnosis, particularly in concealed cases
Chen et al., 2024 [53]Classification of Fabry Cardiomyopathy vs. HCM3D Convolutional Neural Network (3D ResNet18)CMR imagingACC: 91%, AUC: 0.91, F1-score: 0.85; XAI with Grad-CAM
Cohen-Shelly et al., 2021 [54]Detection of moderate-to-severe AVS from ECGCNNECGAUC: 0.85, SEN: 78%, SPE: 74%; XAI with saliency maps
DeGroat et al., 2024 [41]Novel cardiovascular biomarkers using multi-omics dataXGBoost classifier with Bayesian hyperparameter tuningMulti-omics (RNA-seq, whole-genome sequencing, clinical demographics)High prediction accuracy for cardiovascular disease (identified biomarkers showed strong literature-based validation); XAI with SHAP
Drouard et al., 2024 [42]Prediction of cardiovascular risk factors from multi-omic dataSupervised and semi-supervised autoencoders, meta-learnersBlood-derived metabolomics, epigenetics, transcriptomicsAUC improvement of 0.09–0.14 for transcriptomics and 0.07–0.11 for metabolomics with transfer learning; feature importance analysis
Economou Lundeberg et al., 2023 [22]Predicting the risk of VT using 24-h ambulatory ECG dataElastic Net Regression ModelAmbulatory ECGAUC: 0.76, NPV of bottom quintile: 98.2%
Gavidia et al., 2024 [24]Prediction of AF onset using wearable device dataCNN (479 layers, EfficientNetV2)R-to-R interval signals from wearable devicesACC: 83%, F1-score: 85%, Warning time: 30.8 min before AF onset
Goto et al., 2021 [26]Automated detection of CA using AI-based ECG and TTE modelsECG model: 2D CNN (18 layers); Echo model: 3D-CNN (trained on video sequences of apical 4-chamber views)ECG and TTEECG model: C-statistics of 0.85–0.91 across multiple institutions; Echo model: C-statistics of 0.89–1.00; outperformed expert cardiologists across multiple datasets
Gregoire et al., 2024 [25]Prediction of short-term AF onset using HRV from Holter ECG recordingsXGBoost decision tree trained on HRV parameters2-lead Holter ECG dataAUC: 0.92, AUPRC: 0.92, ACC: 84.5%, SEN: 83.0%, SPE: 86.6%, F1-score: 86.2%
Grogan et al., 2021 [27]Detect CA from a standard 12-lead ECGDeep Neural Network (AI-enhanced ECG model) trained to predict CA presence12-lead ECG, single-lead, and 6-lead ECG subsetsAUC: 0.91, SEN: 0.84, SPE: 0.85, PPV: 0.86, NPV: 0.84; predicted CA more than 6 months before clinical diagnosis
Jentzer et al., 2021 [55]Identification of LVSD using ECGsCNN trained on nearly 100,000 ECGs 12-lead ECG, TTE for ground truthAUC: 0.83, SEN: 72.8%, SPE: 77.8%, NPV: 84.9%, ACC: 76.1%
Jiang et al., 2024 [21]Detection of congenital LQTS and differentiation of genotypes based on 12-lead ECGsCNN (based on ResNetv2, trained using Bayesian optimization)12-lead ECG, genetic testing for KCNQ1 and KCNH2 variantsLQTS detection AUC: 0.93, Genotype differentiation AUC: 0.91, SEN: 0.90, F1-score: 0.84; outperformed expert-measured QTc in detecting LQTS, including concealed cases
Ko et al., 2020 [18]Detection of HCM using a 12-lead ECGCNN12-lead ECGAUC: 0.96, SEN: 87%, SPE: 90%, PPV: 31%, NPV: 99%; outperformed standard ECG interpretation, distinguishing HCM from LVH and normal ECGs
Kolk et al., 2023 [34]Prediction of non-arrhythmic mortality in patients with primary prevention ICDXGBoost Multimodal data: 12-lead ECG and clinical variables (demographics, medical history, medications, lab values)AUC (internal validation): 0.90, AUC (external validation): 0.79, SEN: 70.3%, SPE: 69.6%, F1-score: 74.6%; outperformed traditional risk scores; XAI with SHAP and ECG heatmaps
Kolk et al., 2024 [32]Prediction of malignant ventricular arrhythmias in patients with non-ischemic heart failureDEEP RISK model: Residual VAE, XGBoostCMR (LGE), 12-lead ECG, clinical data (demographics, medical history, lab values, medications)AUC: 0.84, SEN: 0.98, SPE: 0.73, AUPRC: 0.31; XAI with SHAP and latent traversal visualizations
Lampert et al., 2023 [56]Prediction of LVSD in patients with PVCsPretrained CNN (ResNet-152)12-lead ECGAUC: 0.79 (internal validation), AUC: 0.85 (external validation); outperformed traditional PVC burden-based risk assessment methods; XAI with Grad-CAM
Lee et al., 2016 [57]Prediction of VT one hour before its occurrence using NNsNN with one hidden layer (5–13 hidden neurons)14 HRV and RRV parametersAUC: 0.93, SEN: 88.2%, SPE: 82.4%, ACC: 85.3%; outperformed traditional VT prediction methods
Lee et al., 2023 [58]Prediction of significant CAD on coronary CTA in asymptomatic individualsNN with multi-task learning and feature selectionCoronary CTA, demographics, clinical and laboratory dataAUC: 0.78, ACC: 71.6%; outperformed CAD consortium score (AUC: 0.67), UDF score (AUC: 0.71), and PCE (AUC: 0.72); XAI with SHAP
Libiseller-Egger et al., 2022 [59]Identification of the genetic basis of cardiovascular age and its impact on cardiovascular riskCNN12-lead ECG, GWAS using UK Biobank dataIdentified eight genetic loci associated with ECG-derived cardiovascular age, with a correlation between delta age and risk factors; feature importance analysis
Liu et al., 2023 [17]AI TTE for diagnosing four entities: ASD, DCM, HCM, and prior MIPre-trained Inception-V3 for feature extraction from single frames, followed by a multiple-layer 1D CNN for video-level classificationTTE (apical 4-chamber videos)AUCs: 0.99% (ASD), 0.98% (DCM), 0.99% (HCM), 0.98% (prior MI), and 0.98% (Normal); performed comparably to or better than cardiologists; XAI with CAM
Melo et al., 2023 [60]Detection of BrS from an ECG without a sodium channel blocker challengeFeed-forward NN (optimized with ensemble learning)12-lead ECG (9 leads used for classification: I, II, III, V1–V6)Training cohort: ACC: 84%, SEN: 85.4%, SPE: 82.4%, AUC: 0.91; Validation cohort: ACC: 88.4%, SEN: 79.6%, SPE: 93.6%, AUC: 0.93; outperformed clinicians in BrS diagnosis; XAI with ECG heatmaps
Morita et al., 2021 [31]Prediction of the positive genotype in patients with HCM using TTE imagesCNNTTE images (parasternal short- and long-axis, apical 2-, 3-, 4-, and 5-chamber views)AUC: 0.86 (CNN & Mayo Score) vs. 0.72 (Mayo Score), AUC: 0.84 (CNN & Toronto Score) vs. 0.75 (Toronto Score); improved SEN, SPE, PPV, and NPV; outperformed reference models; XAI with Grad-CAM
O’Driscoll et al., 2023 [61]Predicting MACE and all-cause mortality from LVEF and GLSMachine learning-based AI algorithm for automated TTE image processingAI-contoured left ventricular function from TTE images, GLS, and LVEFAI-calculated LVEF and GLS were independently associated with MACE
Oikonomou et al., 2024 [33]AI tool for guiding cardiac testing in stable chest pain patientsMachine-learning-derived algorithm (XGBoost) applied to phenomapping-based decision supportDemographic and clinical data (age, sex, BMI, hypertension, diabetes, lipid panel, medications, others)AI-aligned testing was associated with lower risk of MI or death; improved CAD detection rates in anatomical-first testing; feature importance analysis
Oikonomou et al., 2024 [62]Detection and risk stratification of AVS progression using a video-based AI biomarkerDeep learning-based video AI model (DASSi), saliency mapsTTE, CMRFaster AVS progression prediction, Hazard Ratios for aortic valve replacement: 4.97 for YNHHS cohort, 4.04 for CSMC cohort, 11.38 for UK Biobank; XAI with saliency maps and feature importance analysis
Raghunath et al., 2021 [63]Predicting new-onset AF from a 12-lead ECG to enable targeted screeningCNN trained on 12-lead ECG signals12-lead ECG, patient demographics (age, sex)AUC: 0.85, AUPRC: 0.22 for predicting AF within 1 year; SEN: 69%, SPE: 81%; Hazard Ratio for high- vs. low-risk groups: 7.2 over 30 years; outperformed CHARGE-AF and an XGBoost model using only age and sex
Sahashi et al., 2024 [64]Deep learning-based estimation of CMR tissue characteristics from TTEVideo-based CNN trained on TTE videos to predict CMR-derived labelsTTE, CMR for ground truthAUC: 0.56–0.87 for different CMR parameters
Schwartz et al., 2022 [38]Evaluating the feasibility, accuracy, and safety of AI-based left atrium reconstructionModel-based FAM using an AI algorithm trained on >300 left atrial CTAElectroanatomic mapping with CARTO 3, ICE, fluoroscopy, and cardiac CTAReduced mapping and fluoroscopy time during PVI; no major complications
Sun et al., 2024 [65]Detecting CAD from ECG and PCG signalsParallel CNN network with autoencoder, SVM classifierECG, PCGACC: 98.45%, SEN: 98.6%, SPE: 98.6%, F1-score: 98. 9%; outperformed various CAD detection methods
Surucu et al., 2021 [66]Prediction of AF episodes before they occur with HRVCNN with preprocessing stepsECG-derived HRVACC: 100%, PRE: 100%, SEN: 100%, F1-score: 100%
Tokodi et al., 2023 [67]Prediction of RVEF using 2D TTE videosSpatiotemporal CNNs, ensemble model of three best-performing networks, saliency maps2D TTE videos (apical 4-chamber view)Mean absolute error for RVEF: 4.57% (internal validation), 5.54% (external validation); ACC for RVEF <45%: 78.4% vs. human expert: 77.0%; performance comparable to human experts, but the model had higher SEN at the cost of SPE; XAI with 2D TTE heatmaps
Torres Soto et al., 2022 [68]Differentiating HCM from HTN LVH using multimodal AIMultimodal deep learning model (LVH-Fusion; late-average fusion, late-ranked fusion, and pre-trained and random late fusion models)ECG, TTEF1-score: 0.73 (HCM), 0.96 (HTN); SEN: 0.73; SPE: 0.96; outperforms single-modal models; false discovery rate reduced from 0.59 to 0.27; outperformed cardiologists in correctly classifying HCM and HTN; XAI with SHAP
Venkat et al., 2023 [69]Identification of genes associated with cardiovascular risk using RNA-seqRF model, recursive feature elimination, SelectKBest feature selectionRNA-seq (gene expression), demographic data (age, gender, race)ACC for HF: 90.9%, for AF: 95%, for other cardiovascular diseases: 95.9%; feature importance analysis
Yang et al., 2021 [70]Prediction of ischemia and prognosis from coronary plaque featuresMachine learning-based feature selection (Boruta algorithm, hierarchical clustering)Coronary CTA, FFR and clinical data for ground truth AUC for low FFR: 0.8, for 5-year vessel-oriented composite outcome: 0.71; improved ischemia prediction; feature importance analysis
Zhang et al., 2022 [71]Identification of CAD-related genes from gene networksGraph Convolutional Network, three-layer deep learning modelGene expression, gene interaction networksAUC: 0.75, AUPR: 0.78, ACC: 66.9%; better performance than other models
Zhang et al., 2023 [72]Identification of CA using TTE and machine learningVarious machine models: LR, RF, SVM, others; texture feature extractionTTE, myocardial texture analysisAUC: 0.71–0.81, across models; LR had the best performance (F1-score: 0.34, SEN: 0.21, SPE: 1.0); outperformed traditional TTE
Zhao et al., 2022 [73]Prediction of HF with preserved ejection fraction using DNA methylation and clinical dataDeep learning framework integrating Factorization Machine-based Neural Network, LASSO, and XGBoost-based feature selectionDNA methylation (CpG sites), clinical dataAUC: 0.90, Hosmer–Lemeshow statistic: 6.17 (p: 0.632); outperformed existing risk models
Zhou et al., 2023 [30]Differentiation of ischemic cardiomyopathy from DCM using TTEMachine learning models: XGBoost (best-performing), logistic regression, RF, NNTTE, demographic dataXGBoost: AUC: 0.93, ACC: 75%, SEN: 72%, SPE: 78% (internal validation); AUC: 0.80, ACC: 78%, SEN: 64%, SPE: 93% (external validation)
* Explainability mechanisms are reported only for studies that explicitly incorporate such methods. Abbreviations: ACC, accuracy; AF, atrial fibrillation; AI, artificial intelligence; NN, neural network; ASD, atrial septal defect; AUC, area under the curve; AUPRC, area under the precision–recall curve; AVS, aortic valve stenosis; BMI, body mass index; BrS, Brugada syndrome; CA, cardiac amyloidosis; CAD, Coronary Artery disease; CAM, class activation mapping; CHARGE-AF, cohorts for heart and aging research in genomic epidemiology–atrial fibrillation score; CMR, cardiac magnetic resonance; CNN, convolutional neural network; CTA, computed tomography angiography; DCM, dilated cardiomyopathy; ECG, electrocardiogram; F1-score, F1 Score (harmonic mean of precision and recall); FAM, fast anatomical mapping; FFR, fractional flow reserve; GLS, global longitudinal strain; Grad-CAM, Gradient-weighted Class Activation Mapping; GWAS, genome-wide association study; HCM, hypertrophic cardiomyopathy; HF, heart failure; HRV, heart rate variability; HTN, hypertension; ICD, implantable cardioverter-defibrillator; ICE, intracardiac echocardiography; KCNH2/KCNQ1, potassium voltage-gated channel subfamily h member 2/Q member 1; LGE, late gadolinium enhancement; LR, logistic regression; LQTS, long QT syndrome; LVEF/RVEF, left/right ventricular ejection fraction; LVH, left ventricular hypertrophy; LVSD, left ventricular systolic dysfunction; MACE, major adverse cardiac events; MI, myocardial infarction; NPV, negative predictive value; PCG, phonocardiogram; PRE, precision; PVC, premature ventricular complexes; PVI, pulmonary vein isolation; RF, random forest; RNA, ribonucleic acid; RRV, respiratory rate variability; SEN, sensitivity; SHAP, SHapley Additive exPlanations; SPE, specificity; SVM, support vector machine; TTE, transthoracic echocardiography; VAE, variational autoencoder; VGG16, visual geometry group 16-layer CNN; VT, ventricular tachycardia; XAI, explainable artificial intelligence; XGBoost, extreme gradient boosting.

4. Challenges and Future Opportunities

While artificial intelligence is reshaping cardiovascular care, several challenges must be addressed to ensure its effective and ethical integration into clinical practice. These hurdles range from technical limitations to regulatory and ethical concerns. Overcoming them will require interdisciplinary collaboration, continuous technological advancements, and a commitment to transparency and equity in AI applications.

4.1. The Black-Box Problem: Enhancing AI Transparency and Explainability

One of the most significant challenges in medicinal AI is its “black-box” nature, where models generate predictions without clear explanations in their decision-making process [9]. DL models, particularly neural network-based ones, rely on complex, multi-layered computations that make it difficult for clinicians to interpret or validate their outputs. This lack of transparency fosters skepticism among medical professionals and hinders clinical adoption.
To mitigate this, the field is increasingly embracing explainable AI (XAI), which focuses on developing models that can provide human-understandable explanations for their predictions. Techniques such as SHapley Additive Explanations (SHAP) and feature importance scoring [74], and Gradient-weighted Class Activation Mapping (Grad-CAM) and saliency maps [75], are now starting to appear, offering insights into what the model focuses on. As detailed in Table 1, some recent paradigms of AI in cardiovascular care have also started to adopt XAI methods, from SHAP values to rank pre-determined features [20,34,41,58], to heatmap visualizations for raw ECG signals and imaging [17,31,53,60]. These approaches provide transparency, improve clinical trust, and support hypothesis generation in biomarker discovery and phenotyping.
However, important limitations remain. For instance, saliency maps often only indicate where the model is attending (e.g., a region of an image or a time series segment), but not necessarily why that focus leads to a given classification [76]. Similarly, methods like SHAP provide statistical insights into feature contributions but may not reflect true causal relationships or biological plausibility [77]. Despite the efforts to integrate XAI with clinical reasoning and provide standardized frameworks for AI development in cardiology [78], the complexity of interpreting multimodal model outputs and challenges in validating XAI-derived insights at the bedside represent ongoing gaps. Therefore, while significant progress has been made in embedding explainability into AI systems—especially in genomics, imaging, and electrophysiology—further advances are needed to ensure transparency and trustworthiness in AI-enhanced clinical decision-making. Future efforts should aim to develop clinically-grounded XAI frameworks, integrate domain-specific ontologies, and explore causality-aware explanations that can bridge the gap between prediction and actionable understanding.

4.2. Bias in AI Models: Ensuring Fair and Equitable AI

AI models are only as good as the data they are trained on, and biases present in training datasets can lead to systemic errors in AI-based decision-making [2]. If an AI model is primarily trained on datasets that do not adequately represent diverse populations—including underrepresented racial and ethnic groups, gender minorities, or individuals with rare conditions—it may produce biased and unreliable predictions when applied in broader clinical settings or unseen data.
The risks of biased AI extend beyond incorrect diagnoses; they can also exacerbate existing healthcare disparities. For example, if an AI system is primarily trained on ECG data from male patients, it may perform less accurately for female patients, leading to misdiagnoses or suboptimal treatment recommendations. Healthcare providers and AI developers must collaborate to ensure equitable AI-driven healthcare by incorporating diverse, representative datasets and implementing bias mitigation strategies.

4.3. Regulatory and Ethical Considerations

The deployment of AI in medicine exists at the intersection of healthcare, technology, regulation, and ethics. While AI-powered tools hold immense potential for improving diagnosis and treatment, their regulation remains a challenge. Regulatory bodies such as the Food and Drug Administration (FDA) [79], European Medicines Agency (EMA) [80], and Medicines and Healthcare products Regulatory Agency (MHRA) [81], are continually developing guidelines for AI-driven medical devices and services, but the rapid pace of AI innovation often outstrips regulatory frameworks.
Additionally, AI introduces ethical dilemmas concerning autonomy, liability, and informed consent. When an AI-driven decision results in patient harm, who bears responsibility: the clinician using the tool, the developers designing the algorithm, or the institution deploying it? Furthermore, how can patients give informed consent for AI-driven decisions when even experts struggle to fully understand how these models function? Establishing clear legal frameworks and ethical guidelines will be essential to integrating AI safely into patient care.

4.4. Interdisciplinary Collaboration: Bridging the Gap

AI in cardiovascular medicine sits at the intersection of multiple fields, requiring close collaboration across multiple disciplines, including clinicians, data scientists, biomedical engineers, and regulatory experts. One of the primary barriers to AI adoption is the disconnect between these fields; many clinicians lack the technical expertise to understand and trust AI models, while AI developers often have limited insight into clinical workflows and real-world patient care.
Bridging this gap will require interdisciplinary education and training programs that promote shared knowledge and communication. AI-driven research initiatives should include multidisciplinary teams, ensuring that models are clinically relevant, ethically sound, and aligned with practical medical needs.

4.5. Technological Advances and Open Science

Recent advancements in computational power and data infrastructure have significantly improved AI’s ability to process complex cardiovascular data. Some recent breakthroughs include:
  • High-performance computational hardware: advances in graphic processing units (GPU) and tensor processing units (TPU) enable AI models to analyze continuous ECG recordings, high-resolution imaging, and multimodal data integration in real-time, drastically improving efficiency in AI workflows [82].
  • Self-supervised learning: This method is an emerging paradigm that enables AI models to learn useful representations from unlabeled data by creating surrogate tasks, such as predicting missing parts of ECG signals or reconstructing corrupted imaging inputs. This is particularly valuable in cardiovascular medicine, where labeled datasets are limited and costly to obtain. Self-supervised learning has already shown promising results in certain areas of cardiovascular research, such as echocardiographic view classification [83,84] and anomaly detection in ECGs [85].
  • Federated learning: Federated learning allows multiple institutions to collaboratively train AI models without sharing raw patient data. Instead, model updates are exchanged, preserving patient privacy and complying with data protection regulations [86]. This decentralized approach facilitates access to diverse and representative datasets, improving model generalizability and robustness. Federated learning has already marked some use cases in the field, such as distributed ECG classification [87], cross-center imaging tasks [88], and model development for genomic research [89].
  • Foundational models in cardiovascular AI: Foundational models—large-scale, pre-trained neural networks initially developed for broad tasks—are increasingly adapted for cardiovascular medicine [90,91]. These models, such as HeartBEiT for ECG interpretation and EchoCLIP for echo-imaging analysis, are trained on vast and diverse datasets, can be fine-tuned for a variety of downstream clinical applications [92,93,94]. Similar to ChatGPT (OpenAI) in natural language processing, foundational models in medicine can capture universal patterns across different data modalities and patient populations, and offer enhanced performance, adaptability, and transferability to novel cardiovascular tasks, even in low-resource or data-scarce environments.
Despite these technological strides, data and code accessibility remain major barriers to AI-driven cardiology [2]. Many state-of-the-art AI models are developed within academic and private-sector silos, with limited transparency regarding their training and validation processes. This lack of open access hinders reproducibility, making it difficult for the broader scientific community to verify results and build upon existing advancements.
To address this, data-sharing initiatives and open-source AI models should be prioritized. Large-scale cardiovascular datasets, such as the UK Biobank [95] and the Multi-Ethnic Study of Atherosclerosis (MESA) [96], along with AI-driven repositories like PhysioNet [97], can facilitate collaborative research and innovation. Governments, research institutions, and industry leaders must balance data privacy concerns with the need for open AI development. Additionally, standardized evaluation benchmarks should be established to ensure that AI models are rigorously tested across diverse patient populations before clinical deployment. By fostering an open-science approach, AI advancements can be accelerated while maintaining transparency, fairness, and clinical trust.

5. Conclusions

Artificial intelligence is transforming cardiovascular medicine, not by replacing clinicians, but by augmenting their capabilities. AI should not be limited to automating existing workflows or mimicking human decision-making. From ECG interpretation to multi-omics integration for diagnostics, prognostics, and preventive medicine, AI excels where human cognition meets its limits: in synthesizing multimodal data and integrating diverse information sources to drive the next generation of precision medicine.
Challenges still persist since many models remain opaque, raising concerns around explainability, bias, and clinical trust. While recent advances in AI, such as XAI, federated learning, and foundational models, show promise, broader adoption will require transparent validation, open and diverse datasets, and stronger interdisciplinary collaboration.
The future of cardiovascular AI lies not in mimicking human decision-making but in extending it by empowering physicians with insights beyond human cognitive reach. We do not expect a bird to swim or a fish to fly; similarly, we should not request from AI to think like a human but rather to do what clinicians cannot. Embracing this synergy will redefine the frontiers of precision cardiovascular care.

Author Contributions

Conceptualization, P.P., P.D., R.D.L. and E.O.; methodology, P.P., S.R.-C., A.G. (Alexios Giannakodimos), O.K., K.Z., K.K., E.O. and G.S.; software, P.P., S.R.-C. and P.T.; validation, P.D., R.D.L., K.K., E.O. and G.S.; formal analysis, P.P., S.R.-C., A.G. (Athina Goliopoulou), A.G. (Alexios Giannakodimos) and P.T.; investigation, P.D., O.K., K.Z., K.K. and G.S.; resources, P.P., P.D., S.R.-C., R.D.L., O.K., K.Z., K.K. and G.S.; data curation, P.D., S.R.-C., R.D.L., O.K., K.Z., K.K. and G.S.; writing—original draft preparation, P.P., P.D., S.R.-C., A.G. (Athina Goliopoulou), A.G. (Alexios Giannakodimos) and P.T.; writing—review and editing, R.D.L., O.K., K.Z., K.K., E.O. and G.S.; visualization, P.P., S.R.-C. and E.O.; supervision, E.O. and G.S.; project administration, E.O. and G.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sun, X.; Yin, Y.; Yang, Q.; Huo, T. Artificial Intelligence in Cardiovascular Diseases: Diagnostic and Therapeutic Perspectives. Eur. J. Med. Res. 2023, 28, 242. [Google Scholar] [CrossRef]
  2. Faust, O.; Salvi, M.; Barua, P.D.; Chakraborty, S.; Molinari, F.; Acharya, U.R. Issues and Limitations on the Road to Fair and Inclusive AI Solutions for Biomedical Challenges. Sensors 2025, 25, 205. [Google Scholar] [CrossRef] [PubMed]
  3. Takita, H.; Kabata, D.; Walston, S.L.; Tatekawa, H.; Saito, K.; Tsujimoto, Y.; Miki, Y.; Ueda, D. Diagnostic Performance Comparison between Generative AI and Physicians: A Systematic Review and Meta-Analysis. medRxiv 2024. [Google Scholar] [CrossRef]
  4. Korteling, J.E.H.; Van De Boer-Visschedijk, G.C.; Blankendaal, R.A.M.; Boonekamp, R.C.; Eikelboom, A.R. Human-versus Artificial Intelligence. Front. Artif. Intell. 2021, 4, 622364. [Google Scholar] [CrossRef]
  5. Krittanawong, C.; Zhang, H.; Wang, Z.; Aydar, M.; Kitai, T. Artificial Intelligence in Precision Cardiovascular Medicine. J. Am. Coll. Cardiol. 2017, 69, 2657–2664. [Google Scholar] [CrossRef] [PubMed]
  6. Wohlin, C. Guidelines for Snowballing in Systematic Literature Studies and a Replication in Software Engineering. In Proceedings of the 18th International Conference on Evaluation and Assessment in Software Engineering, London, UK, 13–14 May 2014; pp. 1–10. [Google Scholar]
  7. Monteith, S.; Glenn, T.; Geddes, J.R.; Achtyes, E.D.; Whybrow, P.C.; Bauer, M. Differences between Human and Artificial/Augmented Intelligence in Medicine. Comput. Hum. Behav. Artif. Hum. 2024, 2, 100084. [Google Scholar] [CrossRef]
  8. Lussier, M.-T.; Richard, C. Handling Cues from Patients. Can. Fam. Physician Med. Fam. Can. 2009, 55, 1213–1214. [Google Scholar]
  9. Kelly, C.J.; Karthikesalingam, A.; Suleyman, M.; Corrado, G.; King, D. Key Challenges for Delivering Clinical Impact with Artificial Intelligence. BMC Med. 2019, 17, 195. [Google Scholar] [CrossRef]
  10. Pantelidis, P.; Bampa, M.; Oikonomou, E.; Papapetrou, P. Machine Learning Models for Automated Interpretation of 12-Lead Electrocardiographic Signals: A Narrative Review of Techniques, Challenges, Achievements and Clinical Relevance. J. Med. Artif. Intell. 2023, 6, 6. [Google Scholar] [CrossRef]
  11. Moradi, A.; Olanisa, O.O.; Nzeako, T.; Shahrokhi, M.; Esfahani, E.; Fakher, N.; Khazeei Tabari, M.A. Revolutionizing Cardiac Imaging: A Scoping Review of Artificial Intelligence in Echocardiography, CTA, and Cardiac MRI. J. Imaging 2024, 10, 193. [Google Scholar] [CrossRef]
  12. Pinsky, M.R.; Bedoya, A.; Bihorac, A.; Celi, L.; Churpek, M.; Economou-Zavlanos, N.J.; Elbers, P.; Saria, S.; Liu, V.; Lyons, P.G.; et al. Use of Artificial Intelligence in Critical Care: Opportunities and Obstacles. Crit. Care 2024, 28, 113. [Google Scholar] [CrossRef] [PubMed]
  13. Hampton, J.R.; Hampton, J. The ECG Made Easy, 9th ed.; Elsevier: Edinburgh, UK; London, UK; New York, NY, USA; Oxford, UK; Philadelphia, PA, USA; St. Louis, MO, USA; Sydney, Australia, 2019; ISBN 978-0-7020-7457-8. [Google Scholar]
  14. Kaddoura, S. Echo Made Easy, 3rd ed.; Elsevier: Edinburgh, UK; London, UK; New York, NY, USA; Oxford, UK; Philadelphia, PA, USA; St. Louis, MO, USA; Sydney, Australia; Toronto, ON, Canada, 2016; ISBN 978-0-7020-6656-6. [Google Scholar]
  15. Pantelidis, P.; Oikonomou, E.; Gialamas, I.; Goliopoulou, A.; Sarantos, S.; Zakynthinos, G.E.; Anastasiou, A.; Gounaridi, M.I.; Kalogeras, K.; Siasos, G. Decoding the Heart: How Artificial Intelligence Is Transforming Cardiology. J. Med. Artif. Intell. 2025, 8, 9. [Google Scholar] [CrossRef]
  16. Attia, Z.I.; Kapa, S.; Lopez-Jimenez, F.; McKie, P.M.; Ladewig, D.J.; Satam, G.; Pellikka, P.A.; Enriquez-Sarano, M.; Noseworthy, P.A.; Munger, T.M.; et al. Screening for Cardiac Contractile Dysfunction Using an Artificial Intelligence–Enabled Electrocardiogram. Nat. Med. 2019, 25, 70–74. [Google Scholar] [CrossRef] [PubMed]
  17. Liu, B.; Chang, H.; Yang, D.; Yang, F.; Wang, Q.; Deng, Y.; Li, L.; Lv, W.; Zhang, B.; Yu, L.; et al. A Deep Learning Framework Assisted Echocardiography with Diagnosis, Lesion Localization, Phenogrouping Heterogeneous Disease, and Anomaly Detection. Sci. Rep. 2023, 13, 3. [Google Scholar] [CrossRef]
  18. Ko, W.-Y.; Siontis, K.C.; Attia, Z.I.; Carter, R.E.; Kapa, S.; Ommen, S.R.; Demuth, S.J.; Ackerman, M.J.; Gersh, B.J.; Arruda-Olson, A.M.; et al. Detection of Hypertrophic Cardiomyopathy Using a Convolutional Neural Network-Enabled Electrocardiogram. J. Am. Coll. Cardiol. 2020, 75, 722–733. [Google Scholar] [CrossRef]
  19. Attia, Z.I.; Noseworthy, P.A.; Lopez-Jimenez, F.; Asirvatham, S.J.; Deshmukh, A.J.; Gersh, B.J.; Carter, R.E.; Yao, X.; Rabinstein, A.A.; Erickson, B.J.; et al. An Artificial Intelligence-Enabled ECG Algorithm for the Identification of Patients with Atrial Fibrillation during Sinus Rhythm: A Retrospective Analysis of Outcome Prediction. Lancet 2019, 394, 861–867. [Google Scholar] [CrossRef]
  20. Kolk, M.Z.H.; Ruipérez-Campillo, S.; Alvarez-Florez, L.; Deb, B.; Bekkers, E.J.; Allaart, C.P.; Van Der Lingen, A.-L.C.J.; Clopton, P.; Išgum, I.; Wilde, A.A.M.; et al. Dynamic Prediction of Malignant Ventricular Arrhythmias Using Neural Networks in Patients with an Implantable Cardioverter-Defibrillator. eBioMedicine 2024, 99, 104937. [Google Scholar] [CrossRef]
  21. Jiang, R.; Cheung, C.C.; Garcia-Montero, M.; Davies, B.; Cao, J.; Redfearn, D.; Laksman, Z.M.; Grondin, S.; Atallah, J.; Escudero, C.A.; et al. Deep Learning–Augmented ECG Analysis for Screening and Genotype Prediction of Congenital Long QT Syndrome. JAMA Cardiol. 2024, 9, 377. [Google Scholar] [CrossRef]
  22. Economou Lundeberg, J.; Måneheim, A.; Persson, A.; Dziubinski, M.; Sridhar, A.; Healey, J.S.; Slusarczyk, M.; Engström, G.; Johnson, L.S. Ventricular Tachycardia Risk Prediction with an Abbreviated Duration Mobile Cardiac Telemetry. Heart Rhythm O2 2023, 4, 500–505. [Google Scholar] [CrossRef]
  23. Kolk, M.Z.H.; Frodi, D.M.; Langford, J.; Jacobsen, P.K.; Risum, N.; Andersen, T.O.; Tan, H.L.; Hastrup Svendsen, J.; Knops, R.E.; Diederichsen, S.Z.; et al. Artificial Intelligence-Enhanced Wearable Technology Enables Ventricular Arrhythmia Prediction. Eur. Heart J. Digit. Health 2024, ztae069. [Google Scholar] [CrossRef]
  24. Gavidia, M.; Zhu, H.; Montanari, A.N.; Fuentes, J.; Cheng, C.; Dubner, S.; Chames, M.; Maison-Blanche, P.; Rahman, M.M.; Sassi, R.; et al. Early Warning of Atrial Fibrillation Using Deep Learning. Patterns 2024, 5, 100970. [Google Scholar] [CrossRef] [PubMed]
  25. Gregoire, J.M.; Gilon, C.; Bersini, H.; Groben, L.; Nguyen, T.; Godart, P.; Carlier, S. Heart Rate Variability to Predict 1-Day Atrial Fibrillation Episode Onset Using Machine Learning. Eur. Heart J. 2024, 45, ehae666.339. [Google Scholar] [CrossRef]
  26. Goto, S.; Mahara, K.; Beussink-Nelson, L.; Ikura, H.; Katsumata, Y.; Endo, J.; Gaggin, H.K.; Shah, S.J.; Itabashi, Y.; MacRae, C.A.; et al. Artificial Intelligence-Enabled Fully Automated Detection of Cardiac Amyloidosis Using Electrocardiograms and Echocardiograms. Nat. Commun. 2021, 12, 2726. [Google Scholar] [CrossRef]
  27. Grogan, M.; Lopez-Jimenez, F.; Cohen-Shelly, M.; Dispenzieri, A.; Attia, Z.I.; Abou Ezzedine, O.F.; Lin, G.; Kapa, S.; Borgeson, D.D.; Friedman, P.A.; et al. Artificial Intelligence–Enhanced Electrocardiogram for the Early Detection of Cardiac Amyloidosis. Mayo Clin. Proc. 2021, 96, 2768–2778. [Google Scholar] [CrossRef]
  28. Feng, R.; Deb, B.; Ganesan, P.; Tjong, F.V.Y.; Rogers, A.J.; Ruipérez-Campillo, S.; Somani, S.; Clopton, P.; Baykaner, T.; Rodrigo, M.; et al. Segmenting Computed Tomograms for Cardiac Ablation Using Machine Learning Leveraged by Domain Knowledge Encoding. Front. Cardiovasc. Med. 2023, 10, 1189293. [Google Scholar] [CrossRef]
  29. Akita, K.; Kusunose, K.; Haga, A.; Shimomura, T.; Kosaka, Y.; Ishiyama, K.; Hasegawa, K.; Fifer, M.A.; Maurer, M.S.; Shimada, Y.J. Deep Learning of Echocardiography Distinguishes between Presence and Absence of Late Gadolinium Enhancement on Cardiac Magnetic Resonance in Patients with Hypertrophic Cardiomyopathy. Echo Res. Pract. 2024, 11, 23. [Google Scholar] [CrossRef] [PubMed]
  30. Zhou, M.; Deng, Y.; Liu, Y.; Su, X.; Zeng, X. Echocardiography-Based Machine Learning Algorithm for Distinguishing Ischemic Cardiomyopathy from Dilated Cardiomyopathy. BMC Cardiovasc. Disord. 2023, 23, 476. [Google Scholar] [CrossRef] [PubMed]
  31. Morita, S.X.; Kusunose, K.; Haga, A.; Sata, M.; Hasegawa, K.; Raita, Y.; Reilly, M.P.; Fifer, M.A.; Maurer, M.S.; Shimada, Y.J. Deep Learning Analysis of Echocardiographic Images to Predict Positive Genotype in Patients with Hypertrophic Cardiomyopathy. Front. Cardiovasc. Med. 2021, 8, 669860. [Google Scholar] [CrossRef]
  32. Kolk, M.Z.H.; Ruipérez-Campillo, S.; Allaart, C.P.; Wilde, A.A.M.; Knops, R.E.; Narayan, S.M.; Tjong, F.V.Y.; DEEP RISK investigators. Multimodal Explainable Artificial Intelligence Identifies Patients with Non-Ischaemic Cardiomyopathy at Risk of Lethal Ventricular Arrhythmias. Sci. Rep. 2024, 14, 14889. [Google Scholar] [CrossRef]
  33. Oikonomou, E.K.; Aminorroaya, A.; Dhingra, L.S.; Partridge, C.; Velazquez, E.J.; Desai, N.R.; Krumholz, H.M.; Miller, E.J.; Khera, R. Real-World Evaluation of an Algorithmic Machine-Learning-Guided Testing Approach in Stable Chest Pain: A Multinational, Multicohort Study. Eur. Heart J. Digit. Health 2024, 5, 303–313. [Google Scholar] [CrossRef]
  34. Kolk, M.Z.H.; Ruipérez-Campillo, S.; Deb, B.; Bekkers, E.J.; Allaart, C.P.; Rogers, A.J.; Van Der Lingen, A.-L.C.J.; Alvarez Florez, L.; Isgum, I.; De Vos, B.D.; et al. Optimizing Patient Selection for Primary Prevention Implantable Cardioverter-Defibrillator Implantation: Utilizing Multimodal Machine Learning to Assess Risk of Implantable Cardioverter-Defibrillator Non-Benefit. EP Eur. 2023, 25, euad271. [Google Scholar] [CrossRef] [PubMed]
  35. Pirruccello, J.P.; Di Achille, P.; Choi, S.H.; Rämö, J.T.; Khurshid, S.; Nekoui, M.; Jurgens, S.J.; Nauffal, V.; Kany, S.; FinnGen; et al. Deep Learning of Left Atrial Structure and Function Provides Link to Atrial Fibrillation Risk. Nat. Commun. 2024, 15, 4304. [Google Scholar] [CrossRef] [PubMed]
  36. Shin, D.; Sami, Z.; Cannata, M.; Ciftcikal, Y.; Caron, E.; Thomas, S.V.; Porter, C.R.; Tsioulias, A.; Gujja, M.; Sakai, K.; et al. Artificial Intelligence in Intravascular Imaging for Percutaneous Coronary Interventions: A New Era of Precision. J. Soc. Cardiovasc. Angiogr. Interv. 2025, 4, 102506. [Google Scholar] [CrossRef]
  37. Samant, S.; Panagopoulos, A.N.; Wu, W.; Zhao, S.; Chatzizisis, Y.S. Artificial Intelligence in Coronary Artery Interventions: Preprocedural Planning and Procedural Assistance. J. Soc. Cardiovasc. Angiogr. Interv. 2025, 4, 102519. [Google Scholar] [CrossRef] [PubMed]
  38. Schwartz, A.L.; Chorin, E.; Mann, T.; Levi, M.Y.; Hochstadt, A.; Margolis, G.; Viskin, S.; Banai, S.; Rosso, R. Reconstruction of the Left Atrium for Atrial Fibrillation Ablation Using the Machine Learning CARTO 3 M-FAM Software. J. Interv. Card. Electrophysiol. 2022, 64, 39–47. [Google Scholar] [CrossRef]
  39. Bahlke, F.; Englert, F.; Popa, M.; Bourier, F.; Reents, T.; Lennerz, C.; Kraft, H.; Martinez, A.T.; Kottmaier, M.; Syväri, J.; et al. First Clinical Data on Artificial Intelligence-guided Catheter Ablation in Long-standing Persistent Atrial Fibrillation. J. Cardiovasc. Electrophysiol. 2024, 35, 406–414. [Google Scholar] [CrossRef]
  40. Martelli, E.; Capoccia, L.; Di Francesco, M.; Cavallo, E.; Pezzulla, M.G.; Giudice, G.; Bauleo, A.; Coppola, G.; Panagrosso, M. Current Applications and Future Perspectives of Artificial and Biomimetic Intelligence in Vascular Surgery and Peripheral Artery Disease. Biomimetics 2024, 9, 465. [Google Scholar] [CrossRef]
  41. DeGroat, W.; Abdelhalim, H.; Peker, E.; Sheth, N.; Narayanan, R.; Zeeshan, S.; Liang, B.T.; Ahmed, Z. Multimodal AI/ML for Discovering Novel Biomarkers and Predicting Disease Using Multi-Omics Profiles of Patients with Cardiovascular Diseases. Sci. Rep. 2024, 14, 26503. [Google Scholar] [CrossRef]
  42. Drouard, G.; Mykkänen, J.; Heiskanen, J.; Pohjonen, J.; Ruohonen, S.; Pahkala, K.; Lehtimäki, T.; Wang, X.; Ollikainen, M.; Ripatti, S.; et al. Exploring Machine Learning Strategies for Predicting Cardiovascular Disease Risk Factors from Multi-Omic Data. BMC Med. Inform. Decis. Mak. 2024, 24, 116. [Google Scholar] [CrossRef]
  43. Wilson, P.W.F.; D’Agostino, R.B.; Levy, D.; Belanger, A.M.; Silbershatz, H.; Kannel, W.B. Prediction of Coronary Heart Disease Using Risk Factor Categories. Circulation 1998, 97, 1837–1847. [Google Scholar] [CrossRef]
  44. Sytkowski, P.A.; Kannel, W.B.; D’Agostino, R.B. Changes in Risk Factors and the Decline in Mortality from Cardiovascular Disease: The Framingham Heart Study. N. Engl. J. Med. 1990, 322, 1635–1641. [Google Scholar] [CrossRef] [PubMed]
  45. Weng, S.F.; Reps, J.; Kai, J.; Garibaldi, J.M.; Qureshi, N. Can Machine-Learning Improve Cardiovascular Risk Prediction Using Routine Clinical Data? PLoS ONE 2017, 12, e0174944. [Google Scholar] [CrossRef]
  46. Gerculy, R.; Benedek, I.; Kovács, I.; Rat, N.; Halațiu, V.B.; Rodean, I.; Bordi, L.; Blîndu, E.; Roșca, A.; Mátyás, B.-B.; et al. CT-Assessment of Epicardial Fat Identifies Increased Inflammation at the Level of the Left Coronary Circulation in Patients with Atrial Fibrillation. J. Clin. Med. 2024, 13, 1307. [Google Scholar] [CrossRef]
  47. Sardu, C.; Paolisso, G.; Marfella, R. Inflammatory Related Cardiovascular Diseases: From Molecular Mechanisms to Therapeutic Targets. Curr. Pharm. Des. 2020, 26, 2565–2573. [Google Scholar] [CrossRef] [PubMed]
  48. Sardu, C.; D’Onofrio, N.; Torella, M.; Portoghese, M.; Mureddu, S.; Loreni, F.; Ferraraccio, F.; Panarese, I.; Trotta, M.C.; Gatta, G.; et al. Metformin Therapy Effects on the Expression of Sodium-Glucose Cotransporter 2, Leptin, and SIRT6 Levels in Pericoronary Fat Excised from Pre-Diabetic Patients with Acute Myocardial Infarction. Biomedicines 2021, 9, 904. [Google Scholar] [CrossRef]
  49. Rudnicka, Z.; Proniewska, K.; Perkins, M.; Pregowska, A. Cardiac Healthcare Digital Twins Supported by Artificial Intelligence-Based Algorithms and Extended Reality—A Systematic Review. Electronics 2024, 13, 866. [Google Scholar] [CrossRef]
  50. Dalakoti, M.; Wong, S.; Lee, W.; Lee, J.; Yang, H.; Loong, S.; Loh, P.H.; Tyebally, S.; Djohan, A.; Ong, J.; et al. Incorporating AI into Cardiovascular Diseases Prevention-Insights from Singapore. Lancet Reg. Health West. Pac. 2024, 48, 101102. [Google Scholar] [CrossRef]
  51. Agibetov, A.; Kammerlander, A.; Duca, F.; Nitsche, C.; Koschutnik, M.; Donà, C.; Dachs, T.-M.; Rettl, R.; Stria, A.; Schrutka, L.; et al. Convolutional Neural Networks for Fully Automated Diagnosis of Cardiac Amyloidosis by Cardiac Magnetic Resonance Imaging. J. Pers. Med. 2021, 11, 1268. [Google Scholar] [CrossRef]
  52. Bos, J.M.; Attia, Z.I.; Albert, D.E.; Noseworthy, P.A.; Friedman, P.A.; Ackerman, M.J. Use of Artificial Intelligence and Deep Neural Networks in Evaluation of Patients with Electrocardiographically Concealed Long QT Syndrome from the Surface 12-Lead Electrocardiogram. JAMA Cardiol. 2021, 6, 532. [Google Scholar] [CrossRef]
  53. Chen, W.-W.; Kuo, L.; Lin, Y.-X.; Yu, W.-C.; Tseng, C.-C.; Lin, Y.-J.; Huang, C.-C.; Chang, S.-L.; Wu, J.C.-H.; Chen, C.-K.; et al. A Deep Learning Approach to Classify Fabry Cardiomyopathy from Hypertrophic Cardiomyopathy Using Cine Imaging on Cardiac Magnetic Resonance. Int. J. Biomed. Imaging 2024, 2024, 6114826. [Google Scholar] [CrossRef]
  54. Cohen-Shelly, M.; Attia, Z.I.; Friedman, P.A.; Ito, S.; Essayagh, B.A.; Ko, W.-Y.; Murphree, D.H.; Michelena, H.I.; Enriquez-Sarano, M.; Carter, R.E.; et al. Electrocardiogram Screening for Aortic Valve Stenosis Using Artificial Intelligence. Eur. Heart J. 2021, 42, 2885–2896. [Google Scholar] [CrossRef] [PubMed]
  55. Jentzer, J.C.; Kashou, A.H.; Attia, Z.I.; Lopez-Jimenez, F.; Kapa, S.; Friedman, P.A.; Noseworthy, P.A. Left Ventricular Systolic Dysfunction Identification Using Artificial Intelligence-Augmented Electrocardiogram in Cardiac Intensive Care Unit Patients. Int. J. Cardiol. 2021, 326, 114–123. [Google Scholar] [CrossRef]
  56. Lampert, J.; Vaid, A.; Whang, W.; Koruth, J.; Miller, M.A.; Langan, M.-N.; Musikantow, D.; Turagam, M.; Maan, A.; Kawamura, I.; et al. A Novel ECG-Based Deep Learning Algorithm to Predict Cardiomyopathy in Patients with Premature Ventricular Complexes. JACC Clin. Electrophysiol. 2023, 9, 1437–1451. [Google Scholar] [CrossRef] [PubMed]
  57. Lee, H.; Shin, S.-Y.; Seo, M.; Nam, G.-B.; Joo, S. Prediction of Ventricular Tachycardia One Hour before Occurrence Using Artificial Neural Networks. Sci. Rep. 2016, 6, 32390. [Google Scholar] [CrossRef]
  58. Lee, H.; Kang, B.G.; Jo, J.; Park, H.E.; Yoon, S.; Choi, S.-Y.; Kim, M.J. Deep Learning-Based Prediction for Significant Coronary Artery Stenosis on Coronary Computed Tomography Angiography in Asymptomatic Populations. Front. Cardiovasc. Med. 2023, 10, 1167468. [Google Scholar] [CrossRef] [PubMed]
  59. Libiseller-Egger, J.; Phelan, J.E.; Attia, Z.I.; Benavente, E.D.; Campino, S.; Friedman, P.A.; Lopez-Jimenez, F.; Leon, D.A.; Clark, T.G. Deep Learning-Derived Cardiovascular Age Shares a Genetic Basis with Other Cardiac Phenotypes. Sci. Rep. 2022, 12, 22625. [Google Scholar] [CrossRef]
  60. Melo, L.; Ciconte, G.; Christy, A.; Vicedomini, G.; Anastasia, L.; Pappone, C.; Grant, E. Deep Learning Unmasks the ECG Signature of Brugada Syndrome. PNAS Nexus 2023, 2, pgad327. [Google Scholar] [CrossRef]
  61. O’Driscoll, J.M.; Tuttolomondo, D.; Gaibazzi, N. Artificial Intelligence Calculated Global Longitudinal Strain and Left Ventricular Ejection Fraction Predicts Cardiac Events and All-cause Mortality in Patients with Chest Pain. Echocardiography 2023, 40, 1356–1364. [Google Scholar] [CrossRef]
  62. Oikonomou, E.K.; Holste, G.; Yuan, N.; Coppi, A.; McNamara, R.L.; Haynes, N.A.; Vora, A.N.; Velazquez, E.J.; Li, F.; Menon, V.; et al. A Multimodal Video-Based AI Biomarker for Aortic Stenosis Development and Progression. JAMA Cardiol. 2024, 9, 534. [Google Scholar] [CrossRef]
  63. Raghunath, S.; Pfeifer, J.M.; Ulloa-Cerna, A.E.; Nemani, A.; Carbonati, T.; Jing, L.; vanMaanen, D.P.; Hartzel, D.N.; Ruhl, J.A.; Lagerman, B.F.; et al. Deep Neural Networks Can Predict New-Onset Atrial Fibrillation From the 12-Lead ECG and Help Identify Those at Risk of Atrial Fibrillation–Related Stroke. Circulation 2021, 143, 1287–1298. [Google Scholar] [CrossRef]
  64. Sahashi, Y.; Ouyang, D.; Kwan, A. Using Deep Learning to Predict Cardiovascular Magnetic Resonance Findings from Echocardiography Videos. Eur. Heart J. 2024, 45, ehae666.3458. [Google Scholar] [CrossRef]
  65. Sun, C.; Liu, C.; Wang, X.; Liu, Y.; Zhao, S. Coronary Artery Disease Detection Based on a Novel Multi-Modal Deep-Coding Method Using ECG and PCG Signals. Sensors 2024, 24, 6939. [Google Scholar] [CrossRef]
  66. Surucu, M.; Isler, Y.; Perc, M.; Kara, R. Convolutional Neural Networks Predict the Onset of Paroxysmal Atrial Fibrillation: Theory and Applications. Chaos Interdiscip. J. Nonlinear Sci. 2021, 31, 113119. [Google Scholar] [CrossRef] [PubMed]
  67. Tokodi, M.; Magyar, B.; Soós, A.; Takeuchi, M.; Tolvaj, M.; Lakatos, B.K.; Kitano, T.; Nabeshima, Y.; Fábián, A.; Szigeti, M.B.; et al. Deep Learning-Based Prediction of Right Ventricular Ejection Fraction Using 2D Echocardiograms. JACC Cardiovasc. Imaging 2023, 16, 1005–1018. [Google Scholar] [CrossRef]
  68. Soto, J.T.; Weston Hughes, J.; Sanchez, P.A.; Perez, M.; Ouyang, D.; Ashley, E.A. Multimodal Deep Learning Enhances Diagnostic Precision in Left Ventricular Hypertrophy. Eur. Heart J. Digit. Health 2022, 3, 380–389. [Google Scholar] [CrossRef]
  69. Venkat, V.; Abdelhalim, H.; DeGroat, W.; Zeeshan, S.; Ahmed, Z. Investigating Genes Associated with Heart Failure, Atrial Fibrillation, and Other Cardiovascular Diseases, and Predicting Disease Using Machine Learning Techniques for Translational Research and Precision Medicine. Genomics 2023, 115, 110584. [Google Scholar] [CrossRef]
  70. Yang, S.; Koo, B.-K.; Hoshino, M.; Lee, J.M.; Murai, T.; Park, J.; Zhang, J.; Hwang, D.; Shin, E.-S.; Doh, J.-H.; et al. CT Angiographic and Plaque Predictors of Functionally Significant Coronary Disease and Outcome Using Machine Learning. JACC Cardiovasc. Imaging 2021, 14, 629–641. [Google Scholar] [CrossRef] [PubMed]
  71. Zhang, T.; Lin, Y.; He, W.; Yuan, F.; Zeng, Y.; Zhang, S. GCN-GENE: A Novel Method for Prediction of Coronary Heart Disease-Related Genes. Comput. Biol. Med. 2022, 150, 105918. [Google Scholar] [CrossRef]
  72. Zhang, X.; Liang, T.; Su, C.; Qin, S.; Li, J.; Zeng, D.; Cai, Y.; Huang, T.; Wu, J. Deep Learn-Based Computer-Assisted Transthoracic Echocardiography: Approach to the Diagnosis of Cardiac Amyloidosis. Int. J. Cardiovasc. Imaging 2023, 39, 955–965. [Google Scholar] [CrossRef]
  73. Zhao, X.; Sui, Y.; Ruan, X.; Wang, X.; He, K.; Dong, W.; Qu, H.; Fang, X. A Deep Learning Model for Early Risk Prediction of Heart Failure with Preserved Ejection Fraction by DNA Methylation Profiles Combined with Clinical Features. Clin. Epigenet. 2022, 14, 11. [Google Scholar] [CrossRef]
  74. Lundberg, S.M.; Lee, S.-I. A Unified Approach to Interpreting Model Predictions. In Proceedings of the 31st International Conference on Neural Information Processing Systems, Long Beach, CA, USA, 4–9 December 2017; pp. 4768–4777. [Google Scholar]
  75. Selvaraju, R.R.; Cogswell, M.; Das, A.; Vedantam, R.; Parikh, D.; Batra, D. Grad-CAM: Visual Explanations from Deep Networks via Gradient-Based Localization. In Proceedings of the 2017 IEEE International Conference on Computer Vision (ICCV), Venice, Italy, 22–29 October 2017; pp. 618–626. [Google Scholar]
  76. Arun, N.; Gaw, N.; Singh, P.; Chang, K.; Aggarwal, M.; Chen, B.; Hoebel, K.; Gupta, S.; Patel, J.; Gidwani, M.; et al. Assessing the (Un)Trustworthiness of Saliency Maps for Localizing Abnormalities in Medical Imaging. Radiol. Artif. Intell. 2020, 3, e200267. [Google Scholar] [CrossRef] [PubMed]
  77. Ma, S.; Tourani, R. Predictive and Causal Implications of Using Shapley Value for Model Interpretation. In Proceedings of the 2020 KDD Workshop on Causal Discovery, San Diego, CA, USA, 24 August 2020. [Google Scholar]
  78. Svennberg, E.; Han, J.K.; Caiani, E.G.; Engelhardt, S.; Ernst, S.; Friedman, P.; Garcia, R.; Ghanbari, H.; Hindricks, G.; Man, S.H.; et al. State of the Art of Artificial Intelligence in Clinical Electrophysiology in 2025. A Scientific Statement of the European Heart Rhythm Association (EHRA) of the ESC, the Heart Rhythm Society (HRS), and the ESC Working Group in e-Cardiology. EP Eur. 2025, euaf071. [Google Scholar] [CrossRef]
  79. U.S. Food and Drug Administration. Artificial Intelligence and Machine Learning (AI/ML)-Enabled Medical Devices. Available online: https://www.fda.gov/medical-devices/software-medical-device-samd/artificial-intelligence-and-machine-learning-aiml-enabled-medical-devices (accessed on 1 March 2025).
  80. European Medicines Agency. Artificial Intelligence—EMA. Available online: https://www.ema.europa.eu/en/about-us/how-we-work/big-data/artificial-intelligence (accessed on 1 March 2025).
  81. Medicines and Healthcare Products Regulatory Agency. MHRA’s AI Regulatory Strategy Ensures Patient Safety and Industry Innovation into 2030. Available online: https://www.gov.uk/government/news/mhras-ai-regulatory-strategy-ensures-patient-safety-and-industry-innovation-into-2030 (accessed on 1 March 2025).
  82. Nikolić, G.S.; Dimitrijević, B.R.; Nikolić, T.R.; Stojcev, M.K. Survey of Three Types of Processing Units: CPU, GPU and TPU. In Proceedings of the 2022 57th International Scientific Conference on Information, Communication and Energy Systems and Technologies (ICEST), Ohrid, North Macedonia, 16–18 June 2022; pp. 1–6. [Google Scholar]
  83. Huang, S.-C.; Pareek, A.; Jensen, M.; Lungren, M.P.; Yeung, S.; Chaudhari, A.S. Self-Supervised Learning for Medical Image Classification: A Systematic Review and Implementation Guidelines. npj Digit. Med. 2023, 6, 74. [Google Scholar] [CrossRef] [PubMed]
  84. Mugambi, L.; Wa Maina, C.; Zühlke, L. Self-Supervised Multi-Task Learning for the Detection and Classification of RHD-Induced Valvular Pathology. J. Imaging 2025, 11, 97. [Google Scholar] [CrossRef]
  85. Mehari, T.; Strodthoff, N. Self-Supervised Representation Learning from 12-Lead ECG Data. Comput. Biol. Med. 2022, 141, 105114. [Google Scholar] [CrossRef]
  86. European Union Regulation (EU) 2016/679 of the European Parliament and of the Council of 27 April 2016 on the Protection of Natural Persons with Regard to the Processing of Personal Data and on the Free Movement of Such Data (General Data Protection Regulation). 2016. Available online: https://eur-lex.europa.eu/eli/reg/2016/679/oj/eng (accessed on 1 March 2025).
  87. Manocha, A.; Sood, S.K.; Bhatia, M. Federated Learning-Inspired Smart ECG Classification: An Explainable Artificial Intelligence Approach. Multimed. Tools Appl. 2024. [Google Scholar] [CrossRef]
  88. Goto, S.; Solanki, D.; John, J.E.; Yagi, R.; Homilius, M.; Ichihara, G.; Katsumata, Y.; Gaggin, H.K.; Itabashi, Y.; MacRae, C.A.; et al. Multinational Federated Learning Approach to Train ECG and Echocardiogram Models for Hypertrophic Cardiomyopathy Detection. Circulation 2022, 146, 755–769. [Google Scholar] [CrossRef] [PubMed]
  89. Calvino, G.; Peconi, C.; Strafella, C.; Trastulli, G.; Megalizzi, D.; Andreucci, S.; Cascella, R.; Caltagirone, C.; Zampatti, S.; Giardina, E. Federated Learning: Breaking Down Barriers in Global Genomic Research. Genes 2024, 15, 1650. [Google Scholar] [CrossRef]
  90. Mathew, G.; Barbosa, D.; Prince, J.; Venkatraman, S. Foundation Models for Cardiovascular Disease Detection via Biosignals from Digital Stethoscopes. npj Cardiovasc. Health 2024, 1, 25. [Google Scholar] [CrossRef]
  91. Quer, G.; Topol, E.J. The Potential for Large Language Models to Transform Cardiovascular Medicine. Lancet Digit. Health 2024, 6, e767–e771. [Google Scholar] [CrossRef]
  92. Vaid, A.; Jiang, J.; Sawant, A.; Lerakis, S.; Argulian, E.; Ahuja, Y.; Lampert, J.; Charney, A.; Greenspan, H.; Narula, J.; et al. A Foundational Vision Transformer Improves Diagnostic Performance for Electrocardiograms. npj Digit. Med. 2023, 6, 108. [Google Scholar] [CrossRef] [PubMed]
  93. Christensen, M.; Vukadinovic, M.; Yuan, N.; Ouyang, D. Vision–Language Foundation Model for Echocardiogram Interpretation. Nat. Med. 2024, 30, 1481–1488. [Google Scholar] [CrossRef] [PubMed]
  94. Feng, R.; Brennan, K.A.; Azizi, Z.; Goyal, J.; Deb, B.; Chang, H.J.; Ganesan, P.; Clopton, P.; Pedron, M.; Ruipérez-Campillo, S.; et al. Engineering of Generative Artificial Intelligence and Natural Language Processing Models to Accurately Identify Arrhythmia Recurrence. Circ. Arrhythm. Electrophysiol. 2025, 18, e013023. [Google Scholar] [CrossRef] [PubMed]
  95. Sudlow, C.; Gallacher, J.; Allen, N.; Beral, V.; Burton, P.; Danesh, J.; Downey, P.; Elliott, P.; Green, J.; Landray, M.; et al. UK Biobank: An Open Access Resource for Identifying the Causes of a Wide Range of Complex Diseases of Middle and Old Age. PLoS Med. 2015, 12, e1001779. [Google Scholar] [CrossRef]
  96. Bild, D.E. Multi-Ethnic Study of Atherosclerosis: Objectives and Design. Am. J. Epidemiol. 2002, 156, 871–881. [Google Scholar] [CrossRef]
  97. Goldberger, A.L.; Amaral, L.A.N.; Glass, L.; Hausdorff, J.M.; Ivanov, P.C.; Mark, R.G.; Mietus, J.E.; Moody, G.B.; Peng, C.-K.; Stanley, H.E. PhysioBank, PhysioToolkit, and PhysioNet: Components of a New Research Resource for Complex Physiologic Signals. Circulation 2000, 101, e215–e220. [Google Scholar] [CrossRef]
Figure 1. Pipeline of artificial intelligence (AI) model development in cardiovascular medicine.
Figure 1. Pipeline of artificial intelligence (AI) model development in cardiovascular medicine.
Biomedicines 13 01019 g001
Figure 2. Conceptual comparison of artificial intelligence and human cognition traits in clinical analytical thinking.
Figure 2. Conceptual comparison of artificial intelligence and human cognition traits in clinical analytical thinking.
Biomedicines 13 01019 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pantelidis, P.; Dilaveris, P.; Ruipérez-Campillo, S.; Goliopoulou, A.; Giannakodimos, A.; Theofilis, P.; De Lucia, R.; Katsarou, O.; Zisimos, K.; Kalogeras, K.; et al. Hearts, Data, and Artificial Intelligence Wizardry: From Imitation to Innovation in Cardiovascular Care. Biomedicines 2025, 13, 1019. https://doi.org/10.3390/biomedicines13051019

AMA Style

Pantelidis P, Dilaveris P, Ruipérez-Campillo S, Goliopoulou A, Giannakodimos A, Theofilis P, De Lucia R, Katsarou O, Zisimos K, Kalogeras K, et al. Hearts, Data, and Artificial Intelligence Wizardry: From Imitation to Innovation in Cardiovascular Care. Biomedicines. 2025; 13(5):1019. https://doi.org/10.3390/biomedicines13051019

Chicago/Turabian Style

Pantelidis, Panteleimon, Polychronis Dilaveris, Samuel Ruipérez-Campillo, Athina Goliopoulou, Alexios Giannakodimos, Panagiotis Theofilis, Raffaele De Lucia, Ourania Katsarou, Konstantinos Zisimos, Konstantinos Kalogeras, and et al. 2025. "Hearts, Data, and Artificial Intelligence Wizardry: From Imitation to Innovation in Cardiovascular Care" Biomedicines 13, no. 5: 1019. https://doi.org/10.3390/biomedicines13051019

APA Style

Pantelidis, P., Dilaveris, P., Ruipérez-Campillo, S., Goliopoulou, A., Giannakodimos, A., Theofilis, P., De Lucia, R., Katsarou, O., Zisimos, K., Kalogeras, K., Oikonomou, E., & Siasos, G. (2025). Hearts, Data, and Artificial Intelligence Wizardry: From Imitation to Innovation in Cardiovascular Care. Biomedicines, 13(5), 1019. https://doi.org/10.3390/biomedicines13051019

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop