Abstract
Artificial intelligence (AI) has the potential to transform every facet of cardiovascular practice and research. The exponential rise in technology powered by AI is defining new frontiers in cardiovascular care, with innovations that span novel diagnostic modalities, new digital native biomarkers of disease, and high-performing tools evaluating care quality and prognosticating clinical outcomes. These digital innovations promise expanded access to cardiovascular screening and monitoring, especially among those without historical access to high-quality, specialized care. Moreover, AI is propelling biological and clinical discoveries that will make future cardiovascular care more personalized, precise, and effective. The review brings together these diverse AI innovations, highlighting developments in multimodal cardiovascular AI across clinical practice and biomedical discovery, envisioning this new future backed by contemporary science and emerging discoveries. Finally, we define the critical path and the safeguards essential to realizing this AI-enabled future that helps achieve optimal cardiovascular health and outcomes for all.
Keywords: Artificial intelligence, machine learning, precision medicine, digital biomarkers, multiomics
CONDENSED ABSTRACT
Artificial intelligence (AI) will transform every facet of cardiovascular practice and research. The review summarizes how technology powered by AI is defining new frontiers in cardiovascular care, promising to expand access to cardiovascular screening, diagnosis, and monitoring, especially among those without historical access to high-quality, specialized care. Moreover, it summarizes how AI is propelling biological and clinical discoveries that will make future cardiovascular care more personalized, precise, and effective. We ground this vision in emerging scientific advances, while also defining the critical path and the safeguards essential to realizing this AI-enabled future that helps achieve optimal cardiovascular health for all.
Central Illustration |
The next era of AI in cardiovascular practice and discovery. The rapidly evolving landscape of medical AI will be characterized by progressive multimodal integration for scalable diagnosis and prognosis, multiomic development for the accelerated design and evaluation of novel therapeutics, and digital innovations to accelerate evidence generation through smart clinical trials. To ensure patient safety and a net societal benefit, AI algorithms would need to be prospectively assessed for their real-world effectiveness and developed within a robust regulatory framework with a focus on high-value and equitable care. AI: artificial intelligence.
1. Introduction
Artificial intelligence (AI) is poised to transform every facet of cardiovascular practice and research. Clinicians and scientists already interact with AI in their work, with current applications standardizing, accelerating, and expanding the scope of their traditional workflows.1,2 The simultaneous advances in the digitization of health data, the development of novel computational methods, and the exponential growth in computational capacity have enabled the emergence of computational models that encapsulate complex signatures of human health and disease. These developments signal a transformative shift in cardiovascular medicine and beyond (Figure 1).3 Today, many AI applications aim to augment rather than automate tasks,2,3 learning representations of the world around us by modeling the interconnection of various data types and complex biological systems. These innovations can generalize to never-before-seen tasks and generate new knowledge.2 This evolution highlights the power of AI but also exposes potential risks that arise from integrating AI into critical workflows.4
Figure 1 |. The evolving landscape of medical AI.
Rapid progress in biomedical AI has signaled a shift from feature engineering and the simple encoding of human knowledge to methods that directly augment human abilities through deep learning of complex biomedical signals and disease representations. More recently, there has been a transition from task-specific, unimodal tools to task-agnostic, often multimodal, “foundation models” that learn shared representations of distinct data types, further enabling few-shot learning for never-before-seen tasks. Concept adapted from Howell et al.3 AI: artificial intelligence.
In this article, we provide a comprehensive review of the key axes of cardiovascular practice and research where AI is expected to play a dominant role over the next decade (Central Illustration). These include the new domains of AI-driven diagnostics, digital biomarkers for risk stratification, and novel tools for evaluating care quality and prognosticating outcomes. We examine the future of care through the lens of AI-led innovation in biomedical and clinical research and explore how these innovations will accelerate achieving personalized and precise care. The review then defines the challenges that require attention and the safeguards essential to mitigating potential harm.
2. Landscape of AI innovations in cardiovascular care
2a. Innovations in disease diagnosis
Traditionally, disease diagnosis has relied on human-led interpretation of multimodal inputs in history, examination findings, imaging, and test interpretation. Given their computational simplicity, the earliest studies in AI and machine learning (ML) focused on developing tools that relied only on the structured, tabular data derived from these care encounters. Their focus was the standardized and efficient screening of cardiovascular disease under the premise that several ML algorithms capture non-linear relationships and, therefore, may be more effective than traditional linear models in fitting real-world data where complex relationships between different exposures and outcomes exist. In one example, this was realized in an algorithm for diagnosing coronary artery disease from electronic health record (EHR) datasets.5 However, head-to-head studies that have compared the performance of non-linear models developed in such tabular datasets have not found them superior to linear models developed on the same inputs.6
Instead, AI might offer the greatest value by identifying sources of phenotypic variation and risk in complex signals rather than tabular data. With advances in deep learning models, such as those based on convolutional neural networks and transformers, AI algorithms can process raw, unstructured biometric signals and images, learning new representations from these data beyond what is manually inferred and encoded. Perhaps the most studied example is the AI-guided interpretation of 12-lead electrocardiograms (ECGs). Several studies demonstrate that AI-ECG can screen for the risk of paroxysmal atrial fibrillation using ECGs in sinus rhythm,7 but can also detect structural heart disease through electrocardiographic signatures that may precede overt myocardial function changes on echocardiography (Figure 2),8 including a range of cardiomyopathies such as left ventricular systolic dysfunction (LVSD),9,10 hypertrophic cardiomyopathy (HCM),11,12 transthyretin amyloid cardiomyopathy,13,14 and even valvular heart disorders.15 These applications have been expanded to single-lead ECG acquisitions through AI-assisted stethoscopes, smartwatches, and portable devices that acquire fewer leads,16–18 facilitating easy deployment, scalability, and accessibility at the point-of-care, for both chronic (i.e., LVSD)18 and acutely life-threatening conditions such as ST-elevation myocardial infarction.19
Figure 2 |. AI-guided ECG for efficient cardiomyopathy screening.
The natural history for several forms of cardiomyopathy includes longitudinal remodeling that begins at the cellular and tissue level before resulting in changes detectable on echocardiography and eventual clinical symptoms. AI deployed to ECG signals may capture hidden labels of electrical remodeling that may precede mechanical dysfunction and clinical symptoms, thus enabling timely screening and management. AI: artificial intelligence; ECG: electrocardiography.
AI has also enabled better access to more advanced diagnostic tests that are limited by the need for expertise in acquisition and interpretation more than the equipment. For example, cardiac ultrasounds are increasingly obtainable on portable devices, but their acquisition until now has required specific training. Deep learning-enabled guidance systems provide explicit directions on the appropriate positioning to enable novices to acquire high-quality images.20 Moreover, the interpretation of these videos has been automated by a series of tools to measure and classify observable diseases.21,22 In addition, emerging tools augment the capacity of human readers, identifying individual disorders in complex disease categories,23 defining the presence and degree of valvular disease without requiring advanced testing with Doppler,24,25 and even interpreting prenatal screening ultrasounds for congenital heart disease.26
AI further enables opportunistic screening of cardiovascular disease across modalities. Several studies now show that chest X-rays can be used to detect coronary atherosclerosis,27 aortic stenosis,28 and LVSD.29 Estimation of LVSD may also be feasible from coronary angiography films, enabling assessments during emergent procedures.30 The scalability of some of these algorithms may be augmented by their integration into standard screening protocols, such as by detecting coronary calcifications on low-dose computed tomography scans for lung cancer screening31 or breast arterial calcifications on mammography.32 Finally, automated pipelines may be integrated into routine cardiac diagnostics, such as coronary computed tomography angiography, to better quantify plaque burden33 and provide insights into biological and inflammatory activity,34 thus expanding the diagnostic yield of standard reporting.
Across all these applications, scrutiny is required to ensure the diagnostic signal learned is directly linked to the label of interest rather than a confounder.35 Careful matching of cases and controls on a range of clinical and demographic factors may address measured confounders, but extensive testing across external and independent cohorts is essential to provide further reassurance about the generalizability of any given tool.
2b. Digital biomarkers of disease risk
Most AI developments for digital data streams in healthcare have focused on optimizing disease diagnosis or prognosticating outcomes. However, another key domain is the discovery of new markers for those at risk of developing disease, which previously required detailed clinical evaluation, invasive blood-based biomarkers, or risk scores. The limitation of the current approach is the need for accurate assessment of the individual risk factors and calculating the risk in a busy clinical setting. Digital biomarkers can be passive and computable using existing or easily obtainable clinical data and promise to enable risk-informed care through deployments in existing data ecosystems. The relevant data streams are already available in many individuals before the onset of clinical disease or can be easily acquired.
Commercially available wearable devices are an ideal tool for disease prediction, as they are for disease screening, given their broad potential reach among healthy adults.36 The metrics available on these devices, such as step counts, heart rate variability, and sleep quality, are strong predictors of cardiovascular health and outcomes, portending as much as a 2-fold variation in developing cardiovascular disease and premature death.37,38 Advances in deep learning are now enabling an even more comprehensive use of these data and triangulating information across sources.39
However, the selectivity among those who opt to use these devices and among those who use them consistently likely exerts a potent confounding effect.40,41 The on-device availability of spot measures such as 1-lead ECG can overcome some of these predictive challenges with time-dependent data from wearables, especially with the emerging evidence of a broad suite of novel AI-ECG risk biomarkers. These include serving as a biomarker for accelerated aging as a digital surrogate of the biological age,42,43 with individuals with ECG-defined biological age older than their actual age by 8–10 years having a 2-fold higher hazard of all-cause death than their counterparts.
There are also more specific risk markers that can be derived from ECGs with models developed to identify the cross-sectional presence of LVSD able also to identify those at 3–4-fold higher risk of developing LVSD in the future and those at risk of developing clinical heart failure.9,10 More recent assessments also suggest that these risks may have therapeutic implications. Examples include stratifying the risk of cancer therapeutics-related cardiac dysfunction by deploying AI to baseline and on-treatment ECG studies.44,45 Similarly, perioperative risk-defined AI-ECG among those undergoing non-cardiac surgery exceeds the predictive performance of established risk scores.46
The rich predictive information is not restricted to ECGs, with chest X-rays able to risk stratify patients for coronary artery disease and estimate the 10-year risk of major adverse cardiovascular events.27,47 More recently, deep learning has identified predictive cues in clinical domains where clinically relevant features were not interpretable through standard algorithms. Specifically, a new tool called the Digital Aortic Stenosis Severity Index (DASSi) was able to detect both the cross-sectional presence of severe aortic stenosis on a single cardiac ultrasound view but also identify those without aortic stenosis at risk of developing the disease and those with mild disease at risk of rapid progression.25,48 The same tool was also extensible to other cardiac video-based data formats, such as cine cardiac magnetic resonance imaging.
Overall, since our clinical practice is inherently multimodal, AI will increasingly integrate multimodal elements to refine cardiovascular risk stratification (Figure 3). This includes composites of tabular EHR data with imaging,49 genetic determinants integrated with dynamic cardiometabolic phenotypes,50 environmental parameters (“exposome”),51 including fine particulate matter data52 and images and videos extracted from the patient’s local environment,53 as well as biomarkers derived from natural language processing (NLP) and large language models (LLM) that can ingest clinical notes.54 With the rapid expansion of informatics pipelines that can process this information, universal risk calculators may eventually be replaced by locally adapted and continually validated tools that account for the unique characteristics of local populations and communities.55 Overall, from the simplest to the most complex data streams, AI-based digital biomarkers are enabling a new frontier in cardiovascular risk stratification that is more scalable and broadly usable.56
Figure 3 |. Multimodal, foundation models to encode broad representations of cardiovascular health.
Cardiovascular practice is inherently multimodal, and modern AI architectures can simulate that by learning shared embeddings of cardiovascular health by projecting and contrasting related imaging studies, EHR data, and biomedical signals of thousands of different patients. Such digital representations can fuel several downstream tasks, from timely diagnosis to more individualized estimation of treatment effectiveness. ECG: electrocardiography; EHR: electronic health records.
2c. Novel approaches to disease prognostication
AI-driven tools are also enhancing another domain – defining a person’s unique disease trajectory and risk of adverse outcomes. Developing precise risk stratification tools is essential to guide patient counseling and risk-modifying therapies but is inherently limited by the stochastic nature of clinical events and their uncertainty. Traditionally, these stratification schemes have relied on limited numbers of simplified yet interpretable, human-encoded variables,57 such as age and comorbidities. However, these tend to ignore the complex heterogeneity and interactions seen in biological systems, potentially limiting their prognostic value. The advent of complex models that can account for the time-varying inputs in the EHR and documentation in clinical encounters comes closer to realizing the full potential of common data streams in the EHR.58,59
AI enables the extension of prognostication to unstructured data, such as 12-lead ECGs,43,60,61 chest X-rays,47 and echocardiograms,62 which effectively predict cardiovascular events and all-cause mortality across myriad conditions. This is most likely driven by detecting structural or electrical abnormalities directly or indirectly related to established cardiovascular disease or markers associated with biological aging and systemic risk factors.35
The prognosticating role of AI may also extend to enabling high-value care practices and, as discussed later, individualizing how therapies may affect outcomes.63,64 For instance, despite the heterogeneous risk, primary prevention implantable cardioverter defibrillators (ICD) are recommended in a large subset of patients with heart failure. Through multimodal integration of imaging phenotypes and tabular characteristics, AI tools may provide actionable risk discrimination for arrhythmic versus non-arrhythmic mortality,65 guiding personalized risk-benefit discussions. AI models are also improving upon traditional rule-based approaches in discriminating underlying rhythm, thus preventing inappropriate shocks.66 Further insights into personalized disease trajectories may be provided by “digital twin” approaches that model the effects of varying population features and their simulated effects on outcomes.67
Emerging AI tools will also enable optimal outcomes from therapeutic interventions. In procedural cardiovascular care, for example, atrial fibrillation ablation is aided by deep learning models that integrate intra-atrial and surface signals.68 For matching patients to medications, AI phenotyping of baseline and follow-up 12-lead ECGs may assist in the safe initiation and titration of disease-modifying therapies, such as novel myosin inhibitors in hypertrophic cardiomyopathy.69 Other models can allow safe antiarrhythmic initiation to predict QTc prolongation and safe use.70
Therefore, AI’s role spans the gamut of describing the risk profile of the patient and their disease trajectory, providing actionable insights that may personalize both diagnostic and therapeutic management.
3. Future of AI in cardiovascular care delivery
3a. Locally adapted, continually learning AI tools
While the current phase of AI model development has focused on building the best tools that retain average performance across deployments, key aspects of populations and disease may enable models to perform better if they are explicitly adapted to these unique care settings.55,71 Deep learning models can be explicitly fine-tuned as they encounter more data and, therefore, can adapt to local populations. The key innovations that would enable this future are the ability to consistently define gold standard labels across individual sites and the continued ability to leverage them even after the model is deployed. This may also address population drifts over time or performance degradation in minority populations by exposing the models to populations in the real world that differ from the population where they are developed. This is particularly important in cases where the successful implementation of an AI system results in changes in its input features and target population that may impact its longitudinal performance.72
Some key safeguards are that the data sources in new settings do not reflect a bias in care that is encoded into the models, and that the training process exposes AI algorithms to the full breadth of clinical phenotypes, especially among under-represented communities (e.g. different skin colors in dermatology),73 while preventing them from making predictions through demographic confounders of disease.35 However, significant resources will be needed to steward these AI models and to evaluate and adapt models to local populations.74
3b. AI-augmented clinical encounters
AI at the point-of-care promises to accelerate and standardize tedious and time-consuming tasks, with the potential of maximizing direct patient care during clinical encounters. These now cover the full spectrum of a typical clinical encounter, from standardized examination and assessment to automated clinical documentation. Indeed, assistive AI technologies may automate the grading and characterization of cardiopulmonary auscultation,76 and even augment point-of-care inference with the addition of single-lead ECG recordings. This paradigm has been illustrated in the screening of LVSD, both in a cohort of women at risk for peripartum cardiomyopathy in Nigeria,77 and a select cohort of patients with suspected cardiomyopathy referred for echocardiography in London.17
Additionally, previously underutilized biometric signals may also provide diagnostic value, such as voice-based analytics to estimate changes in volume status,78 sensor-based gait-tracking technologies to grade frailty,79 as well as direct portable echocardiography assessment through simplified protocols,20 which maintain robust performance for the screening of valvular and structural cardiomyopathies.24,25 Concurrently, advances in NLP and LLMs have paved the path for AI solutions that can produce clinical summaries based on recordings of ambient conversations.80
3c. AI to enable high-quality community care
The evolution of remote sensing technologies has enabled a new paradigm of continuous monitoring, thus offering real-world insights into the fluctuation and variation of patient-centered information between healthcare visits.81 This may overcome the limitations of traditional in-person visits that only provide objective metrics at a given time, precluding an assessment of the holistic health and wellness of patients during daily activities. Instead, through extended monitoring of patients in their familiar settings, these tools provide data that could inform risk assessment for adverse cardiovascular outcomes. The deployment of remote monitoring now includes voice-based technologies,78 dielectric sensing tools,82 and image-based assessment of lower extremities or jugular venous pulsations,83,84 in addition to information from current patient-facing devices such as those that leverage photoplethysmography, accelerometers, and on-device 1-lead ECG,85–89 in providing a global, yet non-invasive assessment of the overall cardiovascular status.
These technologies may soon provide a non-invasive alternative to implantable monitoring90 and a long-term solution to ambulatory blood pressure or rhythm monitoring.91 Furthermore, rapid advances in material engineering may boost the usability of these algorithms by embedding reliable sensors into everyday clothes (“smart clothing”),92,93 and by generating wearables patches that can simplify the acquisition process for simple echocardiographic videos in the community (Figure 4).94 The deployment and personalization of these mobile and wearable technologies can be further enhanced through just-in-time adaptive interventions that learn to adapt their notifications and triggers to an individual’s state to maximize their effectiveness,95 and may be further boosted by representation and continual learning frameworks that will adapt them to specific use scenarios, such as in clinics or remote health settings.96
Figure 4 |. The evolving landscape of scalable and globally accessible technologies for cardiovascular disease screening and diagnosis.
To boost the scalability of novel technologies, new tools are now available to enable the high-fidelity acquisition of biometric (i.e., ECG or ultrasound) signals using smartphones and various wearable technologies. ECG: electrocardiography; POCUS: point-of-care ultrasound.
3d. Democratizing global cardiovascular care with AI
The reliance of current diagnostic cascades on advanced diagnostic testing creates stark differences in the appropriate diagnosis and treatment in low-resource settings.97 The timeliness of diagnosis, stage of disease, and the prognosis and outcomes of patients vary substantially across care settings, with the largest gaps observable in the underdeveloped and developing world.98 Even tests that are more accessible, such as ECGs and chest X-rays, require access to trained experts, who are often overworked. In the future, the focus on diagnostic cascades will likely be on risk stratification with AI tools deployed for portable devices that enable untrained individuals to do scalable community-based screening.99 Similarly, ECGs acquired as part of standard clinical care can have automated interpretation for both standard conduction and rhythm disorders,18,100 but also a range of structural heart disorders,18 further enabling risk-informed care. Similarly, handheld ultrasound yielded by minimally trained individuals will allow a clinical-grade echocardiographic evaluation right in small health centers.20 Therefore, AI may narrow the gap in access to high-quality diagnostic care, though only with dedicated effort.
4. Specific challenges to address for realizing an AI-powered future in CV care:
4a. Addressing data privacy and security
AI algorithms, especially the recent generative AI models, require access to large volumes of sensitive, multimodal patient data, the collection, processing, and use of which poses significant security and privacy risks. De-identifying the data used to train AI models is challenging, as the ‘richness’ of the data means that even anonymized patterns can be re-identified by malicious actors if they gain access to training databases.101 Additionally, the methods of data exchange or data sharing used by the various actors in the AI development pipeline are vulnerable, increasing the risk of data breaches.102 Finally, models can be exploited once released. Generative AI models can, for example, be fed prompts that force the release of personal health information that can be used for re-identification or other malicious purposes if trained on clinical data with patient identifiers.
Governments and regulators are aware of the need to mitigate these risks, with privacy and security frequently featured in national AI policy documents. China’s Interim Administrative Measures for Generative AI services, for example, requires generative AI developers to register their models and submit them for security inspection;103 the EU’s General Data Protection Regulation explicitly applies to AI systems; and the first principles of the UK’s proposed regulatory framework for AI is ‘safety, security, and robustness’.104 In the US, many groups are getting involved, including the Health AI Partnership, Coalition for Health AI, the American Medical Association, the Joint Commission (Responsible Use of Health Data), the National Academy of Medicine, and many others.
Consequently, multiple technical solutions to the privacy and security risks posed by AI are available: model-to-data techniques, such as federated learning, where the AI model is sent to local datasets where it is trained ‘in-situ’ limiting the need to rely on risky data sharing methods; synthetic data noise can be added to datasets to mitigate the risk of re-identification or inference attacks;105 and ‘white hat’ attacks such as model inversion attacks can be conducted to identify security weaknesses.106 None of these techniques are entirely immune to attacks, and each has different strengths and weaknesses; which technique is the right or best solution depends on the specific use case of the AI in question and the sensitivity of the data involved.107 Without clear solutions to mitigating data and privacy risks, these challenges risk hampering AI efforts.
4b. Working to achieve interoperability across health systems and communities at large
An essential element of any AI algorithm development is their scaling to clinical practice. However, most AI algorithms in published literature have no clear path to real-world care, and a vast majority are not even externally validated.108,109 Though financial and regulatory considerations are often barriers, the major challenge is that the algorithms are often not designed considering their interoperability across institutions. For example, it can often be challenging to deploy a model that leverages EHR data outside the parent institution due to vast variations in types of EHRs. However, this is feasible, as demonstrated through the multi-site EHR-embedded tools in the COVID-19 pandemic.110 Interoperability standards under national policy seek to address this, in part, by instituting data standards such as FHIR and HL7 and a mandate to support access to these outputs from EHRs via accessible application programming interfaces.111 The emergence of AI applications for EHR has put further pressure on these standards to be realized and instituted consistently.
Moreover, independent groups have championed common data models, such as the OMOP (Observational Medical Outcomes Partnership), that flexibly enable working across different data standards after mapping them to the same common formats, though admittedly do not yet fully capture unstructured data such as images.112 Moreover, these data models also enable federated deployment of statistical or machine learning models without sharing data across institutions.113,114 Similarly, other AI applications, including those meant for language, are more adaptable with the emergence of LLMs that can parse meaning in differently structured text.
Finally, AI models built for different cardiovascular modalities, such ECGs and imaging, are most scalable when they align with the prevailing data structure (such as ECG images vs signals),115 and aligned with standard data storage formats, such as Digital Imaging and Communications in Medicine (DICOM).116 The broad use of DICOM, a recognized data standard for medical imaging, has enabled AI for medical imaging to grow faster than other domains. Therefore, ensuring interoperable models requires the development of algorithms on interoperable data streams and would need to become a prerequisite for AI to be scalable and usable in clinical practice.
4c. Enabling appropriate regulatory controls in the era of foundation models
The development, deployment, and use of AI for health care is legally complex. First, AI designed to support diagnosis, prevention, monitoring, prognosis, or treatment should be considered a medical device and subject to regulatory control intended to ensure quality, safety, and efficacy.117 Yet, evolving medical device laws in the UK, EU, and the US all currently lack clarity - particularly with regard to evidence standards for AI – and do not cover generative AI. Consequently, the evidence base supporting AI interventions is weak. For example, in October 2023, of the 691 AI/ML-enabled medical devices approved by the FDA, just 9 (1.4%) had been tested in an interventional trial, and 96.7% had been approved via the 510(k) pathway,118 which typically does not require submission of clinical data. Second, there are questions regarding accountability and liability. It is not clear, for example, whether clinicians will be held liable in cases of missed or misdiagnosis involving a diagnostic algorithm used in a clinical setting, nor how the burden of proof would be met in such cases.119 Third, it is unknown how general, yet highly relevant, regulations relating to data protection and anti-discrimination will be adapted for AI.120 In the EU and the UK, the legality of medical data processing depends on the context and intended purpose: using data for research requires explicit patient consent, whereas using data for direct care does not. AI blends these different use cases by, for example, providing diagnostic support for direct care and continuing to learn (‘research’),121 and its black-box nature makes obtaining specific and unambiguous consent difficult. Furthermore, EU and US anti-discrimination law is typically technology agnostic but only covers specific areas of activity, such as employment and education, rather than important facets, like bias among algorithms used in healthcare.120 Finally, generative AI introduces a range of novel safety concerns related to, for example, its tendency to hallucinate false information that no existing laws are designed to cover.117 There are developments underway to overcome some of these regulatory limitations. The US Food and Drug Administration (FDA) has, for example, published an AI/ML-enabled medical device action plan,122 but the pace of regulatory change does not match the pace of the technology.
Moreover, given the resources required to train, validate, and test AI tools in specific settings, we anticipate significant ‘off-label’ use of AI in healthcare, that is, unapproved use of an approved model.123 This paradigm is expected to become even more common in the near future with the arrival of task-agnostic foundation models. Since most existing regulatory frameworks require a defined intended use, the feasibility of regulating clinical foundation models likely requires the development of novel regulatory pathways to cover the intended and unintended uses of such models.
5. Landscape of AI innovations in cardiovascular discovery
5a. AI for biological discoveries: mechanistic inference and drug design
AI-driven tools can significantly expedite the drug discovery process, from the initial screening of compound libraries to guided drug discovery to AI agents that can conduct drug screening (Figure 5). In the context of cardiovascular diseases, AI-guided drug discovery offers unprecedented opportunities to identify novel therapeutic targets, predict drug efficacy, and optimize the design of lead compounds.124
Figure 5: The Future of Evidence Generation and Translation.
Deep learning-enabled multiomic platforms and models enable more granular phenotyping of the interaction between a person’s genome, broader phenome, and exposome. Computational models can now learn these complex representations and may boost the development of more individualized, effective, and safe treatments. AI: artificial intelligence.
AlphaFold, developed by DeepMind, has revolutionized protein structure prediction, which is critical in understanding drug-target interactions. AlphaFold and subsequent models like AlphaFold2 and ESMFold can predict protein structures, enabling a detailed map of the protein landscape, which in turn facilitates the identification of drug-binding sites and the design of more efficacious therapeutic molecules.125,126 Integrating AlphaFold’s capabilities with drug discovery platforms has already led to identifying novel small molecule inhibitors for targets like CDK20, a kinase implicated in various biological processes, including those relevant to cardiovascular diseases.127
Beyond predicting the structure of proteins, generative AI models are now being employed to design new proteins and peptides as novel therapies.128 These models have started transforming the approach from a traditionally reactive process to a proactive one. By incorporating principles from diffusion models used in synthetic image generation, researchers have expanded the scope of generative AI to protein design. This involves developing models to iterate on representing the protein’s structure until a desired configuration is achieved, mirroring the process of natural protein folding but directed towards a specific functional goal.129 Furthermore, the design of targeted drugs may be further optimized through Bayesian platforms that can further incorporate prior knowledge to accelerate drug discovery and clinical application.130 Thus, AI has the potential to accelerate the pace of drug discovery, offering a wealth of opportunities for cardiovascular medicine. From AI-driven biological insights to the imaginative design of biological therapies, the future of drug discovery is poised for a significant leap forward.
5b. AI for precision therapeutics: the expanding role of large-scale genomics
With the decreasing cost and improved availability of sequencing technologies, the era of ‘petabyte-scale’ genomics is upon us.131 In cardiovascular care, this means optimizing treatment regimens for patients based on their unique genetic makeup that influences drug metabolism, efficacy, and safety. Even without AI, genomics has begun to play a role in routine clinical care. For instance, pharmacogenomics has been incorporated into cardiovascular medicine, influencing therapeutic strategies for hypercholesterolemia, acute coronary syndrome, and cardiomyopathy.132,133 Mutations in genes like TTR, linked to hereditary cardiac amyloidosis, can guide the use of specific treatments only beneficial to patients with certain mutations, underscoring the need for genetic testing in clinical workflows.134 However, with the advent of multimodal AI, the opportunities to transform care and optimize therapeutics using genomics are now numerous.
First, population-scale genomics can pave the way for simulating responses to treatments using high-fidelity digital twins that incorporate genomics and response to treatments noted in historical data. This approach could significantly enhance precision medicine with the advent of genomically-guided individualized care.135 Polygenic risk scores, as well as their genomic inputs, which capture inherent risk, can now be used as elements within multi-modal risk prediction models and as influencers of therapeutic response.136 The ability to leverage genomic data at scale has required ML to be computationally mature and models to be flexible enough to allow genotypic and phenotypic data to be modeled simultaneously. Finally, current cardiovascular foundation models only utilize clinical data, but incorporating genomic data could lead to an understanding of different axes and scales of biology.
5c. AI for clinical discovery: personalized inference from clinical trials
Beyond the role of AI in biological discovery and optimizing clinical operations, it can also enhance learning precision care from large-scale experiments in the clinical domain in the form of randomized clinical trials (RCTs). Whereas the traditional interpretation of an RCT relies on estimating an average treatment effect across the range of included individuals, conceptually, a patient’s characteristics may modify the efficacy of an intervention.137 Since in most RCTs, individuals are only assigned to one arm, the counterfactual scenario of the same patient receiving a treatment and a control is not knowable.
Thus, an unmet need in our broader quest towards precision medicine is to develop data-driven ways to personalize our estimation of the expected harm and benefit of various interventions. Statistical ML approaches provide us with a set of rules that can estimate counterfactual observations, thus providing a robust way to assess treatment heterogeneity independent of human biases. In one example, computational embeddings (“phenomaps”) of clinical trial populations can be constructed to simulate a universe of potential patient phenotypes, as represented in the original clinical trials,63,138,139 which may flag higher-order interactions not reflected by univariate analyses. Using these phenomaps, in silico simulations, and iterative analyses, one can then estimate a personalized treatment effect estimate for a new individual, further incorporating metrics of representativeness and uncertainty.140 Alternative approaches also adapt ML algorithms, such as causal forests, allowing modeling interactions between treatment assignment and baseline phenotypes.141 There have already been demonstrations of their role in identifying treatment effect heterogeneity, such as in patient selection for intensive blood pressure lowering,63,64 and the appropriate use of cardiovascular risk-reduction therapies for diabetes mellitus,139 among others. Collectively, these techniques can guide the translation of RCT findings into real-world populations but also facilitate next-phase transitions in RCT design, for example, defining appropriate phase III inclusion criteria using phase II inference.
Beyond data collected from RCTs, causal ML approaches can be applied to observational data to estimate heterogeneous treatment effects.142 While such approaches only apply under strict assumptions, they could leverage the large variability already in clinical practice. These estimates from data already available could then be used to inform the design of prospective RCTs,143 or enable the generation of evidence where RCTs may not be feasible due to time, cost, or safety-related considerations.
6. Future cardiovascular optimization
6a. Highly individualized targeted therapeutic selection and design
The advent of multiomics, encompassing genomics, proteomics, metabolomics, and other omics technologies, has been integral to the evolution of cardiovascular risk assessment and outcome prediction. AI can leverage these multi-dimensional datasets to uncover complex biomolecular interactions and pathways contributing to disease pathogenesis, prognosis, and response to therapy. A key application of AI in multiomics is the identification of biomarkers for cardiovascular risk, integrating multiomic data to delineate patient subpopulations with unique molecular signatures, paving the way for personalized medicine. These biomarkers can predict cardiovascular events, suggest individual responses to specific treatments, and define novel therapeutic targets.144,145 Such AI-driven platforms not only identify novel drug targets but also provide a foundation for repositioning existing drugs to treat specific patient endotypes, potentially transforming the treatment landscape for cardiovascular and related metabolic conditions.146 AI’s ability to distill multi-layered omics data into coherent patterns heralds a new era of predictive models that offer greater specificity and sensitivity for cardiovascular risk stratification and therapeutic intervention. This approach not only facilitates early intervention strategies but also refines our understanding of disease progression and therapeutic response at a granular level.
Integrating genomics, proteomics, and metabolomics with AI promises to reshape personalized drug design in cardiovascular care.146 This would entail evaluating known treatments and potentially repurposed therapies and defining a need for custom therapeutic agents. For this, foundation models can be built linking these multiomics data streams with clinical outcomes. With these multiomic foundation models, personalized drug design hopefully becomes a reality.
Moreover, the implementation of such models facilitates the exploration of complex biomolecular networks that underlie cardiovascular conditions, identifying potential new drug targets that are specifically tailored to the nuanced needs of individual patients. This leads to a more efficient drug development process, reducing the time and cost typically associated with bringing new cardiovascular drugs to market.
6b. Acceleration of evidence translation through AI-powered clinical trials
A major barrier to translating discoveries into practice is the pace of conducting large-scale clinical trials to confirm their effectiveness and safety, with the typical development time for an innovative drug estimated at over 9 years.147 Moreover, these trials typically provide evidence of benefit for 10 or fewer individuals per 100 participants treated, leaving the vast majority uncertain whether the benefit will be actualized. Acknowledging the need to improve the speed, efficiency, and representation of RCTs, AI tools are being developed to address key bottlenecks of this process.
First, AI is automating the eligibility screening process through ML- and NLP-based approaches that automate the mapping of patient characteristics to inclusion and exclusion criteria (i.e., Trial Pathfinder, Criteria2Query), allowing efficient screening of large repositories and EHRs.148 Second, with multiple medical therapies competing for the same patient population, AI-guided platform trials may provide a data-driven inference mechanism to guide the allocation and matching of participants to individual strategies based on a priori knowledge regarding the interaction between their disease phenotype and anticipated treatment effects.149 Third, advances in LLMs may streamline operations, accelerating and standardizing time-consuming and error-prone processes such as clinical event adjudication.150,151
Fourth, AI may propel innovation in adaptive clinical trials and data-driven predictive enrichment, an important unmet need, as suggested by the FDA.152 For instance, data-driven ML approaches may be incorporated into RCT protocols to screen for treatment effect heterogeneity based on accumulating signals of benefit and harm. In the context of an adaptive design, this paradigm could maximize RCT efficiency by guiding data-driven predictive enrichment in a timely fashion.153
Finally, digital twin technologies that simulate personalized health and disease conditions and projected trajectories by incorporating data with clinical knowledge may provide efficient alternatives to the somewhat limited traditional trial emulation methods from observational data.67,154 These examples showcase how AI may soon provide end-to-end acceleration of all key steps of RCT design, enrollment, follow-up, and interpretation.
7. Challenges to address to accelerate the future of AI-powered CV discovery
7a. How to evolve evidence-generation and regulation to evaluate personalization
The investment in precision care solutions requires a corresponding investment in strategies to test this personalization of care effectively. This is inherently challenging as these strategies are specific to the population they are tested in. By design, regardless of the strategy to define the precision care solution (genomic, metabolomic, and/or phenomic), the characteristics of the population will determine the range of phenotypes and the possible response to therapies. Therefore, prospective clinical studies and RCTs that deploy a personalization strategy in candidate individuals will require both broad coverage of phenotypes and a commitment to being evaluated in multiple distinct populations and care settings. This also includes defining appropriate continual learning mechanisms, particularly for sensor-derived data, that would enable their robust performance despite unavoidable data drifts. Moreover, since a majority of the data on these novel personalization strategies are derived from selective populations,155 the omics profiles of key demographic groups vary widely,156 and most of these studies are based on observational assessments,157 rigorous prospective evaluation with a focus on clinically relevant outcomes is essential in different population groups. Finally, AI may also be used to confirm the validity of published data and information, thus providing an additional layer of scrutiny to evidence that may impact clinical practice.158
However, given the resource intensiveness of conducting large-scale trials across care settings, especially with the increasing volume of algorithmic care, there will need to be a focus on nimble, pragmatic trials that leverage AI to enhance the efficiency of the process. Ultimately, the approach to personalization, the populations at risk, and how it affects their outcomes are critical to defining their range of use and enabling their eventual translation to clinical care. These decisions will also define the confidence in these precision care solutions and enable a degree of reliability critical for the regulatory pathways.
7b. How do we address health equity, bias, and barriers to implementation
While the central premise of integrating AI in clinical care is to make care delivery simpler, safer, more accessible, and of better quality at lower cost, there are major barriers to realizing this vision. A key barrier is that the development and deployment of technologies aimed at improving cardiovascular health still rely on many data streams only obtained in a subset of the population.40,41,159 Specifically, the more affluent have better access to wearable devices,39 and only those with access to regular care undergo diagnostic testing over which AI will be deployed.160 Moreover, since AI-driven care enhancements are currently largely seen as optional tools that a practice may choose to take up but are not mandated or often even covered by insurance,161 those that cater to a more resource-rich population are likely to bring them forward. A big frontier for AI-driven care innovation is to reach people where they live, using technology deployed in a way that they receive the benefits and with care processes aligned with their goals. This has to achieve balance with the business models that support technology diffusion, especially as AI-powered technology becomes an integral part of care. While ensuring broad access to high-quality care, there will also be a need to build trust in communities that have historically mistrusted medical innovations both due to systemic malpractices against their communities as well as challenges with health and technology literacy.162
Similarly, even in health settings where novel methods will define precision care solutions, combining information drawn from clinical trials, genomics, or multiomics, such an approach could exacerbate disparities by being largely accessed by those with additional resources or seeking care at select institutions that offer them. Therefore, before their selective use becomes the norm, clinical guidelines, payers, and health systems must coalesce around the appropriate standards to enable their broad and equitable use.
8. Conclusions
Over the next decade, AI technologies will become a core component of the diagnostic, prognostic, and therapeutic cardiovascular toolkit. Unimodal and task-specific models will likely be superseded by task-agnostic, multimodal, and semi-autonomous systems, effectively augmenting rather than replacing human intelligence. Innovations in data acquisition through wearables and portable technologies are expected to further broaden the scope of AI solutions focused on cardiovascular health monitoring and screening. Concurrently, AI-driven, integrated multiomic environments will enable deeper phenotyping, redefine disease classifications, guide biomarker discovery, and de-risk the development of novel therapeutics. Meanwhile, as the speed of innovation continues to outpace the reflexes of the regulatory environment, the need for ethical, equitable, and trustworthy AI will need to be embedded into the development and validation processes, and the scope of AI should expand further to include its optimal implementation within real-world, dynamic systems.
KEY POINTS.
Artificial intelligence (AI)-enabled technologies are increasingly integrated into cardiovascular practice and investigation.
Over the next decade, we envision an AI-propelled future in which the cardiovascular diagnostic and therapeutic landscape will effectively leverage multimodal data at the point of care.
Innovations in biomedical discovery and cardiovascular research are also set to make the future of cardiovascular care more personalized, precise, and effective.
The path to this future requires equitable and regulated adoption that prioritizes fairness, equity, safety, and partnerships with innovators as well as our communities and society.
Funding:
National Heart, Lung, and Blood Institute of the National Institutes of Health (under awards R01HL167858 and K23HL153775 to R.K., F32HL170592 to E.K.O., R01HL155915 and R01HL167050 to G.N.N., R01HL158626 to J.W., and UM1 TR004407 to E.J.T.)
Disclosures:
R.K. is an Associate Editor of JAMA and receives research support, through Yale, from the Blavatnik Foundation, Bristol-Myers Squibb, Novo Nordisk, and BridgeBio. He is a coinventor of U.S. Provisional Patent Applications 63/177,117, 63/428,569, 63/346,610, 63/484,426, 63/508,315, 63/580,137, 63/606,203, 63/562,335, and a co-founder of Ensight-AI, Inc and Evidence2Health, LLC. E.K.O. is an academic co-founder of Evidence2Health LLC, and has been a consultant for Caristo Diagnostics, Ltd and Ensight-AI, Inc. He is a co-inventor in patent applications (US17/720,068, 63/619,241, 63/177,117, 63/580,137, 63/606,203, 63/562,335, WO2018078395A1, WO2020058713A1) and has received royalty fees from technology licensed through the University of Oxford. G.N.N. is an academic co-founder of Renalytix, Pensieve, Data2Wisdom. G.N.N. also has patents licensed to Heart Test Laboratories. G.N.N. acts as a consultant to Renalytix, Pensieve and Heart Test Laboratories. A.B. is a co-founder and consultant to Personalis and NuMedii; consultant or advisor to NIH, JAMA, Mango Tree Corporation, Samsung, Geisinger Health, Washington University in Saint Louis, University of Utah, and in the recent past, 10x Genomics, Helix, Pathway Genomics, and Verinata (Illumina); has served on paid advisory panels or boards for Regenstrief Institute, Gerson Lehman Group, AlphaSights, Covance, Novartis, Genentech, and Merck, and Roche; is a shareholder in Personalis and NuMedii; is a minor shareholder in Apple, Meta (Facebook), Alphabet (Google), Microsoft, Amazon, NVIDIA, AMD, Snap, 10x Genomics, Doximity, Regeneron, Sanofi, Pfizer, Royalty Pharma, Moderna, BioNtech, Invitae, Pacific Biosciences, Editas Medicine, Eli Lilly, Nuna Health, Assay Depot (Scientist.com), Vet24seven, Snowflake, Sophia Genetics, and several other non-health related companies and mutual funds; and has received honoraria and travel reimbursement for invited talks from Johnson and Johnson, Roche, Genentech, Pfizer, Merck, Lilly, Takeda, Varian, Mars, Siemens, Optum, Abbott, Celgene, AstraZeneca, AbbVie, Westat, Applied Research Works, Acentrus, ALDA, and many academic institutions, medical or disease specific foundations and associations, and health systems. A.B. receives royalty payments through Stanford University, for several patents and other disclosures licensed to NuMedii and Personalis. A.B.’s research has been funded by NIH, FDA, Peraton (as the prime on an NIH contract), Priscilla Chan and Mark Zuckerberg, the Barbara and Gerson Bakar Foundation, Genentech, Johnson and Johnson, Chan Zuckerberg Science, Robert Wood Johnson Foundation, Leon Lowenstein Foundation, Intervalien Foundation, and in the past, the March of Dimes, Juvenile Diabetes Research Foundation, California Governor’s Office of Planning and Research, California Institute for Regenerative Medicine, L’Oreal, and Progenity. None of these entities had any role in the design, planning, or execution of the study, or interpretation of the findings. E.J.T. is on the scientific advisory board of Tempus, Abridge, and Pheno.ai. The remaining authors have nothing to disclose.
ABBREVIATIONS LIST
- AI
artificial intelligence
- ECG
electrocardiography
- EHR
electronic health record
- FDA
Food and Drug Administration
- HCM
hypertrophic cardiomyopathy
- LLM
large language model
- LVSD
left ventricular systolic dysfunction
- ML
machine learning
- NLP
natural language processing
- RCT
randomized clinical trials
REFERENCES
- 1.Topol EJ. High-performance medicine: the convergence of human and artificial intelligence. Nat Med. 2019;25(1):44–56. [DOI] [PubMed] [Google Scholar]
- 2.Moor M, Banerjee O, Abad ZSH, et al. Foundation models for generalist medical artificial intelligence. Nature. 2023;616(7956):259–265. [DOI] [PubMed] [Google Scholar]
- 3.Howell MD, Corrado GS, DeSalvo KB. Three Epochs of Artificial Intelligence in Health Care. JAMA. 2024;331(3):242–244. [DOI] [PubMed] [Google Scholar]
- 4.Fogo AB, Kronbichler A, Bajema IM. AI’s Threat to the Medical Profession. JAMA. 2024;331(6):471–472. [DOI] [PubMed] [Google Scholar]
- 5.Forrest IS, Petrazzini BO, Duffy Á, et al. Machine learning-based marker for coronary artery disease: derivation and validation in two longitudinal cohorts. Lancet. 2023;401(10372):215–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Khera R, Haimovich J, Hurley NC, et al. Use of Machine Learning Models to Predict Death After Acute Myocardial Infarction. JAMA Cardiol. 2021;6(6):633–641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Attia ZI, Noseworthy PA, Lopez-Jimenez F, 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(10201):861–867. [DOI] [PubMed] [Google Scholar]
- 8.Ulloa-Cerna AE, Jing L, Pfeifer JM, et al. rECHOmmend: An ECG-Based Machine Learning Approach for Identifying Patients at Increased Risk of Undiagnosed Structural Heart Disease Detectable by Echocardiography. Circulation. 2022;146(1):36–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Attia ZI, Kapa S, Lopez-Jimenez F, et al. Screening for cardiac contractile dysfunction using an artificial intelligence-enabled electrocardiogram. Nat Med. 2019;25(1):70–74. [DOI] [PubMed] [Google Scholar]
- 10.Sangha V, Nargesi AA, Dhingra LS, et al. Detection of Left Ventricular Systolic Dysfunction From Electrocardiographic Images. Circulation. 2023;148(9):765–777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Goto S, Solanki D, John JE, et al. Multinational Federated Learning Approach to Train ECG and Echocardiogram Models for Hypertrophic Cardiomyopathy Detection. Circulation. 2022;146(10):755–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Sangha V, Dhingra LS, Oikonomou E, et al. Identification of Hypertrophic Cardiomyopathy on Electrocardiographic Images with Deep Learning. medRxiv. Published online December 28, 2023:2023.12.23.23300490. [Google Scholar]
- 13.Goto S, Mahara K, Beussink-Nelson L, et al. Artificial intelligence-enabled fully automated detection of cardiac amyloidosis using electrocardiograms and echocardiograms. Nat Commun. 2021;12(1):2726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Grogan M, Lopez-Jimenez F, Cohen-Shelly M, et al. Artificial Intelligence-Enhanced Electrocardiogram for the Early Detection of Cardiac Amyloidosis. Mayo Clin Proc. 2021;96(11):2768–2778. [DOI] [PubMed] [Google Scholar]
- 15.Cohen-Shelly M, Attia ZI, Friedman PA, et al. Electrocardiogram screening for aortic valve stenosis using artificial intelligence. Eur Heart J. 2021;42(30):2885–2896. [DOI] [PubMed] [Google Scholar]
- 16.Attia ZI, Harmon DM, Dugan J, et al. Prospective evaluation of smartwatch-enabled detection of left ventricular dysfunction. Nat Med. 2022;28(12):2497–2503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bachtiger P, Petri CF, Scott FE, et al. Point-of-care screening for heart failure with reduced ejection fraction using artificial intelligence during ECG-enabled stethoscope examination in London, UK: a prospective, observational, multicentre study. Lancet Digit Health. 2022;4(2):e117–e125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Khunte A, Sangha V, Oikonomou EK, et al. Detection of left ventricular systolic dysfunction from single-lead electrocardiography adapted for portable and wearable devices. NPJ Digit Med. 2023;6(1):124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gibson CM, Mehta S, Ceschim MRS, et al. Evolution of single-lead ECG for STEMI detection using a deep learning approach. Int J Cardiol. 2022;346:47–52. [DOI] [PubMed] [Google Scholar]
- 20.Narang A, Bae R, Hong H, et al. Utility of a Deep-Learning Algorithm to Guide Novices to Acquire Echocardiograms for Limited Diagnostic Use. JAMA Cardiol. 2021;6(6):624–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ouyang D, He B, Ghorbani A, et al. Video-based AI for beat-to-beat assessment of cardiac function. Nature. 2020;580(7802):252–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Krishna H, Desai K, Slostad B, et al. Fully Automated Artificial Intelligence Assessment of Aortic Stenosis by Echocardiography. J Am Soc Echocardiogr. 2023;36(7):769–777. [DOI] [PubMed] [Google Scholar]
- 23.Duffy G, Cheng PP, Yuan N, et al. High-Throughput Precision Phenotyping of Left Ventricular Hypertrophy With Cardiovascular Deep Learning. JAMA Cardiol. 2022;7(4):386–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Oikonomou EK, Holste G, Coppi A, et al. Artificial intelligence-guided detection of under-recognized cardiomyopathies on point-of-care cardiac ultrasound. medRxiv. Published online March 15, 2024:2024.03.10.24304044. [Google Scholar]
- 25.Holste G, Oikonomou EK, Mortazavi BJ, et al. Severe aortic stenosis detection by deep learning applied to echocardiography. Eur Heart J. 2023;44(43):4592–4604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Arnaout R, Curran L, Zhao Y, Levine JC, Chinn E, Moon-Grady AJ. An ensemble of neural networks provides expert-level prenatal detection of complex congenital heart disease. Nat Med. 2021;27(5):882–891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kamel PI, Yi PH, Sair HI, Lin CT. Prediction of Coronary Artery Calcium and Cardiovascular Risk on Chest Radiographs Using Deep Learning. Radiol Cardiothorac Imaging. 2021;3(3):e200486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ueda D, Yamamoto A, Ehara S, et al. Artificial intelligence-based detection of aortic stenosis from chest radiographs. Eur Heart J Digit Health. 2022;3(1):20–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ueda D, Matsumoto T, Ehara S, et al. Artificial intelligence-based model to classify cardiac functions from chest radiographs: a multi-institutional, retrospective model development and validation study. Lancet Digit Health. 2023;5(8):e525–e533. [DOI] [PubMed] [Google Scholar]
- 30.Avram R, Barrios JP, Abreau S, et al. Automated Assessment of Cardiac Systolic Function From Coronary Angiograms With Video-Based Artificial Intelligence Algorithms. JAMA Cardiol. 2023;8(6):586–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chao H, Shan H, Homayounieh F, et al. Deep learning predicts cardiovascular disease risks from lung cancer screening low dose computed tomography. Nat Commun. 2021;12(1):2963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Iribarren C, Chandra M, Lee C, et al. Breast Arterial Calcification: a Novel Cardiovascular Risk Enhancer Among Postmenopausal Women. Circ Cardiovasc Imaging. 2022;15(3):e013526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.de Vos BD, Lessmann N, de Jong PA, Išgum I. Deep Learning-Quantified Calcium Scores for Automatic Cardiovascular Mortality Prediction at Lung Screening Low-Dose CT. Radiol Cardiothorac Imaging. 2021;3(2):e190219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kotanidis CP, Xie C, Alexander D, et al. Constructing custom-made radiotranscriptomic signatures of vascular inflammation from routine CT angiograms: a prospective outcomes validation study in COVID-19. Lancet Digit Health. 2022;4(10):e705–e716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Duffy G, Clarke SL, Christensen M, et al. Confounders mediate AI prediction of demographics in medical imaging. NPJ Digit Med. 2022;5(1):188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Spatz ES, Ginsburg GS, Rumsfeld JS, Turakhia MP. Wearable Digital Health Technologies for Monitoring in Cardiovascular Medicine. N Engl J Med. 2024;390(4):346–356. [DOI] [PubMed] [Google Scholar]
- 37.Master H, Annis J, Huang S, et al. Association of step counts over time with the risk of chronic disease in the All of Us Research Program. Nat Med. 2022;28(11):2301–2308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Stamatakis E, Ahmadi MN, Gill JMR, et al. Association of wearable device-measured vigorous intermittent lifestyle physical activity with mortality. Nat Med. 2022;28(12):2521–2529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Torres-Soto J, Ashley EA. Multi-task deep learning for cardiac rhythm detection in wearable devices. NPJ Digit Med. 2020;3:116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Dhingra LS, Aminorroaya A, Oikonomou EK, et al. Use of Wearable Devices in Individuals With or at Risk for Cardiovascular Disease in the US, 2019 to 2020. JAMA Netw Open. 2023;6(6):e2316634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Aminorroaya A, Dhingra LS, Nargesi AA, Oikonomou EK, Krumholz HM, Khera R. Use of Smart Devices to Track Cardiovascular Health Goals in the United States. JACC Adv. 2023;2(7). doi: 10.1016/j.jacadv.2023.100544 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Shah M, de A Inácio MH, Lu C, et al. Environmental and genetic predictors of human cardiovascular ageing. Nat Commun. 2023;14(1):4941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Lima EM, Ribeiro AH, Paixão GMM, et al. Deep neural network-estimated electrocardiographic age as a mortality predictor. Nat Commun. 2021;12(1):5117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Oikonomou EK, Sangha V, Dhingra LS, et al. Artificial intelligence-enhanced risk stratification of cancer therapeutics-related cardiac dysfunction using electrocardiographic images. medRxiv. Published online March 19, 2024. doi: 10.1101/2024.03.12.24304047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yagi R, Goto S, Himeno Y, et al. Artificial intelligence-enabled prediction of chemotherapy-induced cardiotoxicity from baseline electrocardiograms. Nat Commun. 2024;15(1):2536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ouyang D, Theurer J, Stein NR, et al. Electrocardiographic deep learning for predicting post-procedural mortality: a model development and validation study. Lancet Digit Health. 2024;6(1):e70–e78. [DOI] [PubMed] [Google Scholar]
- 47.Weiss J, Raghu VK, Paruchuri K, et al. Deep Learning to Estimate Cardiovascular Risk From Chest Radiographs : A Risk Prediction Study. Ann Intern Med. 2024;177(4):409–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Oikonomou EK, Holste G, Yuan N, et al. A Multimodal Video-Based AI Biomarker for Aortic Stenosis Development and Progression. JAMA Cardiol. Published online April 6, 2024. doi: 10.1001/jamacardio.2024.0595 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jabbour S, Fouhey D, Kazerooni E, Wiens J, Sjoding MW. Combining chest X-rays and electronic health record (EHR) data using machine learning to diagnose acute respiratory failure. J Am Med Inform Assoc. 2022;29(6):1060–1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Urbut SM, Cho SMJ, Paruchuri K, et al. Dynamic Importance of Genomic and Clinical Risk for Coronary Artery Disease Over the Life Course. medRxiv. Published online November 4, 2023. doi: 10.1101/2023.11.03.23298055 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Khera R Artificial intelligence-enhanced exposomics: novel insights into cardiovascular health. Eur Heart J. Published online March 28, 2024. doi: 10.1093/eurheartj/ehae159 [DOI] [PubMed] [Google Scholar]
- 52.Newman JD, Bhatt DL, Rajagopalan S, et al. Cardiopulmonary Impact of Particulate Air Pollution in High-Risk Populations: JACC State-of-the-Art Review. J Am Coll Cardiol. 2020;76(24):2878–2894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Chen Z, Dazard JE, Khalifa Y, Motairek I, Al-Kindi S, Rajagopalan S. Artificial intelligence-based assessment of built environment from Google Street View and coronary artery disease prevalence. Eur Heart J. Published online March 28, 2024. doi: 10.1093/eurheartj/ehae158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Abbasian M, Yang Z, Khatibi E, et al. Knowledge-Infused LLM-Powered Conversational Health Agent: A Case Study for Diabetes Patients. arXiv [csCL]. Published online February 15, 2024. http://arxiv.org/abs/2402.10153 [DOI] [PubMed] [Google Scholar]
- 55.Youssef A, Pencina M, Thakur A, Zhu T, Clifton D, Shah NH. External validation of AI models in health should be replaced with recurring local validation. Nat Med. 2023;29(11):2686–2687. [DOI] [PubMed] [Google Scholar]
- 56.Oikonomou EK, Khera R. Leveraging the Full Potential of Wearable Devices in Cardiomyopathies. J Card Fail. Published online March 5, 2024. doi: 10.1016/j.cardfail.2024.02.011 [DOI] [PubMed] [Google Scholar]
- 57.Yadlowsky S, Hayward RA, Sussman JB, McClelland RL, Min YI, Basu S. Clinical Implications of Revised Pooled Cohort Equations for Estimating Atherosclerotic Cardiovascular Disease Risk. Ann Intern Med. 2018;169(1):20–29. [DOI] [PubMed] [Google Scholar]
- 58.Yang X, Chen A, PourNejatian N, et al. A large language model for electronic health records. NPJ Digit Med. 2022;5(1):194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Cao K, Xia Y, Yao J, et al. Large-scale pancreatic cancer detection via non-contrast CT and deep learning. Nat Med. 2023;29(12):3033–3043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Attia ZI, Friedman PA, Noseworthy PA, et al. Age and Sex Estimation Using Artificial Intelligence From Standard 12-Lead ECGs. Circ Arrhythm Electrophysiol. 2019;12(9):e007284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hughes JW, Tooley J, Torres Soto J, et al. A deep learning-based electrocardiogram risk score for long term cardiovascular death and disease. NPJ Digit Med. 2023;6(1):169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ulloa Cerna AE, Jing L, Good CW, et al. Deep-learning-assisted analysis of echocardiographic videos improves predictions of all-cause mortality. Nat Biomed Eng. 2021;5(6):546–554. [DOI] [PubMed] [Google Scholar]
- 63.Oikonomou EK, Spatz ES, Suchard MA, Khera R. Individualising intensive systolic blood pressure reduction in hypertension using computational trial phenomaps and machine learning: a post-hoc analysis of randomised clinical trials. Lancet Digit Health. 2022;4(11):e796–e805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Inoue K, Athey S, Tsugawa Y. Machine-learning-based high-benefit approach versus conventional high-risk approach in blood pressure management. Int J Epidemiol. 2023;52(4):1243–1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kolk MZH, Ruipérez-Campillo S, Deb B, 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. Europace. 2023;25(9). doi: 10.1093/europace/euad271 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tateishi R, Suzuki M, Shimizu M, et al. Risk prediction of inappropriate implantable cardioverter-defibrillator therapy using machine learning. Sci Rep. 2023;13(1):19586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Laubenbacher R, Mehrad B, Shmulevich I, Trayanova N. Digital twins in medicine. Nat Comput Sci. 2024;4(3):184–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Tang S, Razeghi O, Kapoor R, et al. Machine Learning-Enabled Multimodal Fusion of Intra-Atrial and Body Surface Signals in Prediction of Atrial Fibrillation Ablation Outcomes. Circ Arrhythm Electrophysiol. 2022;15(8):e010850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tison Geoffrey H, Siontis Konstantinos C, Abreau Sean, et al. Assessment of Disease Status and Treatment Response With Artificial Intelligence–Enhanced Electrocardiography in Obstructive Hypertrophic Cardiomyopathy. J Am Coll Cardiol. 2022;79(10):1032–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Maille B, Wilkin M, Million M, et al. Smartwatch Electrocardiogram and Artificial Intelligence for Assessing Cardiac-Rhythm Safety of Drug Therapy in the COVID-19 Pandemic. The QT-logs study. Int J Cardiol. 2021;331:333–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Fournier-Tombs E Local transplantation, adaptation, and creation of AI models for public health policy. Front Artif Intell. 2023;6:1085671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Vaid A, Sawant A, Suarez-Farinas M, et al. Implications of the Use of Artificial Intelligence Predictive Models in Health Care Settings : A Simulation Study. Ann Intern Med. 2023;176(10):1358–1369. [DOI] [PubMed] [Google Scholar]
- 73.Daneshjou R, Vodrahalli K, Novoa RA, et al. Disparities in dermatology AI performance on a diverse, curated clinical image set. Sci Adv. 2022;8(32):eabq6147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Eaneff S, Obermeyer Z, Butte AJ. The Case for Algorithmic Stewardship for Artificial Intelligence and Machine Learning Technologies. JAMA. 2020;324(14):1397–1398. [DOI] [PubMed] [Google Scholar]
- 75.Tonekaboni S, Joshi S, Campbell K, Duvenaud D, Goldenberg A. What went wrong and when? Instance-wise feature importance for time-series black-box models. Adv Neural Inf Process Syst. 2020;33. [Google Scholar]
- 76.Chorba JS, Shapiro AM, Le L, et al. Deep Learning Algorithm for Automated Cardiac Murmur Detection via a Digital Stethoscope Platform. J Am Heart Assoc. 2021;10(9):e019905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Adedinsewo DA, Morales-Lara AC, Dugan J, et al. Screening for peripartum cardiomyopathies using artificial intelligence in Nigeria (SPEC-AI Nigeria): Clinical trial rationale and design. Am Heart J. 2023;261:64–74. [DOI] [PubMed] [Google Scholar]
- 78.Amir O, Abraham WT, Azzam ZS, et al. Remote Speech Analysis in the Evaluation of Hospitalized Patients With Acute Decompensated Heart Failure. JACC Heart Fail. 2022;10(1):41–49. [DOI] [PubMed] [Google Scholar]
- 79.Fan S, Ye J, Xu Q, et al. Digital health technology combining wearable gait sensors and machine learning improve the accuracy in prediction of frailty. Front Public Health. 2023;11:1169083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Haberle T, Cleveland C, Snow GL, et al. The impact of nuance DAX ambient listening AI documentation: a cohort study. J Am Med Inform Assoc. 2024;31(4):975–979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Stevenson LW, Ross HJ, Rathman LD, Boehmer JP. Remote Monitoring for Heart Failure Management at Home. J Am Coll Cardiol. 2023;81(23):2272–2291. [DOI] [PubMed] [Google Scholar]
- 82.Amir O, Azzam ZS, Gaspar T, et al. Validation of remote dielectric sensing (ReDSTM) technology for quantification of lung fluid status: Comparison to high resolution chest computed tomography in patients with and without acute heart failure. Int J Cardiol. 2016;221:841–846. [DOI] [PubMed] [Google Scholar]
- 83.Chausiaux OE, Keyser M, Williams GP, et al. Heart failure decompensation alerts in a patient’s home using an automated, AI-driven, point-of-care device. BMJ Case Rep. 2022;15(4). doi: 10.1136/bcr-2021-248682 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.García-López I, Rodriguez-Villegas E. Extracting the Jugular Venous Pulse from Anterior Neck Contact Photoplethysmography. Sci Rep. 2020;10(1):3466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Abbaspourazad S, Elachqar O, Miller AC, Emrani S, Nallasamy U, Shapiro I. Large-scale Training of Foundation Models for Wearable Biosignals. arXiv [csLG]. Published online December 8, 2023. http://arxiv.org/abs/2312.05409 [Google Scholar]
- 86.Lubitz SA, Faranesh AZ, Selvaggi C, et al. Detection of Atrial Fibrillation in a Large Population Using Wearable Devices: The Fitbit Heart Study. Circulation. 2022;146(19):1415–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Perez MV, Mahaffey KW, Hedlin H, et al. Large-Scale Assessment of a Smartwatch to Identify Atrial Fibrillation. N Engl J Med. 2019;381(20):1909–1917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Sel K, Osman D, Huerta N, Edgar A, Pettigrew RI, Jafari R. Continuous cuffless blood pressure monitoring with a wearable ring bioimpedance device. NPJ Digit Med. 2023;6(1):59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Joung J, Jung CW, Lee HC, et al. Continuous cuffless blood pressure monitoring using photoplethysmography-based PPG2BP-net for high intrasubject blood pressure variations. Sci Rep. 2023;13(1):8605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Brugts JJ, Radhoe SP, Clephas PRD, et al. Remote haemodynamic monitoring of pulmonary artery pressures in patients with chronic heart failure (MONITOR-HF): a randomised clinical trial. Lancet. 2023;401(10394):2113–2123. [DOI] [PubMed] [Google Scholar]
- 91.Schutte AE, Kollias A, Stergiou GS. Blood pressure and its variability: classic and novel measurement techniques. Nat Rev Cardiol. 2022;19(10):643–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Hannigan BC, Cuthbert TJ, Ahmadizadeh C, Menon C. Distributed sensing along fibers for smart clothing. Sci Adv. 2024;10(12):eadj9708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Fukuma N, Hasumi E, Fujiu K, et al. Feasibility of a T-Shirt-Type Wearable Electrocardiography Monitor for Detection of Covert Atrial Fibrillation in Young Healthy Adults. Sci Rep. 2019;9(1):11768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Hu H, Huang H, Li M, et al. A wearable cardiac ultrasound imager. Nature. 2023;613(7945):667–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Klasnja P, Hekler EB, Shiffman S, et al. Microrandomized trials: An experimental design for developing just-in-time adaptive interventions. Health Psychol. 2015;34S(0):1220–1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Hughes A, Shandhi MMH, Master H, Dunn J, Brittain E. Wearable Devices in Cardiovascular Medicine. Circ Res. 2023;132(5):652–670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Wurie HR, Cappuccio FP. Cardiovascular disease in low- and middle-income countries: an urgent priority. Ethn Health. 2012;17(6):543–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Mocumbi AO. Cardiovascular Health Care in Low- and Middle-Income Countries. Circulation. 2024;149(8):557–559. [DOI] [PubMed] [Google Scholar]
- 99.Francis JR, Fairhurst H, Yan J, et al. Abbreviated Echocardiographic Screening for Rheumatic Heart Disease by Nonexperts with and without Offsite Expert Review: A Diagnostic Accuracy Study. J Am Soc Echocardiogr. 2023;36(7):733–745. [DOI] [PubMed] [Google Scholar]
- 100.Khunte A, Sangha V, Oikonomou EK, et al. Automated Diagnostic Reports from Images of Electrocardiograms at the Point-of-Care. medRxiv. Published online February 18, 2024:2024.02.17.24302976. [Google Scholar]
- 101.Na L, Yang C, Lo CC, Zhao F, Fukuoka Y, Aswani A. Feasibility of Reidentifying Individuals in Large National Physical Activity Data Sets From Which Protected Health Information Has Been Removed With Use of Machine Learning. JAMA Netw Open. 2018;1(8):e186040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wirth FN, Meurers T, Johns M, Prasser F. Privacy-preserving data sharing infrastructures for medical research: systematization and comparison. BMC Med Inform Decis Mak. 2021;21(1):242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ferrari F, van Dijck J, van den Bosch A. Observe, inspect, modify: Three conditions for generative AI governance. New Media & Society. Published online November 29, 2023:14614448231214812. [Google Scholar]
- 104.Department for Science, Innovation and Technology, Office for Artificial Intelligence. A Pro-Innovation Approach to AI Regulation; 2023. Accessed June 20, 2023. https://www.gov.uk/government/publications/ai-regulation-a-pro-innovation-approach/white-paper
- 105.Jadon A, Kumar S. Leveraging Generative AI Models for Synthetic Data Generation in Healthcare: Balancing Research and Privacy. arXiv [csLG]. Published online May 9, 2023. doi: 10.48550/ARXIV.2305.05247 [DOI] [Google Scholar]
- 106.Ghosheh G, Li J, Zhu T. A review of Generative Adversarial Networks for Electronic Health Records: applications, evaluation measures and data sources. arXiv [csLG]. Published online March 14, 2022. Accessed May 21, 2023. http://arxiv.org/abs/2203.07018 [Google Scholar]
- 107.Chen Y, Esmaeilzadeh P. Generative AI in Medical Practice: In-Depth Exploration of Privacy and Security Challenges. J Med Internet Res. 2024;26:e53008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Cai Y, Cai YQ, Tang LY, et al. Artificial intelligence in the risk prediction models of cardiovascular disease and development of an independent validation screening tool: a systematic review. BMC Med. 2024;22(1):56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ramspek CL, Jager KJ, Dekker FW, Zoccali C, van Diepen M. External validation of prognostic models: what, why, how, when and where? Clin Kidney J. 2021;14(1):49–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kamran F, Tang S, Otles E, et al. Early identification of patients admitted to hospital for covid-19 at risk of clinical deterioration: model development and multisite external validation study. BMJ. 2022;376:e068576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Blumenthal D A Step toward Interoperability of Health IT. N Engl J Med. 2022;387(24):2201–2203. [DOI] [PubMed] [Google Scholar]
- 112.Hripcsak G, Duke JD, Shah NH, et al. Observational Health Data Sciences and Informatics (OHDSI): Opportunities for Observational Researchers. Stud Health Technol Inform. 2015;216:574–578. [PMC free article] [PubMed] [Google Scholar]
- 113.Suchard MA, Schuemie MJ, Krumholz HM, et al. Comprehensive comparative effectiveness and safety of first-line antihypertensive drug classes: a systematic, multinational, large-scale analysis. Lancet. 2019;394(10211):1816–1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Khera R, Dhingra LS, Aminorroaya A, et al. Multinational patterns of second line antihyperglycaemic drug initiation across cardiovascular risk groups: federated pharmacoepidemiological evaluation in LEGEND-T2DM. BMJ Med. 2023;2(1):e000651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Sangha V, Mortazavi BJ, Haimovich AD, et al. Automated multilabel diagnosis on electrocardiographic images and signals. Nat Commun. 2022;13(1):1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Aiello M, Esposito G, Pagliari G, Borrelli P, Brancato V, Salvatore M. How does DICOM support big data management? Investigating its use in medical imaging community. Insights Imaging. 2021;12(1):164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Gilbert S, Harvey H, Melvin T, Vollebregt E, Wicks P. Large language model AI chatbots require approval as medical devices. Nat Med. 2023;29(10):2396–2398. [DOI] [PubMed] [Google Scholar]
- 118.Joshi G, Jain A, Araveeti SR, Adhikari S, Garg H, Bhandari M. FDA-Approved Artificial Intelligence and Machine Learning (AI/ML)-Enabled Medical Devices: An Updated Landscape. Electronics. 2024;13(3):498. [Google Scholar]
- 119.Buonora MJ, Axson SA, Cohen SM, Becker WC. Paths Forward for Clinicians Amidst the Rise of Unregulated Clinical Decision Support Software: Our Perspective on NarxCare. J Gen Intern Med. 2024;39(5):858–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Hacker P, Engel A, Mauer M. Regulating ChatGPT and other Large Generative AI Models. arXiv [csCY]. Published online February 5, 2023. doi: 10.48550/ARXIV.2302.02337 [DOI] [Google Scholar]
- 121.Cohen IG, Amarasingham R, Shah A, Xie B, Lo B. The legal and ethical concerns that arise from using complex predictive analytics in health care. Health Aff. 2014;33(7):1139–1147. [DOI] [PubMed] [Google Scholar]
- 122.Loh E ChatGPT and generative AI chatbots: challenges and opportunities for science, medicine and medical leaders. BMJ Lead. Published online May 2, 2023:leader-2023–000797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Krishnamoorthy M, Sjoding MW, Wiens J. Off-label use of artificial intelligence models in healthcare. Nat Med. Published online March 22, 2024. doi: 10.1038/s41591-024-02870-6 [DOI] [PubMed] [Google Scholar]
- 124.Cong Y, Endo T. Multi-Omics and Artificial Intelligence-Guided Drug Repositioning: Prospects, Challenges, and Lessons Learned from COVID-19. OMICS. 2022;26(7):361–371. [DOI] [PubMed] [Google Scholar]
- 125.Arnold C AlphaFold touted as next big thing for drug discovery - but is it? Nature. 2023;622(7981):15–17. [DOI] [PubMed] [Google Scholar]
- 126.Lin Z, Akin H, Rao R, et al. Evolutionary-scale prediction of atomic-level protein structure with a language model. Science. 2023;379(6637):1123–1130. [DOI] [PubMed] [Google Scholar]
- 127.Ren F, Ding X, Zheng M, et al. AlphaFold accelerates artificial intelligence powered drug discovery: efficient discovery of a novel CDK20 small molecule inhibitor. Chem Sci. 2023;14(6):1443–1452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Runcie NT, Mey ASJS. SILVR: Guided Diffusion for Molecule Generation. J Chem Inf Model. 2023;63(19):5996–6005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Corso G, Stärk H, Jing B, Barzilay R, Jaakkola T. DiffDock: Diffusion Steps, Twists, and Turns for Molecular Docking. arXiv [q-bioBM]. Published online October 4, 2022. http://arxiv.org/abs/2210.01776 [Google Scholar]
- 130.Madhukar NS, Khade PK, Huang L, et al. A Bayesian machine learning approach for drug target identification using diverse data types. Nat Commun. 2019;10(1):5221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Cochrane G, Akhtar R, Bonfield J, et al. Petabyte-scale innovations at the European Nucleotide Archive. Nucleic Acids Res. 2009;37(Database issue):D19–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.McDonough CW. Pharmacogenomics in Cardiovascular Diseases. Curr Protoc. 2021;1(7):e189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Claassens DMF, Vos GJA, Bergmeijer TO, et al. A Genotype-Guided Strategy for Oral P2Y12 Inhibitors in Primary PCI. N Engl J Med. 2019;381(17):1621–1631. [DOI] [PubMed] [Google Scholar]
- 134.Alreshq R, Ruberg FL. Clinical approach to genetic testing in amyloid cardiomyopathy: from mechanism to effective therapies. Curr Opin Cardiol. 2021;36(3):309–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Coorey G, Figtree GA, Fletcher DF, Redfern J. The health digital twin: advancing precision cardiovascular medicine. Nat Rev Cardiol. 2021;18(12):803–804. [DOI] [PubMed] [Google Scholar]
- 136.Klarin D, Natarajan P. Clinical utility of polygenic risk scores for coronary artery disease. Nat Rev Cardiol. 2022;19(5):291–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kent DM, van Klaveren D, Paulus JK, et al. The Predictive Approaches to Treatment effect Heterogeneity (PATH) Statement: Explanation and Elaboration. Ann Intern Med. 2020;172(1):W1–W25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Oikonomou EK, Van Dijk D, Parise H, et al. A phenomapping-derived tool to personalize the selection of anatomical vs. functional testing in evaluating chest pain (ASSIST). Eur Heart J. 2021;42(26):2536–2548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Oikonomou EK, Suchard MA, McGuire DK, Khera R. Phenomapping-Derived Tool to Individualize the Effect of Canagliflozin on Cardiovascular Risk in Type 2 Diabetes. Diabetes Care. 2022;45(4):965–974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Thangaraj PM, Shankar SV, Oikonomou EK, Khera R. RCT-Twin-GAN Generates Digital Twins of Randomized Control Trials Adapted to Real-world Patients to Enhance their Inference and Application. medRxiv. Published online December 15, 2023. doi: 10.1101/2023.12.06.23299464 [DOI] [Google Scholar]
- 141.Edward JA, Josey K, Bahn G, et al. Heterogeneous treatment effects of intensive glycemic control on major adverse cardiovascular events in the ACCORD and VADT trials: a machine-learning analysis. Cardiovasc Diabetol. 2022;21(1):58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Athey S, Imbens G. Recursive partitioning for heterogeneous causal effects. Proc Natl Acad Sci U S A. 2016;113(27):7353–7360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Hernán MA, Wang W, Leaf DE. Target Trial Emulation: A Framework for Causal Inference From Observational Data. JAMA. 2022;328(24):2446–2447. [DOI] [PubMed] [Google Scholar]
- 144.He X, Liu X, Zuo F, Shi H, Jing J. Artificial intelligence-based multi-omics analysis fuels cancer precision medicine. Semin Cancer Biol. 2023;88:187–200. [DOI] [PubMed] [Google Scholar]
- 145.Wang RS, Maron BA, Loscalzo J. Multiomics Network Medicine Approaches to Precision Medicine and Therapeutics in Cardiovascular Diseases. Arterioscler Thromb Vasc Biol. 2023;43(4):493–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Multiomic Health Limited. Leveraging deep omics data to tackle a global health challenge with precision. Springer Nature. Accessed April 27, 2024. https://www.nature.com/articles/d43747-022-00110-9 [Google Scholar]
- 147.Brown DG, Wobst HJ, Kapoor A, Kenna LA, Southall N. Clinical development times for innovative drugs. Nat Rev Drug Discov. 2022;21(11):793–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Liu R, Rizzo S, Whipple S, et al. Evaluating eligibility criteria of oncology trials using real-world data and AI. Nature. 2021;592(7855):629–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Woodcock J, LaVange LM. Master Protocols to Study Multiple Therapies, Multiple Diseases, or Both. N Engl J Med. 2017;377(1):62–70. [DOI] [PubMed] [Google Scholar]
- 150.Cunningham JW, Singh P, Reeder C, et al. Natural Language Processing for Adjudication of Heart Failure in a Multicenter Clinical Trial: A Secondary Analysis of a Randomized Clinical Trial. JAMA Cardiol. 2024;9(2):174–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Nargesi AA, Adejumo P, Dhingra L, et al. Automated Identification of Heart Failure with Reduced Ejection Fraction using Deep Learning-based Natural Language Processing. medRxiv. Published online September 11, 2023. doi: 10.1101/2023.09.10.23295315 [DOI] [PubMed] [Google Scholar]
- 152.Center for Drug Evaluation, Research. Adaptive design clinical trials for drugs and biologics guidance for industry. U.S. Food and Drug Administration. Published April 21, 2020. Accessed April 27, 2024. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/adaptive-design-clinical-trials-drugs-and-biologics-guidance-industry [Google Scholar]
- 153.Oikonomou EK, Thangaraj PM, Bhatt DL, et al. An explainable machine learning-based phenomapping strategy for adaptive predictive enrichment in randomized clinical trials. NPJ Digit Med. 2023;6(1):217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Thangaraj PM, Shankar SV, Huang S, et al. A Novel Digital Twin Strategy to Examine the Implications of Randomized Control Trials for Real-World Populations. medRxiv. Published online March 26, 2024. doi: 10.1101/2024.03.25.24304868 [DOI] [Google Scholar]
- 155.Fatumo S, Chikowore T, Choudhury A, Ayub M, Martin AR, Kuchenbaecker K. A roadmap to increase diversity in genomic studies. Nat Med. 2022;28(2):243–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Hu J, Yao J, Deng S, et al. Differences in Metabolomic Profiles Between Black and White Women and Risk of Coronary Heart Disease: an Observational Study of Women From Four US Cohorts. Circ Res. 2022;131(7):601–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Cruz DE, Tahir UA, Hu J, et al. Metabolomic Analysis of Coronary Heart Disease in an African American Cohort From the Jackson Heart Study. JAMA Cardiol. 2022;7(2):184–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Jones N How journals are fighting back against a wave of questionable images. Nature. 2024;626:697–698. [DOI] [PubMed] [Google Scholar]
- 159.McGuire AL, Gabriel S, Tishkoff SA, et al. The road ahead in genetics and genomics. Nat Rev Genet. 2020;21(10):581–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Caraballo C, Ndumele CD, Roy B, et al. Trends in Racial and Ethnic Disparities in Barriers to Timely Medical Care Among Adults in the US, 1999 to 2018. JAMA Health Forum. 2022;3(10):e223856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Mello MM, Rose S. Denial-Artificial Intelligence Tools and Health Insurance Coverage Decisions. JAMA Health Forum. 2024;5(3):e240622. [DOI] [PubMed] [Google Scholar]
- 162.Vishwanatha JK, Christian A, Sambamoorthi U, Thompson EL, Stinson K, Syed TA. Community perspectives on AI/ML and health equity: AIM-AHEAD nationwide stakeholder listening sessions. PLOS Digit Health. 2023;2(6):e0000288. [DOI] [PMC free article] [PubMed] [Google Scholar]