Next Article in Journal
Effectiveness and Safety of Mitral Valve Plasty in Patients with an Anomalous Origin of the Coronary Artery from the Pulmonary Artery
Next Article in Special Issue
Effectiveness of the Combination of Enalapril and Nifedipine for the Treatment of Hypertension versus Empirical Treatment in Primary Care Patients
Previous Article in Journal
Nomogram and Risk Calculator for Postoperative Tracheostomy after Heart Valve Surgery
Previous Article in Special Issue
H-Type Hypertension among Black South Africans and the Relationship between Homocysteine, Its Genetic Determinants and Estimates of Vascular Function
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Artificial Intelligence in Hypertension Management: An Ace up Your Sleeve

1
Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
2
Department of Advanced Biomedical Sciences, Federico II University of Naples, 80138 Naples, Italy
3
Cardiology Unit, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
4
Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
5
Department of Computer Science, University of Salerno, 84084 Fisciano, Italy
6
Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy
*
Author to whom correspondence should be addressed.
J. Cardiovasc. Dev. Dis. 2023, 10(2), 74; https://doi.org/10.3390/jcdd10020074
Submission received: 27 December 2022 / Revised: 5 February 2023 / Accepted: 7 February 2023 / Published: 9 February 2023
(This article belongs to the Special Issue Recent Advances in the Treatment of Hypertension)

Abstract

:
Arterial hypertension (AH) is a progressive issue that grows in importance with the increased average age of the world population. The potential role of artificial intelligence (AI) in its prevention and treatment is firmly recognized. Indeed, AI application allows personalized medicine and tailored treatment for each patient. Specifically, this article reviews the benefits of AI in AH management, pointing out diagnostic and therapeutic improvements without ignoring the limitations of this innovative scientific approach. Consequently, we conducted a detailed search on AI applications in AH: the articles (quantitative and qualitative) reviewed in this paper were obtained by searching journal databases such as PubMed and subject-specific professional websites, including Google Scholar. The search terms included artificial intelligence, artificial neural network, deep learning, machine learning, big data, arterial hypertension, blood pressure, blood pressure measurement, cardiovascular disease, and personalized medicine. Specifically, AI-based systems could help continuously monitor BP using wearable technologies; in particular, BP can be estimated from a photoplethysmograph (PPG) signal obtained from a smartphone or a smartwatch using DL. Furthermore, thanks to ML algorithms, it is possible to identify new hypertension genes for the early diagnosis of AH and the prevention of complications. Moreover, integrating AI with omics-based technologies will lead to the definition of the trajectory of the hypertensive patient and the use of the most appropriate drug. However, AI is not free from technical issues and biases, such as over/underfitting, the “black-box” nature of many ML algorithms, and patient data privacy. In conclusion, AI-based systems will change clinical practice for AH by identifying patient trajectories for new, personalized care plans and predicting patients’ risks and necessary therapy adjustments due to changes in disease progression and/or therapy response.

1. Introduction

Arterial hypertension (AH) is a global public health problem, and its treatment is primarily aimed at reducing associated cardiovascular (CV) morbidity and mortality [1,2]. AH affected more than 1.13 billion individuals in 2015, and the prevalence appears to affect approximately 35–45% of Campo’s overall population [3]. Moreover, AH is the most significant contributor to the global burden of CV diseases and represents a heavy socio-economic burden for many countries [4,5]; indeed, even moderate elevations in arterial blood pressure (BP) are associated with a significant reduction in life expectancy [6]. Furthermore, current data suggest that over 14 million people are unaware of their abnormal BP level; consequently, they are not receiving appropriate medication for it, nor do they engage in other interventions to maintain BP in the normal range [7]. Regardless of guidelines, BP control in hypertensive patients in treatment is insufficient due to many factors, including poor adherence to therapy [8]. Moreover, the definition of “hypertension” recapitulates several different sub-phenotypes influenced by multiple variables: gender, BMI, lifestyle conditions, and so on [9]. Furthermore, the pharmacological therapy used to treat essential AH has remained substantially unchanged in the last 20 years and is mainly focused on regulating vascular resistance [1,10]. Therefore, there is an evident gap in the knowledge required to deepen the multifaced aspects of AH and prompt the research and development of novel approaches.
In this scenario, the possibility of collecting, storing, and analyzing multiple pieces of information from a single patient in the form of electronic health records (EHRs) requires revising the conventional healthcare model of AH management using new technologies or monitoring techniques [11,12]. In this context, artificial intelligence (AI) is a technological method that has been in development in recent years [13,14,15], and, if used appropriately, it could have surprising results in developing predictive models of AH, formulating the diagnosis, stratifying patients, and identifying the most effective therapy (Table 1).
Specifically, accurate BP estimation is essential because BP is a risk factor for many clinical CV events, including stroke and dementia, and therefore can be integrated into several models for risk stratification [16,17]; moreover, in the computational simulation of CV disease, BP can influence the value of focal hemodynamic metrics, e.g., fractional flow [18]; consequently, patient-specific BP values can improve the accuracy of simulation results and have been applied in some recent models [19,20].
Therefore, AI-based systems might change clinical practice for AH by identifying patient trajectories for new, personalized care plans and predicting patients’ risks and necessary therapy adjustments due to changes in disease progression and/or therapy response. Accordingly, a basic knowledge of this science is essential because AI might change medical practice jobs: tiring and routine tasks could be completed by computers to free up time and allow cardiologists to carry out more difficult and sensitive tasks. Therefore, in this review, we describe multiple applications of AI, encompassing diagnostic, prognostic, and therapeutic issues currently unsolved in managing AH (Tables S1–S4).
Overall, this manuscript aims to provide a complete picture of the state of the art of AI in AH (primary and secondary) management, analyzing every aspect of the diagnosis, treatment, and patient follow-up, without neglecting the limitations and all of the possible tools to overcome them.

2. The Principles of AI

AI is a wide-ranging branch of computer science concerned with building smart machines capable of increasing their knowledge through an automatic learning process that typically requires human intelligence [21,22,23]. Therefore, AI is an interdisciplinary science with multiple approaches that incorporate reasoning (making inferences using data), natural language processing (ability to read and understand human languages), planning (ability to act autonomously and flexibly to create a sequence of actions to achieve a final goal), and machine learning (ML) (algorithms that develop automatically through experience) [21]. Specifically, AI based on ML techniques [15] is used to perform predictive analyses by examining mechanisms and associations among given variables from training datasets, which may consist of a variety of data inputs, including wearable devices, multi-omics, and standardized EHRs [10,24]. Essentially, in ML, the rules would be learned by algorithms directly from a set of data rather than being encoded by hand [25]; consequently, by using specific algorithms, ML can establish complex relationships among data, rules governing a system, behavioral patterns, and classification schemes [15]. The classic ML process begins with data acquisition, continues with feature extraction, algorithm selection, and model development, and leads to model evaluation and application [26] (Figure 1). Supervised and unsupervised learning are the most popular approaches employed in ML. Supervised learning is used to predict unknown outputs from a known labeled dataset, hypotheses, and appropriate algorithms, such as an artificial neural network (ANN), support vector machine (SVM), and K-nearest neighbor. The choice of the technique depends on the dataset’s features, number of variables, learning curve, training, and computation time [27,28]. Specifically, supervised learning provides predictions from big data analytics but requires manually labeled datasets and biases that can arise from the dataset itself or the algorithms [24].
On the other hand, in unsupervised learning techniques, there is no information on the features to be predicted; consequently, these techniques must learn from the relationships among the elements of a dataset and classify them without basing them on categories or labels [22]. Therefore, they look for structures, patterns, or characteristics in the source data that can be reproduced in new datasets [15]. ML mainly mimics the nervous system’s structure by creating ANNs, which are networks of units called artificial neurons structured into layers [29]. The system learns to generate patterns from data entered in the training session [29]. A specific ANN, consisting of more layers that allow for improved predictions from data, is known as a deep neural network (DNN). Its performance could be enhanced as the dimension of the training dataset rises [25]. Still, it largely depends on the distribution gap between training and test datasets: a highly divergent test dataset would test an ML prediction model on a feature space that it was not trained on, resulting in poor testing and results; additionally, a highly overlapping test dataset would not test the model for its generalization ability [30]. Specifically, DL employs algorithms such as DNNs and convolutional neural networks (CNNs) [15]. Nevertheless, regardless of its capability of using unlabeled datasets, unsupervised learning still has some limitations, such as the generalizability of cluster patterns identified from a cohort of patients, which can lead to overfitting to the training dataset, and the need to be validated in different large datasets [24]. In the real world, AI can provide tools to improve and extend the effectiveness of clinical interventions [15]. For example, incorporating AI into hypertension management could improve every stage of patient care, from diagnosis to therapy; consequently, the clinical practice could become more efficient and effective.

3. Methods

We conducted a detailed search on AI applications in AH. The research includes topics ranging from big data to complex technology-based interventions. Specifically, the articles reviewed in this paper were obtained by searching journal databases such as PubMed and subject-specific professional websites, including Google Scholar. The search terms included artificial intelligence, artificial neural network, deep learning, machine learning, big data, arterial hypertension, blood pressure, blood pressure measurement, CV disease, and personalized medicine. The inclusion criteria focus on articles directly or indirectly related to the topic of AI and AH. Specifically, both quantitative (measurable data) and qualitative (reasons, opinions, and motivations) reports were reviewed.
The authors independently screened titles and abstracts; subsequently, full texts were sourced for relevant articles. The reference lists of included trials and meta-analyses were also reviewed for significant articles.

4. AI in the Measurement of Blood Pressure

The commonly used methods for BP monitoring are either non-invasive inflatable cuff-based oscillometric or invasive arterial line manometric measurement. The former takes intermittent measures because a pause of at least 1–2 min between two BP measurements is necessary to avoid errors in the measurement [31,32]; moreover, the inflation of the cuff may disturb the patient, and the consequences of these disturbances are alterations in BP [33]. On the other hand, invasive arterial line manometric measurement has an elevated risk of complications; consequently, these unsolved issues drive the search for new non-invasive BP monitoring techniques.
In this scenario, AI algorithms could help improve precision, accuracy, and reproducibility in diagnosing and managing AH using emerging wearable technologies. Alternatives for monitoring BP are cuff-based devices (such as volume-clamp devices or wrist-worn inflatable cuffs) and cuffless devices that use mechanical and optical sensors to determine features of the blood pulse waveform shape (for example, tonometry [34], photoplethysmography [35], and capacitance [32]). In particular, cuffless blood pressure monitoring has been evaluated using a two-step algorithm with a single-channel photoplethysmograph (PPG). This system achieved an AAMI/ISO standard accuracy in all blood pressure categories except systolic hypotension [36]. Independently of the acquisition method, the received signals are preprocessed and sent for feature extraction and selection. Subsequently, the signals and the gathered data can be used to feed ML to obtain systolic BP (SBP) and diastolic BP (DBP) estimations from the raw signals [35] (Figure 2).
Since the volume and distension of arteries can be related to the pressure in the arteries, the PPG signal produces pulse waveforms that are similar to pressure waveforms created by tonometry. PPG offers the added advantage that it can be measured continuously using miniature, inexpensive, and wearable optical electronics [37]. However, PPG signal measurements are not without technical challenges; indeed, they require noise elimination, multisite measurement, multiphotodector development, event detection, event visualization, different models, the accurate positioning of sensors, and the calculation of propagation distances, without neglecting the impact of the variable PEP time on the pulse wave velocity timing [37]. Moreover, there are several PPG-based methods for estimating BP: the PPG signal alone and its derivate, ECG and PPG signals, BCG and PPG signals, and PCG and PPG signals; each has advantages and limitations [38,39,40,41,42], which, however, are beyond this discussion.

ML Algorithms in BP Estimation

To adapt to the nonlinearity of the dataset and to create a relationship between features and estimated BP, there are different ML approaches [43]:
  • Gaussian Process Regression: A Bayesian regression approach gives a probability distribution over all possible values [35,44].
  • Ensemble trees: The idea is to pull together a set of weak learners to create a strong learner [45].
  • Multivariate Linear Regression: It is a method to analyze the correlation, correlation direction, and strength between multiple independent variables and the dependent variable [33,46,47].
  • Support vector regression: It is a non-parametric algorithm that uses a kernel function (a class of algorithms for pattern analysis, whose general task is to find and study relations in datasets) [48,49,50,51].
  • Random forest, gradient boosting, and adaptive boosting regression [48,49].
  • CNN [52,53].
After hyper-parameter optimization, it is necessary to evaluate the performance of ML algorithms through the correlation between the acquired predicted data and the ground-truth data. The difference between reference and estimated BP could be considered using the following criteria: the mean absolute error, mean squared error, and correlation coefficient [35]. The role of parameter optimization is to lower the value of the predicted error. The mean absolute and standard deviations are the model’s predictive performance indicators.
Specifically, these AI-based systems could help continuously monitor BP using wearable technologies and improve AH management and outcomes [33,50]. Starting from the input (raw signals), we can reach the output (estimated SBP and DBP) through the algorithms of ML [24]. In particular, BP can be estimated from a PPG signal obtained from a smartphone or a smartwatch by using DL [54,55].
Moreover, future studies on AI and wearable devices need to confirm the above results and provide conclusive clinical data to support using a combination of AI and wearable-device-obtained data to correctly perform BP measurements, which may offer an alternative to current oscillometric methods [24].

5. Use of AI for the Prediction of Undiagnosed Hypertension

AH is generally asymptomatic or variably occurs with symptoms such as headache, dizziness, tinnitus, and nosebleeds in a minority of patients. For this reason, it is not always possible to readily identify and predict this disease.
The goal should be to implement primary and secondary prevention methods. This task could greatly benefit AI implementation, featuring the individual identification of each patient’s previous and new CV risk factors and treating modifiable risk factors. AI can predict the patient’s risk of AH development thanks to different algorithm settings with big sets of data. Predicting AH onset has been attempted in the last decade with progressively greater precision thanks to technological advancements.
Accordingly, risk factors for AH, such as low levels of education, a sedentary job, a family history of AH, demographic data, routine blood tests, BMI, waist and hip circumference and ratio, diet, physical exercise, and salt and alcohol intake, showed effectiveness in predicting elevated SBP using ML techniques [56,57,58], for instance, an ANN [59]. On the other hand, AI techniques are also emerging in gene expression analysis; specifically, it is possible to improve AH predictive models by integrating data from a combination of gene expression and next-generation sequencing in ML analysis [60]. Furthermore, thanks to ML algorithms, it is possible to identify new hypertension genes; accordingly, Li et al. [61] analyzed gene expression in hypertensive patients, thus identifying 177 new hypertension genes thanks to ML algorithm development: this was then integrated with environmental factors in evaluating the risk of AH [53,62].
Furthermore, health risk prediction using a DL architecture appears suitable for extensive complex datasets, as shown by Maxwell et al. [63]. Using physical examination records of 110,300 patients, it was possible to examine, classify, and learn each risk factor contributing to chronic diseases such as AH, diabetes, and liver steatosis [63].
With the same principle, Ye et al. [56] validated an accurate 1-year risk prediction model for the incidence of essential AH. In this retrospective and prospective study, risk factors (type 2 diabetes, dyslipidemias, CV diseases, etc.) were calculated, evaluated, and stratified, yielding an accuracy of 91.7% for the prediction of the AH incidence at 1 year, with 87% in the prospective cohort (validation) [56].
Furthermore, the AI risk assessment of AH development can also benefit from cardiorespiratory fitness data obtained from treadmill exercise stress testing [64]. Overall, AI has the potential to increase efficacy by aggregating immense volumes of information, virtually predicting the risk of AH and consequently preventing or delaying the development of this disease by directing early interventions [24].
Making the AH diagnosis using validated guidelines [65,66] is an easy task for AI. Using inputs such as BP, comorbidities, demographic data, and routine blood tests, the accuracy can reach 92.85% with an ANN [67]. With a similar dataset (age, BP, BMI, serum lipoproteins profile, smoking habit, and exercise), using ML, the accuracy is estimated at 82% [68].
The most common example of AI for AH diagnosis, and probably still the most accurate, is twenty-four-hour ambulatory BP monitoring (ABPM) [69]. Thanks to direct and regular interval BP measurements and a relatively easy algorithm, ABPM software can identify and classify AH. ABPM is particularly useful in correctly evaluating white coats and masked AH [70,71].
New approaches based on ANNs try to estimate BP for AH diagnosis in healthy individuals using factors such as BMI, age, exercise, smoking, and alcohol consumption, although with limited accuracy and efficacy [68]. An interesting method for SBP prediction is the use of retinal fundus images. This factor was used by Poplin et al. [72], who showed that DL could be used in this case, even if the BP error was 11.23 mmHg, clearly not accurate enough to be of diagnostic use.
The ability of AI to diagnose AH is dependent on its accuracy. Further studies will identify more methods of BP prediction for the early diagnosis of AH and complication prevention. Looking at the world around us, AI’s future in AH diagnosis and management probably lies in systems that can constantly retrieve vital signs, such as smartphones, smartwatches, and all of the relative accessories [73].

6. Targeting AH by AI

Optimizing pharmacological therapy to achieve the optimal control of BP levels represents a tough challenge for physicians. The management of AH is based on evidence-based medicine and expert opinions; BP control rates remain poor worldwide and are far from satisfactory across Europe [74].
Specifically, the importance of AI in this field is in the robust identification of modifiable factors that impact the evolution of AH; moreover, this new instrument could support the choice of the most appropriate management of AH. Indeed, most hypertensive patients need medication and lifestyle changes to achieve optimal BP control, and AI is helpful due to its better capability of identifying the best combination therapy compared to standard analysis [74].
Several works discussed and analyzed the opportunity of using AI to evaluate the impacts of different factors on BP values. Koren et al. applied ANN and decision trees to identify parameters contributing to Campo’s success in his AH drug treatment [75]. Indeed, ML algorithms allow better real-world data analysis, identifying patterns and trends not easily recognized with classical experimental or observational approaches. Randomized trials usually compare the effectiveness of a drug to a placebo or, at best, to an already-approved compound. However, evaluating the differences between and advantages of drug combinations, frequently necessary for AH treatment, is a more complex task [75] that AI could fulfill. Here, we provide some previous experience and approaches; for example, the computer was presented with a training dataset in the first training phase, and for each instance, it was given the correct classification. Specifically, the appropriate classification of the algorithm was divided into two reference categories: “treatment success”, defined as BP lower than 140/90 mmHg within 90 days after starting therapy, and “treatment failure” in any other case. Finally, AI decision trees and ANN compared various classes of antihypertensives in patients: beta-blockers alone or in combination were the most effective [75]. In another study, ML methods predicted individual treatment effects of intensive BP therapy [76]; notably, the results revealed an improvement in the discrimination and calibration of individualized medications from clinical trial data. Moreover, Ye X. et al. [77] demonstrated the potential of using predictive models to select optimal AH treatment pathways; specifically, along with clinical guidelines and guideline-based CDS systems, the LSTM models show the best prediction for achieving the optimal control of BP with different combinations of treatments. ML can also detect adherence to antihypertensive therapy by analyzing data recorded from a smartphone application to promote patients’ awareness, self-monitoring, and treatment compliance [78].
Nonetheless, the exploitation of AI in the healthcare field is still in its early phases, and its potential is not entirely explored. Indeed, AI represents the opportunity for a global evaluation of the patient in his or her complexity because it allows the integration of clinical, demographic, biochemical, and instrumental data to develop models that physicians and healthcare providers could use to improve AH management and test new drug therapies using the multi-omics method [79,80].
We are now entering an era in which omics-based technology evolution will start to deliver long-promised elements that may improve the understanding of the complex mechanistic basis of this disease [79]. Accordingly, the key to better disease management will pass through personalized medicine. The key to this is future drug discovery that will be possible thanks to enhanced AI technologies exploiting information integration. Data from sequenced genomes, functional genomics, protein profiling, metabolomics, and bioinformatics may ensure a better comprehensive systems-based analysis for further understanding AH disease’s complexities. Integrating AI with omics-based technologies will lead to the definition of the trajectory of the hypertensive patient and the use of the most appropriate drug.
In conclusion, AI can improve intelligent healthcare systems that provide personalized recommendations and treatment approaches [24].

7. Definition of the Hypertensive Patient’s Trajectory: Role of AI in AH Prognosis

According to the recent AHA and ESC guidelines [74,81], the evaluation of the prognosis of AH is related to the demographic characteristics, the progression of organ injury, and comorbidities of the patients. Predicting the outcomes of hypertensive patients is significant research work [82].
There are currently several clinical risk scores intended for specific populations and risk groups [83,84,85]. Nevertheless, most of these scores are based on linear models and might therefore lack specificity and sensitivity in certain subgroups [86]. Moreover, the wide variety of scores may also cause slow adoption in real-world clinical practice [86].
The role of AI concerning the prognostic establishment of AH is due to the utility of ML algorithms trained on large datasets to estimate prognoses and potentially guide medical treatments [87].
The recent focus on AI and ML methods for AH prognosis is related to the critical repercussions of elevated BP for the risk of developing organ damage. Despite commonly used statistical methods, AI systems can process large amounts of complex data. Therefore, AI and ML aim to process a prognostic model that has clinical relevance in managing patients in the real world [88]. The prognostic impact of AH needs the stratification of patients based on their global evaluation with the integration of different parameters, such as the grade and stage of AH, BP control, and concomitant comorbidities. Indeed, AI can stratify patients correctly through the use of classification algorithms (SVM, C4.5 decision tree, random forest (RF), and extreme gradient boosting (XGBoost)); specifically, XGBoost has the best prediction performance [82].
However, the accuracy of the definition of the individual trajectory can be enhanced by a prolonged follow-up [89] and an increase in the number of relevant features, such as genomic variants associated with specific hypertensive phenotypes and different outcomes [61,90].
Moreover, the prognosis determines the frequency and the type of clinical monitoring. Specifically, AI allows the elaboration of new risk stratification to develop novel prognostic layers in favor of personalized medicine with the prospect of efficiently measuring the grade classification of AH [91] and identifying different outcomes’ profiles [89]. In conclusion, the clinical advantages of implementing AI systems and their integration into current prognostic stratification might grant the correct identification of classes of outcomes of patients, improving their clinical management.

8. AI in Secondary Arterial Hypertension

The causes of hypertension are multiple, and we must not overlook secondary arterial hypertension; indeed, in 10–15% of cases, the specific cause underlying hypertension can be identified [92]. Specifically, to arrive at the diagnosis of essential hypertension, all causes of secondary hypertension must be ruled out, and the accurate history, thorough examination, and performance of all necessary tests based on the data collected always place the physician and his or her knowledge of internal medicine at the basis of the evaluation of each hypertensive patient. Consequently, diagnosis and patient management must be connected to the clinician, who can use AI to speed up and facilitate his or her task. In any case, AI and ML cannot wholly replace the doctor’s role, but they represent a useful tool in his hands. Identifying secondary hypertension in its various subtypes is essential to preventing and targeting the treatment of CV complications. However, screening for secondary hypertension can be time-consuming, expensive, and difficult; consequently, simplified diagnostic tests are urgently required to distinguish between primary and secondary hypertension to address the current underdiagnosis of the latter. In children and adolescents, the most common causes of hypertension are renal parenchymal or vascular disease and aortic coarctation [93]; in adults, earlier studies identified renal parenchymal and vascular diseases as the most common causes of secondary hypertension. Obstructive sleep apnea (OSA) was recognized as an exceedingly common cause of secondary hypertension [94]. Among endocrine causes, we include primary aldosteronism (the most common), thyroid disease (hypo- or hyperthyroidism), hypercortisolism (Cushing’s), and finally, phaeochromocytoma [92]. The application of ML methods to the etiological diagnosis of secondary hypertension can be helpful in clinical practice. Accordingly, AI technology should be implemented cautiously; to be a partner of clinicians, there is still a long way to go, but it can serve as a virtual assistant and enable clinicians to promote quality and increase efficiency. Based on EMRs from Fuwai Hospital, five ML prediction models with good performance and applicability to the etiology detection of secondary hypertension were developed by Campo [95], which demonstrated that ML approaches were feasible and effective in diagnosing secondary hypertension. Reel and colleagues [95] showed that the MOmics approach provided better discriminatory power compared to single-omics (monoomics) data analysis and appropriately classified different forms of endocrine hypertension with high sensitivity and specificity, providing potential diagnostic biomarker combinations for diagnosing secondary hypertension subtypes. However, there still needs to be more data in the literature on the application of AI in the field of secondary hypertension; consequently, these innovative and clinically relevant prediction models still require further validation and more clinical tests before being implemented into clinical practice.

9. Limitations of Applying ML in CV Research

Despite the great importance that AI has taken on in the last few years, it is necessary to underline that this system is not free from technical issues and biases (Table S5). The quality of the algorithms underlying AI technology can be affected by some limitations, such as the inconsistent quality of the studies that form the databases at the heart of AI [96].
Specifically, overfitting is a significant issue common to all ML models: this happens when a model has become overly attuned to the training data, such that it does not generalize to new datasets; this is the opposite of underfitting, where the algorithm cannot wholly capture the predictive power of the data. These two issues (over- and underfitting) could be solved by improving the parameters of the model or making modifications to the training set [21].
Moreover, it is also essential to consider the adversarial robustness of a model (ability to resist being fooled), which could be improved by enlarging the training set [97].
Furthermore, many clinicians remain wary of ML because of concerns about the “black-box” nature of many ML algorithms. These models are sufficiently complex that they are not directly interpretable to humans. Subsequently, the lack of interpretability of predictive models can undermine trust in those models, especially in the medical field, in which so many decisions are life-and-death issues [98]. To be trusted, users must comprehend the model outputs [98]. Consequently, the “explainability” technique in ML seeks to imbue humans with a high level of understanding of how an algorithm works and makes decisions without carefully considering each step [99]; however, the nature of explanations as approximations may omit important information about how black-box models work and why they make specific predictions [100].
Another flaw is the lack of discernment of predisposing factors for specific CV diseases from confounding factors. Moreover, it is also possible that, during data analysis processes by specialized clinicians, some may contribute to lowering the accuracy of the databases owing to their biases, thus making their validation even more time-consuming [28].
It would also be necessary for some AI-automated diagnostic CV algorithms to identify CV risk factors that are essential but still not unanimously recognized since, sometimes, there is no consensus among cardiologists. Therefore, it would be desirable to validate data through a clinical consensus before merging them into the AI, though expensive and time-consuming. Another important aspect is the disparity between various racial groups, ethnic minorities, and social classes. Some CV diseases are more represented in some races and/or ethnic minorities and can express themselves with different phenotypes [101].
Prospectively, it will be necessary to render AI accessible to patients and promote awareness, self-monitoring, healthy behaviors, and therapeutic adherence by integrating new technologies, such as wearable devices that can automatically analyze activity levels and give feedback, such as lifestyle and drug dose changes [24].

10. Conclusions

The study of AH needs a revolution, and its future may lie in the favorable convergence of digital data and biotechnological and biomedical sciences and their implementation in healthcare delivery with new delivery models and effective strategies for population health. With AI, we could better understand epigenetic changes relevant to AH onset and progression, potentially classify the risk of AH in individuals, identify the mechanism of poorly controlled AH, and evaluate treatment responses in clinical trials using a multi-omics approach [102]. Furthermore, AI could target healthy individuals who are at higher risk of developing AH and may benefit from lifestyle modifications for the primary prevention of CV disease [24].
In conclusion, AH prediction and management using a combination of AI and wearable technology could potentially be the first real chance for precision CV medicine [102]. Moreover, future AI-enhanced AH care will encourage patient awareness, self-monitoring, healthy behaviors, and treatment adherence, along with developing digital technologies [24]. Therefore, future research needs to focus on precision AH medicine utilizing AI-based technologies to reduce the global burden of AH without neglecting the significant limitations that this approach still has.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/jcdd10020074/s1. Table S1: Major contributions of AI in AH measurement. Table S2: Major contributions of AI in AH prediction and diagnosis. Table S3: Major contributions of AI in AH treatment. Table S4: Major contributions of AI in AH outcome. Table S5: Limitations of applying ML in cardiovascular research Supplementary References [34,35,51,56,57,58,59,60,61,63,64,68,75,76,77,82,89,90,103].

Author Contributions

Conceptualization, V.V. and M.C.; data curation, V.V., M.C., C.I., A.R., M.T., A.G., B.S., A.M., M.R.R. and P.D.P.; writing—original draft preparation, V.V., M.C., C.I., A.R., M.T., N.V., R.G., A.G., A.C. and C.V.; writing—review and editing, V.V., M.C., C.I., C.M., G.D., A.B., G.G. and C.V.; supervision, V.V. and M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the project “SOLOMAX—SOciaLNetwOrk of MedicAlEXperiences” (to MC) from the Italian Ministry of Economic Development (MISE).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sorriento, D.; Rusciano, M.R.; Visco, V.; Fiordelisi, A.; Cerasuolo, F.A.; Poggio, P.; Ciccarelli, M.; Iaccarino, G. The Metabolic Role of GRK2 in Insulin Resistance and Associated Conditions. Cells 2021, 10, 167. [Google Scholar] [CrossRef]
  2. Visco, V.; Finelli, R.; Pascale, A.V.; Giannotti, R.; Fabbricatore, D.; Ragosa, N.; Ciccarelli, M.; Iaccarino, G. Larger Blood Pressure Reduction by Fixed-Dose Compared to Free Dose Combination Therapy of ACE Inhibitor and Calcium Antagonist in Hypertensive Patients. Transl. Med. UniSa 2017, 16, 17–23. [Google Scholar]
  3. NCD Risk Factor Collaboration. Worldwide trends in blood pressure from 1975 to 2015: A pooled analysis of 1479 population-based measurement studies with 19.1 million participants. Lancet 2017, 389, 37–55. [Google Scholar] [CrossRef]
  4. Izzo, C.; Vitillo, P.; Di Pietro, P.; Visco, V.; Strianese, A.; Virtuoso, N.; Ciccarelli, M.; Galasso, G.; Carrizzo, A.; Vecchione, C. The Role of Oxidative Stress in Cardiovascular Aging and Cardiovascular Diseases. Life 2021, 11, 60. [Google Scholar] [CrossRef]
  5. Whitworth, J.A.; World Health Organization; International Society of Hypertension Writing Group. 2003 World Health Organization (WHO)/International Society of Hypertension (ISH) statement on management of hypertension. J. Hypertens. 2003, 21, 1983–1992. [Google Scholar] [CrossRef]
  6. Pereira da Silva, A.; Matos, A.; Aguiar, L.; Ramos-Marques, N.; Ribeiro, R.; Gil, A.; Gorjao-Clara, J.; Bicho, M. Hypertension and longevity: Role of genetic polymorphisms in renin-angiotensin-aldosterone system and endothelial nitric oxide synthase. Mol. Cell Biochem. 2019, 455, 61–71. [Google Scholar] [CrossRef]
  7. Wall, H.K.; Ritchey, M.D.; Gillespie, C.; Omura, J.D.; Jamal, A.; George, M.G. Vital Signs: Prevalence of Key Cardiovascular Disease Risk Factors for Million Hearts 2022—United States, 2011–2016. MMWR Morb. Mortal Wkly. Rep. 2018, 67, 983–991. [Google Scholar] [CrossRef]
  8. O’Brien, E.; Asmar, R.; Beilin, L.; Imai, Y.; Mancia, G.; Mengden, T.; Myers, M.; Padfield, P.; Palatini, P.; Parati, G.; et al. Practice guidelines of the European Society of Hypertension for clinic, ambulatory and self blood pressure measurement. J. Hypertens. 2005, 23, 697–701. [Google Scholar] [CrossRef]
  9. Visco, V.; Pascale, A.V.; Virtuoso, N.; Mongiello, F.; Cinque, F.; Gioia, R.; Finelli, R.; Mazzeo, P.; Manzi, M.V.; Morisco, C.; et al. Serum Uric Acid and Left Ventricular Mass in Essential Hypertension. Front. Cardiovasc. Med. 2020, 7, 570000. [Google Scholar] [CrossRef]
  10. Dzau, V.J.; Balatbat, C.A. Future of Hypertension. Hypertension 2019, 74, 450–457. [Google Scholar] [CrossRef]
  11. De Luca, V.; Tramontano, G.; Riccio, L.; Trama, U.; Buono, P.; Losasso, M.; Bracale, U.M.; Annuzzi, G.; Zampetti, R.; Cacciatore, F.; et al. “One Health” Approach for Health Innovation and Active Aging in Campania (Italy). Front. Public Health 2021, 9, 658959. [Google Scholar] [CrossRef]
  12. Visco, V.; Finelli, R.; Pascale, A.V.; Mazzeo, P.; Ragosa, N.; Trimarco, V.; Illario, M.; Ciccarelli, M.; Iaccarino, G. Difficult-to-control hypertension: Identification of clinical predictors and use of ICT-based integrated care to facilitate blood pressure control. J. Hum. Hypertens. 2018, 32, 467–476. [Google Scholar] [CrossRef]
  13. Visco, V.; Esposito, C.; Manzo, M.; Fiorentino, A.; Galasso, G.; Vecchione, C.; Ciccarelli, M. A Multistep Approach to Deal With Advanced Heart Failure: A Case Report on the Positive Effect of Cardiac Contractility Modulation Therapy on Pulmonary Pressure Measured by CardioMEMS. Front. Cardiovasc. Med. 2022, 9, 874433. [Google Scholar] [CrossRef]
  14. Visco, V.; Esposito, C.; Vitillo, P.; Vecchione, C.; Ciccarelli, M. It is easy to see, but it is better to foresee: A case report on the favourable alliance between CardioMEMS and levosimendan. Eur. Heart J. Case Rep. 2020, 4, 1–5. [Google Scholar] [CrossRef]
  15. Visco, V.; Ferruzzi, G.J.; Nicastro, F.; Virtuoso, N.; Carrizzo, A.; Galasso, G.; Vecchione, C.; Ciccarelli, M. Artificial Intelligence as a Business Partner in Cardiovascular Precision Medicine: An Emerging Approach for Disease Detection and Treatment Optimization. Curr. Med. Chem. 2020, 28, 6569–6590. [Google Scholar] [CrossRef]
  16. Zhou, J.; Lee, S.; Wong, W.T.; Waleed, K.B.; Leung, K.S.K.; Lee, T.T.L.; Wai, A.K.C.; Liu, T.; Chang, C.; Cheung, B.M.Y.; et al. Gender-specific clinical risk scores incorporating blood pressure variability for predicting incident dementia. J. Am. Med. Inform. Assoc. 2022, 29, 335–347. [Google Scholar] [CrossRef]
  17. Tian, X.; Fang, H.; Lan, L.; Ip, H.L.; Abrigo, J.; Liu, H.; Zheng, L.; Fan, F.S.Y.; Ma, S.H.; Ip, B.; et al. Risk stratification in symptomatic intracranial atherosclerotic disease with conventional vascular risk factors and cerebral haemodynamics. Stroke Vasc. Neurol. 2022, svn-2022-001606. [Google Scholar] [CrossRef]
  18. Leng, X.; Lan, L.; Ip, V.H.L.; Liu, H.; Abrigo, J.; Liebeskind, D.S.; Wong, L.K.S.; Leung, T.W. Noninvasive fractional flow in intracranial atherosclerotic stenosis: Reproducibility, limitations, and perspectives. J. Neurol. Sci. 2017, 381, 150–152. [Google Scholar] [CrossRef]
  19. Zhong, L.; Zhang, J.M.; Su, B.; Tan, R.S.; Allen, J.C.; Kassab, G.S. Application of Patient-Specific Computational Fluid Dynamics in Coronary and Intra-Cardiac Flow Simulations: Challenges and Opportunities. Front. Physiol. 2018, 9, 742. [Google Scholar] [CrossRef]
  20. Wang, X.; Liu, H.; Xu, M.; Chen, C.; Ma, L.; Dai, F. Efficacy assessment of superficial temporal artery-middle cerebral artery bypass surgery in treating moyamoya disease from a hemodynamic perspective: A pilot study using computational modeling and perfusion imaging. Acta Neurochir. 2023. [Google Scholar] [CrossRef]
  21. Padmanabhan, S.; Tran, T.Q.B.; Dominiczak, A.F. Artificial Intelligence in Hypertension: Seeing Through a Glass Darkly. Circ. Res. 2021, 128, 1100–1118. [Google Scholar] [CrossRef]
  22. Dorado-Diaz, P.I.; Sampedro-Gomez, J.; Vicente-Palacios, V.; Sanchez, P.L. Applications of Artificial Intelligence in Cardiology. The Future is Already Here. Rev. Esp. Cardiol. Engl. Ed. 2019, 72, 1065–1075. [Google Scholar] [CrossRef]
  23. Bonderman, D. Artificial intelligence in cardiology. Wien. Klin. Wochenschr. 2017, 129, 866–868. [Google Scholar] [CrossRef]
  24. Chaikijurajai, T.; Laffin, L.J.; Tang, W.H.W. Artificial Intelligence and Hypertension: Recent Advances and Future Outlook. Am. J. Hypertens. 2020, 33, 967–974. [Google Scholar] [CrossRef]
  25. Schmidt-Erfurth, U.; Sadeghipour, A.; Gerendas, B.S.; Waldstein, S.M.; Bogunovic, H. Artificial intelligence in retina. Prog. Retin. Eye Res. 2018, 67, 1–29. [Google Scholar] [CrossRef]
  26. Johnson, K.W.; Torres Soto, J.; Glicksberg, B.S.; Shameer, K.; Miotto, R.; Ali, M.; Ashley, E.; Dudley, J.T. Artificial Intelligence in Cardiology. J. Am. Coll. Cardiol. 2018, 71, 2668–2679. [Google Scholar] [CrossRef]
  27. Bzdok, D.; Krzywinski, M.; Altman, N. Machine learning: Supervised methods. Nat. Methods 2018, 15, 5–6. [Google Scholar] [CrossRef]
  28. Krittanawong, C.; Zhang, H.; Wang, Z.; Aydar, M.; Kitai, T. Artificial Intelligence in Precision Cardiovascular Medicine. J. Am. Coll. Cardiol. 2017, 69, 2657–2664. [Google Scholar] [CrossRef]
  29. Ahuja, A.S. The impact of artificial intelligence in medicine on the future role of the physician. PeerJ 2019, 7, e7702. [Google Scholar] [CrossRef]
  30. Turhan, B. On the dataset shift problem in software engineering prediction models. Empir. Softw. Eng. 2012, 17, 62–75. [Google Scholar] [CrossRef]
  31. Campbell, N.R.; Chockalingam, A.; Fodor, J.G.; McKay, D.W. Accurate, reproducible measurement of blood pressure. CMAJ 1990, 143, 19–24. [Google Scholar]
  32. Quan, X.; Liu, J.; Roxlo, T.; Siddharth, S.; Leong, W.; Muir, A.; Cheong, S.M.; Rao, A. Advances in Non-Invasive Blood Pressure Monitoring. Sensors 2021, 21, 4273. [Google Scholar] [CrossRef]
  33. Gesche, H.; Grosskurth, D.; Kuchler, G.; Patzak, A. Continuous blood pressure measurement by using the pulse transit time: Comparison to a cuff-based method. Eur. J. Appl. Physiol. 2012, 112, 309–315. [Google Scholar] [CrossRef]
  34. Huang, K.H.; Tan, F.; Wang, T.D.; Yang, Y.J. A Highly Sensitive Pressure-Sensing Array for Blood Pressure Estimation Assisted by Machine-Learning Techniques. Sensors 2019, 19, 848. [Google Scholar] [CrossRef]
  35. Chowdhury, M.H.; Shuzan, M.N.I.; Chowdhury, M.E.H.; Mahbub, Z.B.; Uddin, M.M.; Khandakar, A.; Reaz, M.B.I. Estimating Blood Pressure from the Photoplethysmogram Signal and Demographic Features Using Machine Learning Techniques. Sensors 2020, 20, 3127. [Google Scholar] [CrossRef]
  36. Khalid, S.; Liu, H.; Zia, T.; Zhang, J.; Chen, F.; Zheng, D. Cuffless Blood Pressure Estimation Using Single Channel Photoplethysmography: A Two-Step Method. IEEE Access 2020, 8, 58146–58154. [Google Scholar] [CrossRef]
  37. Elgendi, M.; Fletcher, R.; Liang, Y.; Howard, N.; Lovell, N.H.; Abbott, D.; Lim, K.; Ward, R. The use of photoplethysmography for assessing hypertension. NPJ Digit. Med. 2019, 2, 60. [Google Scholar] [CrossRef]
  38. Zheng, Y.; Poon, C.C.; Yan, B.P.; Lau, J.Y. Pulse Arrival Time Based Cuff-Less and 24-H Wearable Blood Pressure Monitoring and its Diagnostic Value in Hypertension. J. Med. Syst. 2016, 40, 195. [Google Scholar] [CrossRef]
  39. Pandian, P.S.; Mohanavelu, K.; Safeer, K.P.; Kotresh, T.M.; Shakunthala, D.T.; Gopal, P.; Padaki, V.C. Smart Vest: Wearable multi-parameter remote physiological monitoring system. Med. Eng. Phys. 2008, 30, 466–477. [Google Scholar] [CrossRef]
  40. Plante, T.B.; Urrea, B.; MacFarlane, Z.T.; Blumenthal, R.S.; Miller, E.R., 3rd; Appel, L.J.; Martin, S.S. Validation of the Instant Blood Pressure Smartphone App. JAMA Intern. Med. 2016, 176, 700–702. [Google Scholar] [CrossRef]
  41. Zhang, Q.; Zhou, D.; Zeng, X. Highly wearable cuff-less blood pressure and heart rate monitoring with single-arm electrocardiogram and photoplethysmogram signals. Biomed. Eng. Online 2017, 16, 23. [Google Scholar] [CrossRef]
  42. Radha, M.; de Groot, K.; Rajani, N.; Wong, C.C.P.; Kobold, N.; Vos, V.; Fonseca, P.; Mastellos, N.; Wark, P.A.; Velthoven, N.; et al. Estimating blood pressure trends and the nocturnal dip from photoplethysmography. Physiol. Meas. 2019, 40, 025006. [Google Scholar] [CrossRef] [Green Version]
  43. Hare, A.J.; Chokshi, N.; Adusumalli, S. Novel Digital Technologies for Blood Pressure Monitoring and Hypertension Management. Curr. Cardiovasc. Risk Rep. 2021, 15, 11. [Google Scholar] [CrossRef]
  44. Nour, M.; Kandaz, D.; Ucar, M.K.; Polat, K.; Alhudhaif, A. Machine Learning and Electrocardiography Signal-Based Minimum Calculation Time Detection for Blood Pressure Detection. Comput. Math. Methods Med. 2022, 2022, 5714454. [Google Scholar] [CrossRef]
  45. Kumar, P.S.; Rai, P.; Ramasamy, M.; Varadan, V.K.; Varadan, V.K. Multiparametric cloth-based wearable, SimpleSense, estimates blood pressure. Sci. Rep. 2022, 12, 13059. [Google Scholar] [CrossRef]
  46. Mase, M.; Mattei, W.; Cucino, R.; Faes, L.; Nollo, G. Feasibility of cuff-free measurement of systolic and diastolic arterial blood pressure. J. Electrocardiol. 2011, 44, 201–207. [Google Scholar] [CrossRef]
  47. Park, M.; Kang, H.; Huh, Y.; Kim, K.C. Cuffless and noninvasive measurement of systolic blood pressure, diastolic blood pressure, mean arterial pressure and pulse pressure using radial artery tonometry pressure sensor with concept of Korean traditional medicine. Annu. Int. Conf. IEEE Eng. Med. Biol. Soc. 2007, 2007, 3597–3600. [Google Scholar] [CrossRef]
  48. Kachuee, M.; Kiani, M.M.; Mohammadzade, H.; Shabany, M. Cuffless Blood Pressure Estimation Algorithms for Continuous Health-Care Monitoring. IEEE Trans. Biomed. Eng. 2017, 64, 859–869. [Google Scholar] [CrossRef]
  49. Monte-Moreno, E. Non-invasive estimate of blood glucose and blood pressure from a photoplethysmograph by means of machine learning techniques. Artif. Intell. Med. 2011, 53, 127–138. [Google Scholar] [CrossRef]
  50. Peng, R.C.; Yan, W.R.; Zhang, N.L.; Lin, W.H.; Zhou, X.L.; Zhang, Y.T. Cuffless and Continuous Blood Pressure Estimation from the Heart Sound Signals. Sensors 2015, 15, 23653–23666. [Google Scholar] [CrossRef]
  51. Khalid, S.G.; Zhang, J.; Chen, F.; Zheng, D. Blood Pressure Estimation Using Photoplethysmography Only: Comparison between Different Machine Learning Approaches. J. Healthc. Eng. 2018, 2018, 1548647. [Google Scholar] [CrossRef]
  52. Yan, C.; Li, Z.; Zhao, W.; Hu, J.; Jia, D.; Wang, H.; You, T. Novel Deep Convolutional Neural Network for Cuff-less Blood Pressure Measurement Using ECG and PPG Signals. Annu. Int. Conf. IEEE Eng. Med. Biol. Soc. 2019, 2019, 1917–1920. [Google Scholar] [CrossRef]
  53. Rastegar, S.; Gholamhosseini, H.; Lowe, A.; Mehdipour, F.; Linden, M. Estimating Systolic Blood Pressure Using Convolutional Neural Networks. Stud. Health Technol. Inform. 2019, 261, 143–149. [Google Scholar]
  54. Tison, G.H.; Singh, A.C.; Ohashi, D.A.; Hsieh, J.T.; Ballinger, B.M.; Olgin, J.E.; Marcus, G.M.; Pletcher, M.J. Abstract 21042: Cardiovascular Risk Stratification Using Off-the-Shelf Wearables and a Multi-Task Deep Learning Algorithm. Circulation 2017, 136, A21042. [Google Scholar] [CrossRef]
  55. Banerjee, R.; Choudhury, A.D.; Sinha, A.; Visvanathan, A. HeartSense: Smart phones to estimate blood pressure from photoplethysmography. In Proceedings of the 12th ACM Conference on Embedded Network Sensor Systems, Memphis, TN, USA, 3–6 November 2014. [Google Scholar]
  56. Ye, C.; Fu, T.; Hao, S.; Zhang, Y.; Wang, O.; Jin, B.; Xia, M.; Liu, M.; Zhou, X.; Wu, Q.; et al. Prediction of Incident Hypertension Within the Next Year: Prospective Study Using Statewide Electronic Health Records and Machine Learning. J. Med. Internet Res. 2018, 20, e22. [Google Scholar] [CrossRef]
  57. Kanegae, H.; Suzuki, K.; Fukatani, K.; Ito, T.; Harada, N.; Kario, K. Highly precise risk prediction model for new-onset hypertension using artificial intelligence techniques. J. Clin. Hypertens. 2020, 22, 445–450. [Google Scholar] [CrossRef]
  58. Golino, H.F.; Amaral, L.S.; Duarte, S.F.; Gomes, C.M.; Soares Tde, J.; Dos Reis, L.A.; Santos, J. Predicting increased blood pressure using machine learning. J. Obes. 2014, 2014, 637635. [Google Scholar] [CrossRef]
  59. Huang, S.; Xu, Y.; Yue, L.; Wei, S.; Liu, L.; Gan, X.; Zhou, S.; Nie, S. Evaluating the risk of hypertension using an artificial neural network method in rural residents over the age of 35 years in a Chinese area. Hypertens. Res. 2010, 33, 722–726. [Google Scholar] [CrossRef]
  60. Held, E.; Cape, J.; Tintle, N. Comparing machine learning and logistic regression methods for predicting hypertension using a combination of gene expression and next-generation sequencing data. BMC Proc. 2016, 10, 141–145. [Google Scholar] [CrossRef]
  61. Li, Y.H.; Zhang, G.G.; Wang, N. Systematic Characterization and Prediction of Human Hypertension Genes. Hypertension 2017, 69, 349–355. [Google Scholar] [CrossRef]
  62. Pei, Z.; Liu, J.; Liu, M.; Zhou, W.; Yan, P.; Wen, S.; Chen, Y. Risk-Predicting Model for Incident of Essential Hypertension Based on Environmental and Genetic Factors with Support Vector Machine. Interdiscip. Sci. 2018, 10, 126–130. [Google Scholar] [CrossRef]
  63. Maxwell, A.; Li, R.; Yang, B.; Weng, H.; Ou, A.; Hong, H.; Zhou, Z.; Gong, P.; Zhang, C. Deep learning architectures for multi-label classification of intelligent health risk prediction. BMC Bioinform. 2017, 18, 523. [Google Scholar] [CrossRef]
  64. Sakr, S.; Elshawi, R.; Ahmed, A.; Qureshi, W.T.; Brawner, C.; Keteyian, S.; Blaha, M.J.; Al-Mallah, M.H. Using machine learning on cardiorespiratory fitness data for predicting hypertension: The Henry Ford ExercIse Testing (FIT) Project. PLoS ONE 2018, 13, e0195344. [Google Scholar] [CrossRef] [Green Version]
  65. Fernandes, M.; Olde Rikkert, M.G.M. The new US and European guidelines in hypertension: A multi-dimensional analysis. Contemp. Clin. Trials 2019, 81, 44–54. [Google Scholar] [CrossRef]
  66. McCormack, T.; Boffa, R.J.; Jones, N.R.; Carville, S.; McManus, R.J. The 2018 ESC/ESH hypertension guideline and the 2019 NICE hypertension guideline, how and why they differ. Eur. Heart J. 2019, 40, 3456–3458. [Google Scholar] [CrossRef]
  67. Diciolla, M.; Binetti, G.; Di Noia, T.; Pesce, F.; Schena, F.P.; Vagane, A.M.; Bjorneklett, R.; Suzuki, H.; Tomino, Y.; Naso, D. Patient classification and outcome prediction in IgA nephropathy. Comput. Biol. Med. 2015, 66, 278–286. [Google Scholar] [CrossRef]
  68. Lafrenière, D.; Zulkernine, F.H.; Barber, D.; Martin, K. Using machine learning to predict hypertension from a clinical dataset. In Proceedings of the IEEE Symposium Series on Computational Intelligence (SSCI), Athens, Greece, 6–9 December 2016; pp. 1–7. [Google Scholar]
  69. Hermida, R.C.; Smolensky, M.H.; Ayala, D.E.; Portaluppi, F. Ambulatory Blood Pressure Monitoring (ABPM) as the reference standard for diagnosis of hypertension and assessment of vascular risk in adults. Chronobiol. Int. 2015, 32, 1329–1342. [Google Scholar] [CrossRef]
  70. Pierdomenico, S.D.; Cuccurullo, F. Prognostic value of white-coat and masked hypertension diagnosed by ambulatory monitoring in initially untreated subjects: An updated meta analysis. Am. J. Hypertens. 2011, 24, 52–58. [Google Scholar] [CrossRef]
  71. Asayama, K.; Thijs, L.; Li, Y.; Gu, Y.M.; Hara, A.; Liu, Y.P.; Zhang, Z.; Wei, F.F.; Lujambio, I.; Mena, L.J.; et al. Setting thresholds to varying blood pressure monitoring intervals differentially affects risk estimates associated with white-coat and masked hypertension in the population. Hypertension 2014, 64, 935–942. [Google Scholar] [CrossRef]
  72. Poplin, R.; Varadarajan, A.V.; Blumer, K.; Liu, Y.; McConnell, M.V.; Corrado, G.S.; Peng, L.; Webster, D.R. Prediction of cardiovascular risk factors from retinal fundus photographs via deep learning. Nat. Biomed. Eng. 2018, 2, 158–164. [Google Scholar] [CrossRef]
  73. Persell, S.D.; Peprah, Y.A.; Lipiszko, D.; Lee, J.Y.; Li, J.J.; Ciolino, J.D.; Karmali, K.N.; Sato, H. Effect of Home Blood Pressure Monitoring via a Smartphone Hypertension Coaching Application or Tracking Application on Adults With Uncontrolled Hypertension: A Randomized Clinical Trial. JAMA Netw. Open 2020, 3, e200255. [Google Scholar] [CrossRef]
  74. Williams, B.; Mancia, G.; Spiering, W.; Agabiti Rosei, E.; Azizi, M.; Burnier, M.; Clement, D.L.; Coca, A.; de Simone, G.; Dominiczak, A.; et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension. Eur. Heart J. 2018, 39, 3021–3104. [Google Scholar] [CrossRef]
  75. Koren, G.; Nordon, G.; Radinsky, K.; Shalev, V. Machine learning of big data in gaining insight into successful treatment of hypertension. Pharmacol. Res. Perspect. 2018, 6, e00396. [Google Scholar] [CrossRef] [Green Version]
  76. Duan, T.; Rajpurkar, P.; Laird, D.; Ng, A.Y.; Basu, S. Clinical Value of Predicting Individual Treatment Effects for Intensive Blood Pressure Therapy. Circ. Cardiovasc. Qual. Outcomes 2019, 12, e005010. [Google Scholar] [CrossRef]
  77. Ye, X.; Zeng, Q.T.; Facelli, J.C.; Brixner, D.I.; Conway, M.; Bray, B.E. Predicting Optimal Hypertension Treatment Pathways Using Recurrent Neural Networks. Int. J. Med. Inform. 2020, 139, 104122. [Google Scholar] [CrossRef]
  78. da Silva, V.J.; da Silva Souza, V.; Guimaraes da Cruz, R.; Mesquita Vidal Martinez de Lucena, J.; Jazdi, N.; Ferreira de Lucena Junior, V. Commercial Devices-Based System Designed to Improve the Treatment Adherence of Hypertensive Patients. Sensors 2019, 19, 4539. [Google Scholar] [CrossRef]
  79. Matthews, H.; Hanison, J.; Nirmalan, N. “Omics”-Informed Drug and Biomarker Discovery: Opportunities, Challenges and Future Perspectives. Proteomes 2016, 4, 28. [Google Scholar] [CrossRef]
  80. Monte, A.A.; Vasiliou, V.; Heard, K.J. Omics Screening for Pharmaceutical Efficacy and Safety in Clinical Practice. J. Pharm. Pharm. 2012, S5, 001. [Google Scholar] [CrossRef]
  81. Unger, T.; Borghi, C.; Charchar, F.; Khan, N.A.; Poulter, N.R.; Prabhakaran, D.; Ramirez, A.; Schlaich, M.; Stergiou, G.S.; Tomaszewski, M.; et al. 2020 International Society of Hypertension global hypertension practice guidelines. J. Hypertens. 2020, 38, 982–1004. [Google Scholar] [CrossRef]
  82. Chang, W.; Liu, Y.; Xiao, Y.; Yuan, X.; Xu, X.; Zhang, S.; Zhou, S. A Machine-Learning-Based Prediction Method for Hypertension Outcomes Based on Medical Data. Diagnostics 2019, 9, 178. [Google Scholar] [CrossRef]
  83. SCORE2-OP Working Group; ESC Cardiovascular Risk Collaboration. SCORE2-OP risk prediction algorithms: Estimating incident cardiovascular event risk in older persons in four geographical risk regions. Eur. Heart J. 2021, 42, 2455–2467. [Google Scholar] [CrossRef]
  84. Crea, F. The new SCORE2 risk prediction algorithms and the growing challenge of risk factors not captured by traditional risk scores. Eur. Heart J. 2021, 42, 2403–2407. [Google Scholar] [CrossRef]
  85. Conroy, R.M.; Pyorala, K.; Fitzgerald, A.P.; Sans, S.; Menotti, A.; De Backer, G.; De Bacquer, D.; Ducimetiere, P.; Jousilahti, P.; Keil, U.; et al. Estimation of ten-year risk of fatal cardiovascular disease in Europe: The SCORE project. Eur. Heart J. 2003, 24, 987–1003. [Google Scholar] [CrossRef] [Green Version]
  86. Sabovcik, F.; Ntalianis, E.; Cauwenberghs, N.; Kuznetsova, T. Improving predictive performance in incident heart failure using machine learning and multi-center data. Front. Cardiovasc. Med. 2022, 9, 1011071. [Google Scholar] [CrossRef]
  87. Diller, G.P.; Kempny, A.; Babu-Narayan, S.V.; Henrichs, M.; Brida, M.; Uebing, A.; Lammers, A.E.; Baumgartner, H.; Li, W.; Wort, S.J.; et al. Machine learning algorithms estimating prognosis and guiding therapy in adult congenital heart disease: Data from a single tertiary centre including 10 019 patients. Eur. Heart J. 2019, 40, 1069–1077. [Google Scholar] [CrossRef]
  88. Santhanam, P.; Ahima, R.S. Machine learning and blood pressure. J. Clin. Hypertens. 2019, 21, 1735–1737. [Google Scholar] [CrossRef]
  89. Wu, X.; Yuan, X.; Wang, W.; Liu, K.; Qin, Y.; Sun, X.; Ma, W.; Zou, Y.; Zhang, H.; Zhou, X.; et al. Value of a Machine Learning Approach for Predicting Clinical Outcomes in Young Patients With Hypertension. Hypertension 2020, 75, 1271–1278. [Google Scholar] [CrossRef]
  90. Huan, T.; Meng, Q.; Saleh, M.A.; Norlander, A.E.; Joehanes, R.; Zhu, J.; Chen, B.H.; Zhang, B.; Johnson, A.D.; Ying, S.; et al. Integrative network analysis reveals molecular mechanisms of blood pressure regulation. Mol. Syst. Biol. 2015, 11, 799. [Google Scholar] [CrossRef]
  91. Srivastava, P.; Srivastava, A.; Burande, A.; Khandelwal, A. A Note on Hypertension Classification Scheme and Soft Computing Decision Making System. ISRN Biomath. 2013, 2013, 342970. [Google Scholar] [CrossRef]
  92. Rimoldi, S.F.; Scherrer, U.; Messerli, F.H. Secondary arterial hypertension: When, who, and how to screen? Eur. Heart J. 2014, 35, 1245–1254. [Google Scholar] [CrossRef]
  93. Arar, M.Y.; Hogg, R.J.; Arant, B.S., Jr.; Seikaly, M.G. Etiology of sustained hypertension in children in the southwestern United States. Pediatr. Nephrol. 1994, 8, 186–189. [Google Scholar] [CrossRef]
  94. Pedrosa, R.P.; Drager, L.F.; Gonzaga, C.C.; Sousa, M.G.; de Paula, L.K.; Amaro, A.C.; Amodeo, C.; Bortolotto, L.A.; Krieger, E.M.; Bradley, T.D.; et al. Obstructive sleep apnea: The most common secondary cause of hypertension associated with resistant hypertension. Hypertension 2011, 58, 811–817. [Google Scholar] [CrossRef]
  95. Reel, P.S.; Reel, S.; van Kralingen, J.C.; Langton, K.; Lang, K.; Erlic, Z.; Larsen, C.K.; Amar, L.; Pamporaki, C.; Mulatero, P.; et al. Machine learning for classification of hypertension subtypes using multi-omics: A multi-centre, retrospective, data-driven study. EBioMedicine 2022, 84, 104276. [Google Scholar] [CrossRef]
  96. Miller, D.D. Machine Intelligence in Cardiovascular Medicine. Cardiol. Rev. 2020, 28, 53–64. [Google Scholar] [CrossRef]
  97. Chen, J.; Qian, L.; Urakov, T.; Gu, W.; Liang, L. Adversarial Robustness Study of Convolutional Neural Network for Lumbar Disk Shape Reconstruction from MR images. SPIE Med. Imaging Image Process. 2021, 11596, 1159615. [Google Scholar]
  98. D’Angelo, G.; Della-Morte, D.; Pastore, D.; Donadel, G.; De Stefano, A.; Palmieri, F. Identifying patterns in multiple biomarkers to diagnose diabetic foot using an explainable genetic programming-based approach. Future Gener. Comput. Syst. 2023, 140, 138–150. [Google Scholar] [CrossRef]
  99. Goldstein, A.; Kapelner, A.; Bleich, J.; Pitkin, E. Peeking inside the black box: Visualizing statistical learning with plots of individual conditional expectation. J. Comput. Graph. Stat. 2015, 24, 44–65. [Google Scholar] [CrossRef]
  100. Petch, J.; Di, S.; Nelson, W. Opening the Black Box: The Promise and Limitations of Explainable Machine Learning in Cardiology. Can. J. Cardiol. 2022, 38, 204–213. [Google Scholar] [CrossRef]
  101. Tat, E.; Bhatt, D.L.; Rabbat, M.G. Addressing bias: Artificial intelligence in cardiovascular medicine. Lancet Digit. Health 2020, 2, e635–e636. [Google Scholar] [CrossRef]
  102. Krittanawong, C.; Bomback, A.S.; Baber, U.; Bangalore, S.; Messerli, F.H.; Wilson Tang, W.H. Future Direction for Using Artificial Intelligence to Predict and Manage Hypertension. Curr. Hypertens. Rep. 2018, 20, 75. [Google Scholar] [CrossRef]
  103. Chen, S.; Ji, Z.; Wu, H.; Xu, Y. A Non-Invasive Continuous Blood Pressure Estimation Approach Based on Machine Learning. Sensors 2019, 19, 2585. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The typical ML workflow in healthcare research.
Figure 1. The typical ML workflow in healthcare research.
Jcdd 10 00074 g001
Figure 2. Block diagram of the blood pressure estimation process using ML techniques. In detail, the raw signals are prepared through normalization, the correction of baseline wandering due to respiration, and finally, signal filtration. Specifically, to construct a dataset for BP estimation models, it is necessary to accurately extract the features of the original waveform (and underlying demographic and statistical data) and select effective features, improving the generalization and reducing the risk of overfitting the algorithms. PPG: photoplethysmograph; ML: machine learning.
Figure 2. Block diagram of the blood pressure estimation process using ML techniques. In detail, the raw signals are prepared through normalization, the correction of baseline wandering due to respiration, and finally, signal filtration. Specifically, to construct a dataset for BP estimation models, it is necessary to accurately extract the features of the original waveform (and underlying demographic and statistical data) and select effective features, improving the generalization and reducing the risk of overfitting the algorithms. PPG: photoplethysmograph; ML: machine learning.
Jcdd 10 00074 g002
Table 1. AI application in hypertension management.
Table 1. AI application in hypertension management.
ApplicationsBenefits
Measuring BPEstimate BP by analyzing PPG signal with ML and DL algorithms.Self-monitoring BP for hypertension
Predicting AH developmentPredict the risk of developing AH by using genetics, medical data, and behavioral, environmental, and socioeconomic factors.Timely intervention
Diagnosing AHAccurately diagnosing AH by using CV risk factors, anthropometric data, vital signs, and laboratory data.Precision diagnosis
Predicting AH treatment successIdentify factors contributing to treatment success.Personalized treatment plan
Predicting AH prognosisStratify patients and predict CV outcomes.Treatment plan adjustment
AI: artificial intelligence; BP: blood pressure; PPG: photoplethysmograph; ML: machine learning; DL: deep learning; AH: arterial hypertension; CV: cardiovascular.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Visco, V.; Izzo, C.; Mancusi, C.; Rispoli, A.; Tedeschi, M.; Virtuoso, N.; Giano, A.; Gioia, R.; Melfi, A.; Serio, B.; et al. Artificial Intelligence in Hypertension Management: An Ace up Your Sleeve. J. Cardiovasc. Dev. Dis. 2023, 10, 74. https://doi.org/10.3390/jcdd10020074

AMA Style

Visco V, Izzo C, Mancusi C, Rispoli A, Tedeschi M, Virtuoso N, Giano A, Gioia R, Melfi A, Serio B, et al. Artificial Intelligence in Hypertension Management: An Ace up Your Sleeve. Journal of Cardiovascular Development and Disease. 2023; 10(2):74. https://doi.org/10.3390/jcdd10020074

Chicago/Turabian Style

Visco, Valeria, Carmine Izzo, Costantino Mancusi, Antonella Rispoli, Michele Tedeschi, Nicola Virtuoso, Angelo Giano, Renato Gioia, Americo Melfi, Bianca Serio, and et al. 2023. "Artificial Intelligence in Hypertension Management: An Ace up Your Sleeve" Journal of Cardiovascular Development and Disease 10, no. 2: 74. https://doi.org/10.3390/jcdd10020074

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

Article Metrics

Back to TopTop