IMR Press / FBL / Volume 28 / Issue 10 / DOI: 10.31083/j.fbl2810248
Open Access Review
A Pharmaceutical Paradigm for Cardiovascular Composite Risk Assessment Using Novel Radiogenomics Risk Predictors in Precision Explainable Artificial Intelligence Framework: Clinical Trial Tool
Show Less
1 Department of Radiology, Azienda Ospedaliero Universitaria, 40138 Cagliari, Italy
2 Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
3 School of Bioengineering Research and Sciences, Maharashtra Institute of Technology's Art, Design and Technology University, 412021 Pune, India
4 Department of Cardiology, Indraprastha APOLLO Hospitals, 110001 New Delhi, India
5 Department of Medicine, Division of Cardiology, Queen's University, Kingston, ON K7L 1C2, Canada
6 Department of Medicine, Division of Cardiology, University of Toronto, Toronto, ON M5S 1A1, Canada
7 Heart and Vascular Institute, Adventist Health St. Helena, St Helena, CA 94574, USA
8 Department of Vascular Surgery, Central Clinic of Athens, 11526 Athens, Greece
9 Invasive Cardiology Division, University of Szeged, 6725 Szeged, Hungary
10 Department of Radiology, Harvard Medical School, Boston, MA 02118, USA
11 Department of Vascular Surgery, University of Lisbon, 1600-209 Lisbon, Portugal
12 Department of Neurology & Stroke Program, University of Maryland, Baltimore, MD 21201, USA
13 Vascular Screening and Diagnostic Centre and University of Nicosia Medical School, 2368 Agios Dometios, Cyprus
14 Nephrology Department, Kaiser Permanente, Sacramento, CA 95823, USA
15 Department of Food Science and Technology, Graphic Era Deemed to be University, Dehradun, 248002 Uttarakhand, India
16 Department of Electrical and Computer Engineering, Idaho State University, Pocatello, ID 83209, USA
17 Department of Radiobiology and Molecular Genetics, National Institute of The Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
18 Allergy, Clinical Immunology and Rheumatology Institute, Toronto, ON L4Z 4C4, Canada
19 MV Diabetes Centre, Royapuram, Chennai, 600013 Tamil Nadu, India
20 Department of Computer Engineering, Graphic Era Deemed to be University, 248002 Dehradun, India
*Correspondence: jasjit.suri@atheropoint.com (Jasjit S. Suri)
Front. Biosci. (Landmark Ed) 2023, 28(10), 248; https://doi.org/10.31083/j.fbl2810248
Submitted: 10 May 2023 | Revised: 2 August 2023 | Accepted: 28 August 2023 | Published: 19 October 2023
(This article belongs to the Special Issue Explainable Artificial Intelligence in Biomedicine)
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Cardiovascular disease (CVD) is challenging to diagnose and treat since symptoms appear late during the progression of atherosclerosis. Conventional risk factors alone are not always sufficient to properly categorize at-risk patients, and clinical risk scores are inadequate in predicting cardiac events. Integrating genomic-based biomarkers (GBBM) found in plasma/serum samples with novel non-invasive radiomics-based biomarkers (RBBM) such as plaque area, plaque burden, and maximum plaque height can improve composite CVD risk prediction in the pharmaceutical paradigm. These biomarkers consider several pathways involved in the pathophysiology of atherosclerosis disease leading to CVD. Objective: This review proposes two hypotheses: (i) The composite biomarkers are strongly correlated and can be used to detect the severity of CVD/Stroke precisely, and (ii) an explainable artificial intelligence (XAI)-based composite risk CVD/Stroke model with survival analysis using deep learning (DL) can predict in preventive, precision, and personalized (aiP3) framework benefiting the pharmaceutical paradigm. Method: The PRISMA search technique resulted in 214 studies assessing composite biomarkers using radiogenomics for CVD/Stroke. The study presents a XAI model using AtheroEdgeTM 4.0 to determine the risk of CVD/Stroke in the pharmaceutical framework using the radiogenomics biomarkers. Conclusions: Our observations suggest that the composite CVD risk biomarkers using radiogenomics provide a new dimension to CVD/Stroke risk assessment. The proposed review suggests a unique, unbiased, and XAI model based on AtheroEdgeTM 4.0 that can predict the composite risk of CVD/Stroke using radiogenomics in the pharmaceutical paradigm.

Keywords
cardiovascular disease
stroke
biomarkers
radiomics
genomics
deep learning
bias
pruning
cloud
multicenter
pharmaceutical
explainable artificial intelligence
1. Introduction

Cardiovascular diseases (CVD) kill 17.3 million people each year. This figure is predicted to increase to 23.6 million by 2030 [1, 2]. By 2030, this will cost $920 billion in direct healthcare costs, a challenge for the public health system [3]. Among the various types of CVD, coronary heart disease (CHD) is the leading cause. This disease is due to atherosclerosis, the chronic inflammatory condition of the artery wall and one of the most common causes of death [4, 5]. CVD is caused by many things, including genetic, metabolomic, environmental, behavioural, and lifestyle factors [6, 7]. Although traditional risk factors such as age, gender, high cholesterol, high blood pressure [8], smoking, and comorbidities like diabetes mellitus [9], obesity [10], and hypertension [11, 12] are commonly used risk factors for predicting CVD risk, laboratory-based biomarkers are not always feasible, particularly in developing countries with resource constraints [13, 14]. Also, most scoring systems for CVD risk were made for Caucasians, and their validity and usefulness in other ethnic groups are still unknown [15]. As a result, CVD risk is frequently misdiagnosed. Although patients gain in some ways, it lacks the ethnic effect of medication design as well as the benefits of tailored therapy due to genomes and proteomics [16, 17].

Furthermore, the current design of the risk assessment tool lacks CVD risk stratification due to the non-linear behavior caused by the amalgamation of all the risk predictors [18]. However, the benefit to the drug industry in medication design has not been realized due to the absence of artificial intelligence (AI) methodologies, which leads to CVD risk stratification with different grades [19]. So, to achieve the above objectives, one certainly needs AI strategies for CVD risk assessment, particularly in big data or large cohorts [20, 21]. This requires composite CVD risk designs that can operate in batch (large cohort side) mode, sometimes called Pharma (Pharmaceutical) Mode, for automatically processing a large set of patients [22]. Previous studies conducted clinical trials but lacked AI-based solutions [22].

Further, it did not consider the composite risk of CVD stratification [23, 24]. Studies that did CVD also lack AI-based solutions [25]. Thus, there is a need for AI-based CVD composite risk in large cohorts for pharmaceutical companies to benefit from their drug design [26].

The Precision Medicine Initiative (PMI), introduced by former US President Obama in 2015, can potentially reduce the worldwide burden of CVD by mobilizing collective resources and expertise to develop and spread knowledge [27]. The precision medicine approach makes it possible to start treatments earlier, use more advanced diagnostics, and make treatments more personalized and affordable [28]. The precision medicine approach, with the help of AI, can improve symptom-driven care by proactively combining multi-omics assessments with clinical [28], imaging [29, 30], epidemiological [31], and demographic variables [32].

Precision medicine allows for earlier treatments for advanced diagnostics and tailoring better and more affordable personal treatment [29, 33, 34]. Advanced data analysis techniques are being employed more and more to aid in the development of successful CVD treatments [35]. Batch mode processing allows large data sets to be analyzed at once, facilitating a more thorough examination of patient risk factors and comorbidities to determine the best course of treatment [36]. Additionally, composite CVD risk stratification considers the combined effect of multiple risk factors to create a more comprehensive assessment of a patient’s risk [37]. Finally, the ability to categorize patients into different risk classes ranging from low to high allows for more personalized and effective treatment approaches to be developed based on a patient’s risk profile [38]. These tools provide valuable support to healthcare providers in managing CVD and improving patient outcomes. In this paragraph, we have to first give the spirit of using DL due to the success of DL in healthcare [39]. As a result, we propose that DL can be adapted for routine mode and pharmacological models for assessing CVD/Stroke risk.

This study proposes deep learning (DL) to stratify the risk of CVD/Stroke using radiomics-based biomarker (RBBM) and genomics-based biomarker (GBBM) as covariates in pharmaceutical mode. The study also investigates pruned or compacted AI systems for CVD risk using multi-omics data and uses an explainability model to illuminate AI’s “Black Box Nature” [40]. Fig. 1 depicts the explainable AI (XAI) model integration of various CVD biomarkers, including (i) office-based biomarkers (OBBM), such as age, gender, ethnicity, BMI, hypertension, and smoking, (ii) laboratory-based biomarkers (LBBM), such as LDL, HDL, triglycerides, eGFR, and ESR, (iii) RBBM, such as plaque burden, cIMT, PA, and maximum each patient tries to help clinicians figure out how personalized medical information differences might affect health, so they can diagnose and plan the best way to treat the patient [41].

Fig. 1.

An overview of composite biomarkers using the XAI model for CVD risk stratification in the pharmaceutical domain (Original image, AtheroPoint™ LLC, Roseville, CA, USA).

Finally, a cloud-based framework is implemented to make the results clinically explainable [42] and accessible through telemedicine [43, 44]. The presented study aims to reduce bias, increase compression, and achieve aiP3 goals using the composite risk radio genomics model. The presented model discusses reliability, stability, and survival analysis by AtheroPoint 4.0™, Roseville, CA, USA, for CVD risk stratification.

2. Search Strategy

The search methodology employed the PRISMA framework, illustrated in Fig. 2; we employed various keywords to search for articles such as “cardiovascular disease”, “stroke”, “CVD”, “genomics and CVD”, “radiomics and CVD and pharmamode”, “radiomics and stroke”, “genomics and stroke batch mode”, “prevention medicine and clinical trial”, “preventive medicine and CVD”, “personalized medicine and artificial intelligence”, “atherosclerotic in genomics”, “radiomics and AI”, “genomics and radiomics”. Out of these 698 articles, 326 publications were considered, while 289 studies were excluded based on three criteria: unrelated research, irrelevant papers, and inadequate data. These criteria led to eliminating 289, 174, and 21 studies, as indicated by E1, E2, and E3 (Fig. 2), respectively, resulting in a final assessment of 214 studies. Our focus was solely on articles that linked CVD/Stroke with radiomics and GBBM. Studies investigating diseases, such as Parkinson’s, cancer, and diabetes, were not considered.

Fig. 2.

PRISMA model for study selection.

The omitted E1 category included 289 studies unrelated to CVD/Stroke or RBBM/GBBM. The E2 category contained 174 papers that either needed more information to be included in our analysis or needed to show a link between RBBM/GBBM and CVD/Stroke. Additionally, these studies lacked selectable AI and CVD/Stroke characteristics for analysis that could be utilized for CVD/stroke risk stratification. Finally, we found 27 research studies with inadequate data sets designated as E3 in the PRISMA model.

3. Radiomics-Based CVD Risk Stratification in Pharmaceutical Mode

Various radiological methods have been invented and widely used in recent years to rule out and/or identify preclinical atherosclerotic-based CVD to advise optimal prophylactic therapy. Since the carotid artery can be used for the prediction of coronary artery disease [45, 46, 47, 48], thus the most commonly used imaging modalities for its screening are Magnetic Resonance Imaging (MRI) [49, 50, 51], Computed Tomography Angiography (CTA) [52, 53, 54, 55, 56], Optical Coherence Tomography (OCT) [57], and Ultrasound (US) [58, 59]. The US is the most common, user-friendly, cost-effective, high-resolution, non-invasive image acquisition modality capable of imaging and recognizing atherosclerotic plaque [58, 60, 61].

3.1 Non-AI-Based Radiomics and CVD Risk Stratification

Plaque segmentation attempts to identify plaques precisely and consistently to measure plaque burden and track progression over time. This can be used to diagnose and plan treatment for CVD patients. The segmentation results can be utilized to quantitatively analyze plaque features such as volume, shape, and composition. Regression and progression of the plaque in over 10,000 patients in a few hours retrospectively. Fig. 3 shows the carotid ultrasound image phenotype (CUSIP) parameters derived from carotid artery scans representing the surrogate makers for coronary atherosclerotic disease.

Fig. 3.

Carotid artery scans show plaque in the far wall of the carotid artery. Carotid ultrasound image phenotype (CUSIP) consists of carotid intima-media thickness (IMT, ave., max., min.) and total plaque area (TPA). The shaded yellow region corresponds to the region of plaque burden. (A) Moderate plaque burden in carotid bulb; (B) Moderate plaque burden in common carotid artery; (C) Moderate-High plaque burden in carotid blub artery; (D) High plaque burden in carotid blub artery; and (E) High plaque burden in carotid blub artery. LCB, left carotid bulb; RCB, right carotid bulb (Original image, AtheroPointTM LLC, Roseville, CA, USA).

The segmentation process can be challenging due to the variability in plaque appearance and the presence of other structures in the images. A combination of multiple methods can be used to improve the accuracy and reliability of plaque segmentation as part of the radiomics features. Carotid ultrasound image phenotypes (CUSIP) have been tried before [62, 63, 64]. Therefore, it offers a wide range of applications for regular proactive monitoring of atherosclerotic plaque for CVD risk assessment [65, 66, 67, 68, 69, 70]. The statistical reports in the Pharma Mode shown in Fig. 4 will give the regression or progression of the plaque based on the statins during the 5–10-year follow-up of thousands of patients. The surrogate carotid artery disease and genetic-based biomarkers gathered from blood plasma will be used to track the atherosclerotic disease of the coronary artery disease. In addition, statistical regression plots will be constructed to link CVD risk to other clinical biomarkers.

Fig. 4.

GUI view of AtheroEdge™ 2.0, showing carotid artery plaque measurement. (A) Pharma Mode selection for selection of the image resolution. (B) CUSIP parameters showing cIMT and TPA measurements for low plaque patient. (C) CUSIP parameters (cIMT/TPA) measurements for the high-plaque patient. (D) Pharma Mode report showing the CUSIP parameters and composite CVD risk (marked in color code). This includes (i) renal effect on CVD risk via estimated glomerular filtration rate (eGFR) biomarkers, (ii) arthritis effect on CVD using erythrocyte sedimentation rate (ESR) biomarkers, (iii) diabetic effect on CVD using HbA1c, (iv) changes in CVD due to low-density lipoprotein (LDL) (Original image, AtheroPoint™ LLC, Roseville, CA, USA).

3.2 AI-Based Radiomics and CVD Risk Stratification

AI-based radiomics techniques have been recently used to extract and analyze segmented features from the carotid artery based on carotid ultrasound imaging modalities [30, 71]. The studies have used different DL algorithms, including convolutional neural networks (CNN), UNet, UNet+, and DenseNet, along with other machine learning (ML) algorithms such as support vector machine (SVM), random forest (RF) and logistic regression (LR) for classification. An example of AtheroEdge™ 3.0 in Pharma mode is seen in Fig. 5A. On clicking the “Pharma Mode” feature of the system, the epidemiologist or physician conducting the Pharma trial will select the spreadsheet with 100,000 patients.

Fig. 5.

GUI view of AtheroEdge™ 3.0, AI-based Cardiovascular Disease Risk Stratification system in Pharma mode. (A) AI-based system in Pharma Mode with 70% (green) patient cohort processed. (B) AtheroEdge™ 3.0: An example of the results of the Pharma Mode trial. The color code shows the CVD risk class. The percentage risk is shown in the second column from the right. The rest of the columns are the CVD risk predictors (zoomed version of Fig. 5B is shown in the Supplementary Material) (Original image, AtheroPoint™ LLC, Roseville, CA, USA).

The patient data file will be loaded into the system and ready to select the trained AI model for CVD risk prediction. On clicking the “Trained Model” button, the user can fetch the trained AI model and the number of risk classes. Finally, on clicking the “Predict” button, the user AtheroEdge™ 3.0 system will compute the risk of all the 100,000 patients in, say, five seconds per patient. Fig. 5B depicts an example report. Table 1 (Ref. [30, 72, 73, 74, 75, 76, 77, 78, 79, 80]) also includes a list of the segment characteristics used in each investigation, such as carotid intima-media thickness (cMIT), carotid wall thickness (cWT), lumen diameter (LD), plaque load (PB), and plaque area (PA). Table 1 also displays the accuracy (ACC), area under the curve (AUC), and study conclusion for each algorithm. Saba et al. [72] used ML-based radiomics to assess intra/inter-observer variability in detecting the common carotid artery (CCA) using SVM and RF classifiers, achieving an ACC of 98.32%. Biswas et al. [30] used DL-based radiomics to detect joint cWT, and PB features with an accuracy of 93% and AUC of 0.89. Vila et al. [73] also used a DenseNet CNN model to detect and estimate the cIMT with an ACC of 96.45% and an AUC of 0.89. Jain et al. [74] used a UNet-based model to detect and segment the CCA plaque area (PA) with an ACC of 88% and an AUC of 0.91. In another study, Jain et al. [75] used UNet+ to detect and segment the PA in the internal carotid artery (ICA) with an ACC of 97% and AUC of 0.99%. Yuan et al. [76] used UNet-based radiomics to segment and analyze cMIT and plaque with an ACC of 97% and a Dice score of 83.3–85.7.

Table 1.Studies using DL-based radiomics (covariates) for segmented features using CCA.
SN Studies Year DS AS IM AI (ML/DL) AA Segment Features Performance Conclusion
Segmentation
1 Jain et al. [74] 2021 970 CCA US DL UNet, UNet+ PA ACC: 88%, AUC: 0.91 (p < 0.0001) Detection of PA and segmentation
2 Jain et al. [75] 2022 379 ICA US DL UNet, UNet+ PA AUC: 97%, AUC: 0.99 (p < 0.0001) Detection of PA and segmentation
3 Yuan et al. [76] 2022 115 CCA US DL UNet cMIT ACC: 97%, Dice 83.3–85.7 cMIT and Plaque segmentation
4 Gago et al. [78] 2022 8000 CCA US DL UNet PA, cMIT, and cWT estimation ACC: 79.00% Tissue characterization of plaque
5 Lainé et al. [80] 2022 2676 CCA US DL UNet cWT ACC: 86.00% Dilated U-net architecture is used for cWT
Classification
6 Saba et al. [72] 2018 100 CCA US ML SVM, RF LD ACC: 98.32% intra/inter-observer variability
7 Biswas et al. [30] 2020 250 CCA US DL CNN, LR cWT, PB cMIT error < 0.093 ± 0.0677 mm, AUC: 0.89 (p < 0.0001) Joint detection cWT and PB
8 Vila et al. [73] 2020 8000 CCA US DL CNN (Dense Net) cIMT ACC: 96.45%, AUC: 0.89 (p < 0.0001) Plaque detection and cIMT estimation
9 Molinari et al. [77] 2022 500 CCA US DL CNN cMIT and cWT ACC: 95.6%, AUC:0.83 (p < 0.0001) cMIT and cWT measurement
10 Shin et al. [79] 2022 1440 CCA US DL CNN Plaque viscous index ACC: 83.00%, AUC: 0.87 (p < 0.0001) Viscoelasticity index

SN, Serial number; DS, Data size; AS, Artery Segment; IM, Imaging modality; CCA, Carotid artery; COA, Coronary artery; CLA, Classifier; FE, Feature extraction; IVUS, Intra-vascular ultrasound; US, Ultrasound; cWT, Carotid wall thickness; cMIT, Carotid intima-media thickness; LD, Lumen diameter; PB, Plaque burden; PA, Plaque area; CCA, Common carotid artery; ICA, Internal carotid artery; ACC, Accuracy; AUC, Area under the curve; AI, Artificial Intelligence; AA, AI Algorithms; ML, Machine Learning; DL, Deep Learning; HDL, Hybrid Deep Learning; SVM, Support vector machine; RF, Random Forest; DT, Decision Tree; CNN, Convolution neural network; LR, Logistic regression; NR, Not Reported.

Molinari et al. [77] used a CNN-based radiomics model to analyze cMIT and cWT features, achieving an ACC of 95.6% and an AUC of 0.83. UNet-based tissue characterization of CCA plaque by Gago et al. [78] yielded an ACC of 79.00%. Shin et al. [79] used a CNN-based model to analyze the viscous plaque index with an ACC of 83% and an AUC of 0.87, while Lainé et al. [80] segmented the cWT with an ACC of 86% using a dilated U-Net architecture.

4. Genomics-Based CVD Risk Stratification in Pharmaceutical Model

Genetic factors play a significant role in the development and progression of plaque buildup in the arteries, and understanding an individual’s genetic makeup can provide valuable insights into their risk for CVD. Genetic data is used to identify potential treatment and prevention strategies targets and can help personalize care for individual patients. To accurately assess and understand the plaque burden in an individual, it is essential to have access to genetic data. This section highlights conventional risk scores and AI-based composite risk score methods for the personalized predication of CVD.

4.1 Conventional Polygenic Risk Score and CVD Risk Stratification

A polygenic risk score, often known as a “polygenic risk score” or “PRS”, is a prediction tool that assesses an individual’s genetic predisposition to various diseases, such as CVD [81]. The PRS is estimated by taking into account the presence and frequency of genetic variants associated with the disease [82, 83].

SNPs associated with CVD can be used as markers to establish an individual’s genetic profile [84]. Individual genotyping data is collected after SNP identification. Large-scale genotyping initiatives or DNA tests can provide genotype data [85]. After that, each person’s PRS is calculated based on their genome’s risk allele count [86]. The CVD association of risk alleles influences their weight [81]. The “effect size” measures how much an SNP affects CVD risk [87]. This statistic typically measures the strength of the relationship between two variables. The odds ratio compares the risk of CVD for those with a particular SNP to those without [88]. The odds ratio usually quantifies the effect [82]. An individual’s PRS is calculated by summing the weights of all risk alleles found in their genotyping data [89]. Higher PRS scores indicate a higher CVD predisposition. Fig. 6 shows using the molecular data to calculate the PRS.

Fig. 6.

Molecular data used for prediction of PRS for CVD (Original image, AtheroPoint™ LLC, Roseville, CA, USA).

The PRS is not a diagnostic tool and should not be used alone to make clinical decisions [90]. The PRS should be used with family history, lifestyle, and other risk factors to make a more informed decision about CVD risk [91, 92]. Table 2 (Ref. [93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114]) lists In-vitro biomarkers that can be identified in the serum and plasma of asymptomatic and CVD patients. To improve diagnosis and CVD risk stratification, multivariate genomic-based biomarkers and multivariable prediction models have been used [93, 115, 116]. The prediction models assess multivariate atherosclerosis and coronary artery development indicators in circulating molecules. Cellular, biochemical, epigenetic, and/or transcriptional GBBM contributing to CVD are addressed below. ACS biomarkers include C-reactive protein (CRP) [94], a liver protein released in response to inflammation, which increases cardiovascular risk. Another study by Barwari et al. [95] and Barwari et al. [95] found plasma LDL-cholesterol levels to be a biomarker for ACS. LDL-cholesterol is considered “bad” because it causes arterial plaque. Joshi et al. [96] and Cristell et al. [97] noted that PCSK9 regulates cholesterol metabolism and that the identified variant is associated with higher LDL-cholesterol levels and an increased risk of cardiovascular events. Pedicino et al. [98] and Partida et al. [99] identified pro-inflammatory CD4+ cells with poor CD28 expression as a biomarker for ACS. Sato et al. [100] and Montone et al. [101] found that biomarkers of ACS include a drop in the number of regulatory T lymphocytes and their ability to stop cancer cells from growing. The following rows list plaque rupture biomarkers without systemic inflammation. Kelley et al. [102] found inflammasome activation, while Kreiner et al. [103], Toldo et al. [104], and Omerovic et al. [105] found interleukins 1 and 18 and emotional disturbance-induced catecholamine release as CVD biomarkers. Repova et al. [106] found neutrophil activity, while Dang et al. [107] found macrophages or T lymphocytes as ACS biomarkers, eroding plaque. In summary, Table 2 provides a comprehensive overview of different biomarkers associated with plaque progression in patients with CVD. The table highlights the complexity of CVD and the need for personalized treatment approaches based on individual patient characteristics.

Table 2.Studies show the genomics biomarkers responsible for plaque progression.
SN Author Disease Phenotype Biomarker
1 Thibaut et al. [93], Padua et al. [94] CVD common phenotype C-reactive protein (CRP)
2 Barwari et al. [95], Cristell et al. [97] CVD common phenotype Plasma levels of low low-density protein cholesterol (LDL-cholesterol)
3 Joshi et al. [96], Cristell et al. [97] CVD common phenotype PCSK9 variant
4 Pedicino et al. [98], Partida et al. [99] CVD systemic inflammation and plaque rupture CD4+ cells that are pro-inflammatory and have a poor expression of the co-stimulatory molecule CD28 on their cell surfaces
5 Sato et al. [100], Montone et al. [101] ACS systemic inflammation and plaque rupture a decrease in the number of circulating regulatory T lymphocytes as well as their inhibitory function
6 Montone et al. [101] CVD systemic inflammation and plaque rupture CD31 and protein tyrosine phosphatase N22, upstream regulatory mediators, modulate T-cell number and function.
7 Kelley et al. [102] CVD plaque rupture without systemic inflammation Activation of Inflammasome
8 Kreiner et al. [103] ACS plaque rupture without systemic inflammation Interleukin (IL)-1
9 Toldo et al. [104] CVD plaque rupture without systemic inflammation Interleukin (IL)-18
10 Omerovic et al. [105] CVD plaque rupture without systemic inflammation Catecholamine release due to emotional disturbance
11 Repova et al. [106] CVD erosion of plaque activation of Neutrophil
12 Dang et al. [107] CVD erosion of plaque Macrophages or T lymphocytes
13 Thota et al. [108] CVD erosion of plaque Proteoglycans
14 Wang et al. [109] ACS erosion of plaque Glycosaminoglycans
15 Worssam et al. [110] CVD erosion of plaque Arterial Smooth Muscle Cells (SMCs)
16 Romo et al. [111] CVD erosion of plaque Monocyte HYAL2 expression increased.
17 Pinheiro-de-Sousa et al. [112] CVD erosion of plaque up regulation of Endothelial cell CD44
18 Chong et al. [113] CVD plaque in the absence of thrombus Vasospasm
19 Shimokawa et al. [114] CVD plaque in the absence of thrombus Rho-kinase activity

ACS, Acute Coronary syndrome.

Radiogenomics Risk Predictors are valuable biomarkers used for CVD risk stratification, combining radiological imaging data and genetic information to assess an individual’s CVD risk [75, 117]. These predictors analyze quantitative features extracted from imaging modalities like MRI, CTA, OCT, and US, alongside genetic data, serving as specific indicators of pathological changes in the arteries [118]. Referred to as “RBBMs”, they are not mere symptoms but powerful tools for risk evaluation [119]. Numerous studies, detailed in Table 1, have explored and emphasized the significance of these risk predictors.

CAD affects major blood vessels supplying the heart muscle, primarily due to cholesterol deposits causing atherosclerosis, leading to reduced blood flow and potential heart attacks, angina, or strokes [120]. Symptoms of CVD may differ between genders, with men experiencing more chest pain and women often presenting shortness of breath, nausea, and extreme fatigue, indicating underlying heart or blood vessel issues [121]. Abnormal anatomical conditions in CVD, such as cardiomegaly, myocardial infarction, vasculitis [122], atherosclerosis, and thrombosis [123], are observable through medical imaging, physical examinations [124], or laboratory tests, providing critical insights into disease development. Studying these pathological changes is vital for understanding CVD and utilizing radiological biomarkers, to detect stenosis and other risk factors, contributing to a comprehensive comprehension of CVD and its related conditions.

4.2 AI Genomics-Based Composite Risk Score for CVD Risk Stratification

The medical imaging field has noted the progress made in ML and DL [125, 126, 127, 128]. Deep neural networks (DNNs), a subset of DL, work like the human brain [39, 117, 129]. AI has been used in recent studies to figure out the risk of CVD using RBBM [11, 12, 38, 130, 131, 132, 133, 134] and GBBM [33, 135, 136] frameworks. DL is becoming more popular because (i) it automatically extracts features [137], (ii) it can fuse with different ML configurations for classification [117, 138], (iii) it uses UNet and Hybrid UNet-based DL strategies for segmentation [43, 119], and (iv) Lastly, it gives more accurate segmentation and solo or ensemble-based classification because it can go through forward and backward propagation by reducing the loss using different kinds of loss functions [74].

4.2.1 Training and Prediction

The architecture consists of two halves. The left is the training subsystem, while the right is the prediction subsystem.

The DL training classifiers consist of one of the DL classifiers, namely, long short-term memory network (LSTM), recurrent neural network (RNN), gated recurrent units (GRU), bidirectional-LSMT (BiLSTM), bidirectional-RNN (BiRNN), and bidirectional-GRU (BiGRU) (presented in the following subsection).

Along with the DL classifier bank, there are supervised clinical risk labels representing ground truth (GT), such as heart failure (or high CVD risk) and stroke [130, 139]. This GT representing the CAD includes computed tomography (CT) coronary score [140] or quantification of CAD lesions using intravascular ultrasound (IVUS) [141, 142]. Several non-linear training-based approaches have been shown in heart disease risk stratification [12, 132, 134, 143]. Fig. 7 shows AI-based architecture for CVD risk assessment.

Fig. 7.

AI genomics-based architecture for CVD risk assessment (Original image, AtheroPoint™ LLC, Roseville, CA, USA).

4.2.2 Deep Learning Classifier Banks

The RNN [144], BiRNN [145], LSTM [146], BiLSTM [147], GRU [148], and BiGRU [149]models evaluate sequential data, such as Electrocardiograph (ECG) [148, 150], text [146], speech [151], localization of myocardial infraction [147], and handwriting [152, 153]. Table 3 (Ref. [154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168]) below summarises AI-based genomics studies that make a personalized and accurate prediction of CVD. A total of 15 studies are listed in the table and described with sample size, ground truth, technology, benchmark, source description, AI type, classifier type, cross-validation technique, and performance characteristics. In the first investigation, Davies et al. [154] (2010) tried to predict CVD using SNP data in conjunction with ML techniques using SVM and LR. The total number of participants in the study was 2333, and the ACC and SPE parameters each attained an AUC of 0.80 and 0.78, respectively. The study was prospective.

Table 3.AI-based genomics studies that predict CVD in a personalized and precision framework.
SN Studies Year SS GT BM SD Tech AI (ML/DL) CT CVP Performance parameters CTR
ACC SEN SPE AUC F1
1 Davies et al. [154] 2010 2333 CVD LBBM, OBBM SNP GWAS ML SVM, LR 10K 0.61, 0.55 NR NR 0.80, 0.78 NR Prospective
2 Valavanis et al. [155] 2010 63 CVD LBBM, OBBM DNA DEG DL PDM-ANN 3K 0.95 0.98 0.94 0.96 NR Retrospective
3 Beigi et al. [156] 2011 23 CVD LBBM, OBBM SNP Proteinase k ML SVM 10K 0.91 NR NR NR NR Prospective
4 Montañez et al. [157] 2017 12 CVD LBBM, OBBM SNP DEG ML SVM, RF, XGB 10K NR 0.88, 0.52, 0.70 0.86, 0.95, 0.82 0.90, 0.87, 0.85 NR Retrospective
5 Zhao et al. [158] 2019 490 CVD LBBM, OBBM SNP DEG DL CNN 10K 0.81 0.83 0.79 0.82 NR Prospective
6 Schiano et al. [159] 2021 443 DCM LBBM mRNA DEG ML DT 10K 0.79 NR NR NR NR Prospective
7 Yang et al. [160] 2021 117 CVD LBBM, OBBM SNP DEG ML SVM, RF 10K 0.86, 0.89 NR NR 0.91, 0.93 NR Retrospective
8 Akbarzadeh et al. [161] 2022 4756 CVD LBBM DNA Proteinase k ML SVM, DT, RF, LR 5K 0.72,0.73, 0.74, 0.70 0.66,0.66, 0.69, 0.67 0.78,0.80, 0.78,0.73 0.78, 0.77, 0.80, 0.77 NR Prospective
9 Kesar et al. [162] 2022 203 CVD LBBM, OBBM NR NR ML LR, XGB 10K NR NR NR 0.75, 0.76 NR Prospective
10 Pirruccello et al. [163] 2022 116 CVD LBBM, OBBM SNP DEG DL UNet 10K 0.96 NR NR NR NR Retrospective
11 Steinfeldt et al. [164] 2022 395713 CVD LBBM NR NR DL CNN 10K 0.78 NR 0.81 NR NR Prospective
12 Lee et al. [165] 2022 283 CVD LBBM SNP DEG ML LR 10K 0.81 0.79 0.83 NR NR Retrospective
13 Libiseller‑Egger et al. [166] 2022 34432 CVD LBBM, OBBM SNP GWAS DL SVM, LR 5K 0.76 0.81 0.84 0.81 NR Prospective
14 Kang et al. [167] 2023 17044 AMI LBBM mRNA and miRNA DEG ML SVM, RF, DT 10K 0.81, 0.85, 0.81 079, 0.89, 0.77 0.84, 0.82, 0.86 0.92, 0.96, 0.88 NR Retrospective
15 Venkat et al. [168] 2023 82 CVD LBBM, OBBM SNP DEG ML RF 5K 0.80 NR NR NR NR Prospective

SN, serial number; SS, Sample size; GT, Ground Truth; Tech, Technique; BM, Benchmark; LBBM, Lab-base biomarkers; OBBM, Office base biomarkers; SD, Source description; AI, Artificial Intelligence; CT, classifier type; CVP, Cross-validation protocol; CTR, Clinical trials; ACC, Accuracy; SEN, Sensitivity; SPE, Specificity; AUC, Area under the curve; ML, Machine Learning; DL, Deep Learning; CNN, Convolution neural network; PDM, parameter decreasing method; ANN, Artificial neural network; PDM, parameter decreasing method; DEG, Differential expression gene; SVM, Support vector machine; RF, Random forest; DT, Decision tree; NR, Not Reported; DCM, Dilated cardiomyopathy; AMI, Acute myocardial infarction.

Valavanis et al. [155] (2010) demonstrated the ability to predict CVD using DNA data and the DL algorithm PDM-ANN. With a sample size of 63 in a retrospective study, the accuracy of the prediction was found to be 95.5%. However, Beigi et al. [156] (2011) used SNP data with an SVM-based ML paradigm to predict CVD. In a prospective study, the sample size was only 23, but it still reached a very high accuracy of 91%. By combining SNP data with ML algorithms like SVM, RF, and XGB, Montañez et al. [157] (2017) conducted a retrospective study that predicted CVD. In the study, there were 12 participants, and the SPE parameter managed to reach an AUC of 0.9.

Zhao et al. [158] (2019) carried out a prospective study that uses SNP data in conjunction with the DL technique known as CNN to predict CVD. The total number of participants in the trial was 490, and the AUC was 0.82. To predict dilated cardiomyopathy (DCM), Schiano et al. [159] (2021) used mRNA data in conjunction with the DT-based ML technique. The study has an accuracy of 0.79 with a sample size of 443. Yang et al. [160] (2021) carried out the study, which used SNP data and ML algorithms like SVM and RF to predict CVD. The retrospective study included 117 participants and obtained an AUC value of 0.91 for the SEN of 0.93. The study by Akbarzadeh et al. [161] (2022) uses DNA data and ML algorithms SVM, DT, RF, and LR to predict CVD. The prospective research study utilized a sizable sample size of 4756 participants and successfully achieved a range of performance criteria, one of which was an AUC of 0.78 for the ACC. The study, which was conducted by Kesar et al. [162] (2022), forecasted CVD using both LBBM and OBBM; however, the data types were not stated. The research had a total of 203 participants as its sample size, and it managed to get an AUC of 0.75 and an F1 score of 0.76. The research was prospective in scope.

Overall, these studies demonstrate the potential of AI-based genomics approaches to predict CVD accurately and provide personalized and precise care for patients. However, more studies are needed to validate these approaches’ effectiveness and assess their impact on clinical practice.

5. Explainable AI, Reliability Stability, and Survival Analysis of Model for CVD Risk Stratification in Joint Cloud and Pharmaceutical Framework

This section discusses using explainable AI, reliability stability, and survival analysis techniques to design and develop the CVD risk stratification in a joint cloud and pharmaceutical framework. It examines the benefits and limitations of these techniques and provides examples of successful implementations. Section 5.1 focuses on the importance of explainability in AI-based decision-making in the pharmaceutical paradigm. It discusses different techniques to ensure explainability, provides examples of use cases, and examines potential benefits and limitations.

Section 5.2 discusses the use of cloud-based solutions for AI in the pharmaceutical industry, including the benefits of scalability, flexibility, and cost-effectiveness. It also examines potential challenges and limitations, such as data security and privacy concerns, and provides case studies of successful implementations. Subsection 5.3 explains the use of statistical methods to analyze the reliability and accuracy of AI prediction models in the pharmaceutical industry. It provides an overview of common statistical techniques, different types of AI prediction models, and case studies of successful applications. Lastly, Subsection 5.4 focuses on applying AI-based survival analysis in the pharmaceutical industry. It examines different types of AI-based survival analysis models, their benefits, and the importance of interpretability and explainability. It also provides case studies of successful applications.

5.1 Explainable AI-Based Decision-Making in Pharmaceutical Model

In the past, AI models were considered “black boxes”, meaning that it was unknown what specific patterns in the input data would result in the desired output. To put it another way, even if a classification algorithm is nearly perfect, humans must still determine what circumstances led to the categorisation [169]. The lack of transparency in model decisions becomes problematic when a model is trained to make accurate predictions based on features that are irrelevant to the situation at hand, and, as a result, it lacks generalization for that dataset [170]. Fig. 8 (Ref. [171]) shows the eight customizable DL processes: (i) DL training, (ii) quality assurance (QA), (iii) installation, (iv) deployment, (v) prediction, (vi) cross-validation-based testing (A/B test), (vii) monitoring, and (viii) debugging. These processes are made possible by incorporating the feedback loop in XAI. Incorporating XAI enhances its usability, increasing demand for such systems and generating higher revenue [172]. Moreover, it offers greater stability to the AI system, thus extending the lifespan of the product design. XAI strategies expose sophisticated model decision patterns [173]. XAI can provide new information about a dataset and the disease by revealing input variables’ predictive importance, including risk factors, traits, and protein expression levels. However, models like principal component analysis (PCA), LR, and Cox linear regression use model parameters to determine feature relevance [174].

Fig. 8.

Eight aspects of XAI [171].

The average importance of features across samples is determined using feature analysis [175]. Training models to distinguish across classes can be expanded to numerous classes, providing insights such as specific patient groups [176]. XAI clinical power stems from strategies that explain patient decisions. Fig. 9 illustrates the clinical decision system concept employing XAI. If the AI model predicts a patient’s risk, the doctor can inquire about the model’s output for clinical, imaging, or molecular features route databases and gene set enrichment estimates can uncover risk variables and molecular disease pathways. This information can help clinicians choose better treatment plans. Furthermore, by understanding the decision process, the physician will be able to judge the accuracy of the predictions. Explainability has thus been recommended as an ethical requirement for prospective clinical decision systems [177]. This section describes various XAI techniques and summarises medical and clinical applications for patient evaluation [8, 42, 43, 153].

Fig. 9.

Clinical features decisions system using XAI. AI models are trained using clinical, imaging, or multi-omics data into different classes (low, mild, moderate, and high). The trained algorithm predicts (prediction blocks) patient disease or symptom risk. Lastly, the XAI presents decision patterns to help the doctor evaluate the forecast and plan treatment (Original image, AtheroPoint™ LLC, Roseville, CA, USA).

Explainable Modeling

XAI can help interpret the results of a clinical Pharma Mode, such as the diseases like CVD. An ML model was trained in Pharma Mode to predict the risk of CVD based on clinical and demographic variables. Local interpretable model-agnostic explanations (LIME) work by training a local linear model to approximate the behavior of the ML model in the neighbourhood of a specific instance. The local model can be used to identify which features strongly influence the predicted outcome for that instance. The equation for the local linear model is:

(1) f ( x ) = w 0 + w 1 * x 1 + w 2 * x 2 + + w n * x n

where, x is a modified version of the original input x, and w0, w1, …, wn are the weights of the linear model. LIME uses a technique called feature perturbation to generate modified instances x that are similar to the original instance but have one or more features changed. The local model is trained on a set of perturbed instances and their corresponding predictions, and the weights are learned using linear regression. On the other hand, Shapley additive explanations (SHAP) use a game-theoretic approach to assign a contribution score to each feature in the model. The contribution score represents the degree to which each feature contributes to the difference between the model’s and average predictions across all possible feature combinations. The equation for the SHAP value of a feature i is:

(2) ϕ i ( x ) = ( 1 M ) * m j 1 M [ f ( x j ) - E [ f ( z j ) ] * h i ( z j ) ]

where x is the instance to be explained, M is the number of feature combinations, f is the ML model, E[f(zj)] is the expected value of foverall feature combinations that include feature i, and hi is a function that measures the difference between the prediction of the model with and without feature i for a given instance zj. The SHAP value of a feature i represents the average contribution of that feature to the difference between the model’s prediction for x and the average prediction for all possible feature combinations. In the context of the CVD Pharma Mode, LIME and SHAP techniques can be used to identify which clinical and demographic features are most important for predicting the risk of CVD and to explain how the model arrives at its predictions for individual patients. This information can help physicians make more informed decisions about patient care and identify areas where further research is needed.

5.2 Cloud-based Explainable AI in Pharmaceutical Model

Cloud-based XAI is an approach that allows researchers and healthcare professionals to access powerful ML/DL models and tools for data analysis and prediction without needing specialized hardware or software. In the context of predicting CVD, cloud-based XAI can provide an efficient and cost-effective way to analyze large amounts of data and generate accurate predictions. Cloud-based XAI relies on cloud computing technology, which involves using remote servers to store and process data rather than relying on local resources [43, 72, 178, 179]. Fig. 10 shows a cloud-based XAI system. By leveraging cloud-based infrastructure, researchers can access powerful computing resources and AI models that can process vast amounts of data quickly and efficiently.

Fig. 10.

AtheroCloud™ system depicts a typical daily routine. A yellow color fills the plaque region, representing the total plaque area after the image has been loaded in the Routine mode and the “Auto Trace” button has been clicked (Original image, AtheroPoint™ LLC, Roseville, CA, USA).

To build a cloud-based XAI system for predicting CVD reserchers can follow a few steps: (i) Feature selection: Feature selection is the process of identifying the most important features contributing to predicting CVD. This can be accomplished through the use of statistical approaches such as regression analysis or machine learning/deep learning algorithms that automatically choose the most relevant features. (ii) Model interpretability: The model can be made more interpretable using algorithms that explain individual predictions, such as LIME and SHAP. These algorithms provide feature importance scores and highlight the most important features for a specific prediction. (iii) Data visualization: Data visualization can be utilized to explore data and uncover patterns and relationships among features. Visualization tools such as scatter plots, heatmaps, and box plots can help researchers identify outliers and correlations that may be useful for predicting CVD. (iv) Cloud-based infrastructure: Cloud-based infrastructure can be used to store and process large amounts of data, making it possible to build more complex and accurate models.

Cloud-based AI platforms such as Amazon Web Services (AWS) and Google Cloud Platform (GCP) provide powerful tools for building and deploying ML/DL models [180]. (v) Collaboration: Collaboration is essential for developing accurate and reliable models for CVD prediction. Cloud-based XAI platforms enable researchers to share data and models, collaborate on model development, and work together to identify new insights and approaches [181]. Cloud-based XAI has several advantages over traditional on-premises solutions. First, researchers can access powerful computing resources without needing specialized hardware or software. Second, it provides a scalable and flexible platform that can be easily adapted to meet changing healthcare needs. Finally, it enables researchers and healthcare professionals to collaborate and share data and models, leading to better and more accurate predictions for CVD and other health conditions.

5.3 Explainable AI Using Heatmaps

Heatmaps in Explainable AI work by highlighting the regions of input data that significantly influence the model’s output. For image-based tasks, such as object detection or medical image analysis, heatmaps visually depict the importance of different pixels or regions within the input image. The intensity of colors in the heatmap indicates the magnitude of the model’s focus on particular areas [182]. The generation of heatmaps often involves techniques like Gradient-weighted Class Activation Mapping (Grad-CAM) and Guided Backpropagation. Grad-CAM computes the gradients of the target class with respect to the final convolutional layer’s feature maps. These gradients are then global-average-pooled to obtain the heatmap, which highlights the relevant regions in the image. Guided Backpropagation, on the other hand, is used to ensure that only meaningful features are highlighted in the heatmap by preventing the model from backpropagating through unimportant regions [183]. By interpreting heatmaps, researchers, domain experts, and end-users gain valuable insights into why a specific decision was made by the AI model. For instance, in medical diagnosis, heatmaps can help doctors understand which regions of an X-ray or MRI scan contributed most to the AI model’s identification of a certain condition [184].

Skandha et al. [138] proposed the first of its kind method to characterize and classify the carotid plaque using an optimized transfer learning approach and SuriNet (a class of Atheromatic™ 2.0 TL). Eleven Atheromatic™ models were implemented, and the best AUC was 0.961 (p < 0.0001) from MobileNet and 0.927 (p < 0.0001) from SuriNet. We validated the performance using grayscale median, fractal dimension, higher-order spectra, and spatial heatmaps. TL showed equal and comparable performance to deep learning. The Atheromatic™ 2.0 TL model showed a performance improvement of 12.9% over Atheromatic™ 1.0 ML (AtheroPoint™, Roseville, CA, USA) compared with the previous. Moreover, the Atheromatic™ 2.0 TL model displayed an outstanding 12.9% performance improvement over the previous Atheromatic™ 1.0ML (AtheroPoint™, Roseville, CA, USA), which was based on traditional machine learning techniques. Notably, the system’s validity was established using a widely accepted dataset, reinforcing the robustness and reliability of the findings. This study sets a significant milestone in understanding carotid plaque characteristics, opening new avenues for enhanced diagnosis and treatment in the field of carotid artery disease.

In conclusion, heatmaps are a crucial tool in the toolkit of Explainable AI, offering visual interpretations of AI model decisions and making complex AI systems more transparent and interpretable. As AI continues to impact various aspects of our lives, XAI using heatmaps will play an instrumental role in building responsible and trustworthy AI applications.

5.4 Statistical Analysis of Artificial Intelligence Prediction Models

The statistical analysis involved the presentation of continuous variables as mean ± standard deviation. The normal distribution of continuous variables was verified using the Kolmogorov-Smirnov test [185]. The independent samples t-test or Mann-Whitney U test was used to compare continuous data, while the chi-square test or Fisher’s exact test was used for categorical variables, as appropriate [186]. For comparisons involving multiple groups, one-way analysis of variance (ANOVA) or Kruskal-Wallis H test with Bonferroni test was used, as appropriate [187]. Pearson r and Spearman rho coefficient were used for correlation assessment, depending on the nature of the data [188].

Multivariable linear regression analysis assessed the association between left atrium (LA) function parameters and myocardial scar size/location [118]. The regression models included demographic characteristics, traditional cardiovascular risk factors, and Late gadolinium enhancement (LGE) location/size as covariates. To avoid co-linearity, Spearman correlation coefficients were used to test for correlations between continuous variables, and variables with r >0.50 were excluded from the same multivariable model [189]. A p-value < 0.05 was considered statistically significant. The analysis was performed using IBM SPSS Statistics version 22 (SPSS Inc., Chicago, IL, USA) [190].

5.5 Artificial Intelligence-Based Survival Analysis

AI-based survival analysis is a cutting-edge technique used to predict the risk of CVD [134]. Using huge amounts of medical data, AI algorithms may estimate the chance of an individual having CVD based on parameters such as age, gender, blood pressure, cholesterol levels, and lifestyle patterns [129]. This approach can give physicians valuable insights into patient health and help them create personalized treatment plans for each individual [191].

By utilizing AI-based survival analysis, doctors can also identify high-risk individuals who may require closer monitoring or more aggressive treatment to prevent the development of CVD [192]. AI technology can revolutionize the healthcare industry by improving the accuracy of CVD diagnosis and treatment, leading to better patient outcomes and improved overall public health [69, 193].

6. Critical Discussions

The ML/DL system needs to overcome critical concerns like bias, explainability, ergonomic design, and affordability to ensure the safety and effectiveness of the medical product, such as CVD risk stratification.

6.1 Principal Findings

This is the first study of its kind (a) that combines radiomics and genomic biomarkers to detect the severity of CVD and stroke risk precisely, and (b) an explainable XAI-based composite risk CVD/Stroke model using DL can predict with precision in a preventive and personalized (aiP3) framework benefiting pharmaceutical paradigm. Using these two hypotheses, we demonstrated that CVD and stroke risk severity could be determined using RBBM and GBBM biomarkers in the DL framework. Such models can be considered “personalized medicine frameworks”. One significant advancement is the inclusion of carotid imaging and CVD genetic indicators in the DL framework for CVD risk classification. A set of six recommendations for accurate, robust, real-time CVD risk assessment utilizing combined RBBM and GBBM were offered. The study proposed model having significant potential such as (a) radiomics and genomic biomarkers have a strong correlation and can be used to detect the severity of CVD and stroke precisely, and (b) The proposed review suggests a unique, unbiased, and XAI model (AtheroEdgeTM 4.0) that can predict the composite risk of CVD/Stroke using radiogenomics in the pharmaceutical paradigm. The proposed model required less time than the traditional model to stratify CVD/stroke risk, saving thousands of hours using phrmamode.

AtheroCloudTM can be utilized in either the (a) “Routine” mode or the (b) “Pharma” trial mode. Both the batch of carotid ultrasound scans and individual scans are automatically processed in real-time in the Routine mode and Pharma trial mode, respectively, during the patient visit. The system supports large databases, from gigabytes to terabytes, and can operate in batch mode, processing as many as 10,000 images simultaneously.

6.2 Benchmarking

Table 4 (Ref. [27, 41, 75, 119, 143, 159, 194, 195, 196, 197, 198, 199, 200, 201]) displays the benchmarking studies. It has 17 attributes, namely, K0: Serial Number, K1: Studies, K2: Year, K3: References, K4: OBBM, K5: LBBM, K6: RBBM, K7: GBBM, K8: PBBM, K9: EBBM, K10: Preventive, K11: Prediction, K12: Personalized, K13: AI Type, K14: FDA discussion, K15: Clinical Setting, K16: Risk of bias; K17: AI explainability. Our observations showed that there were only eight studies [27, 41, 159, 194, 195, 196, 197] that discussed the role of GBBM in CVD using DL, while the remaining studies predicted CVD using RBBM [75, 119, 143, 198, 199, 200, 202, 203]. However, all the studies mentioned the role of OBBM and LBBM in CVD risk. Five studies adopted DL technologies [27, 41, 75, 196, 198, 199], while eight studies used ML [11, 131, 197, 202], and two studies used HDL [143, 201]. Only four studies discussed the risk of bias (RoB) [75, 143, 202]. Except for the proposed study, no other study addressed clinical settings, AI explainability, and fusion of GBBM and RBBM.

Table 4.Benchmarking table for CVD risk using multivariate biomarkers.
K0 K1 K2 K3 K4 K5 K6 K7 K8 K9 K10 K11 K12 K13 K14 K15 K16 K17
1 Krittanawong et al. [194] 2017 31 × × NR × × × ×
2 Arena et al. [195] 2018 202 × × × × NR × × × ×
3 Krittanawong et al. [27] 2018 88 × × × × DL × × × ×
4 Jamthikar et al. [143] 2019 110 × × × × HDL × × × ×
5 Saba et al. [199] 2019 125 × × × × DL × × × ×
6 Dainis et al. [41] 2019 83 × × × × DL × × × ×
7 Gruson et al. [201] 2020 42 × × × HDL × × × ×
8 Alimadadi et al. [197] 2020 56 × × × × ML × × × ×
9 Saba et al. [198] 2020 69 × × × × ML × × ×
10 Westerlund et al. [196] 2021 167 × × × × DL × × × ×
11 Schiano et al. [159] 2021 29 × × × ML × × × ×
12 Jain et al. [119] 2022 67 × × × × × ML × × × ×
13 Staub et al. [200] 2022 25 × × × × × NR × × × ×
14 Jain et al. [75] 2022 85 × × × × DL × ×
15 Saba et al. (Proposed) 2023 214 DL × ×

K0: Serial Number, K1: Studies, K2: Year, K3: References, K4: OBBM, K5: LBBM, K6: RBBM, K7: GBBM, K8: PBBM, K9: EBBM, K10: Preventive, K11: Prediction, K12: Personalized, K13: AI Type, K14: FDA discussion, K15: Clinical Setting, K16: Risk of Bias, K17: AI explainability. CVD, Cardiovascular Disease; DL, Deep Learning; ML, Machine Learning; HDL, Hybrid deep learning; NR, Not reported.

6.3 The Role of AI Bias during Designs

Evaluating bias in AI models has gained much greater significance in recent years [204, 205]. Earlier computer-aided diagnosis techniques showed a lack of bias in evaluations [206]. To reduce bias, a large sample size, appropriate clinical testing, the incorporation of comorbidities, the use of big data configurations, the usage of unseen data analysis, and the scientific validation of training model design are all strategies that can be utilized [140, 207]. Essential phases in patient risk stratification include determining the AI RoB [191, 207, 208] and suitably modifying diagnostics and treatment.

6.4 The Role of Pruning-Based AI Systems

Edge devices are becoming increasingly important as cloud-based systems and the internet improve. Edge devices are extremely important when using trained AI models for future predictions or disease risk stratifications in mobile frameworks. There is a requirement to deploy compressed models since huge data models cannot be deployed on edge devices. Image-based DL models such as fully convolutional networks (FCN) or segmentation networks (SegNet) [40] can be pruned using evolutionary algorithms such as Particle Swarm Optimization (PSO), Genetic Algorithms (GA), Wolf Optimization (WO), and Differential Evolution (DE) [209]. The future of radiomics-based CVD risk stratification fused genetic-based paradigms can be compressed and deployed on edge devices for rural areas, especially in third-world nations.

6.5 Recommendations

The following are recommendations for a suggested Radiogenomics model that can be used to stratify CVD/stroke risk. The study proposes two hypotheses: (a) radiomics and genomic biomarkers have a strong correlation and can be used to detect the severity of CVD and stroke precisely, and (b) The proposed review suggests a unique, unbiased, and XAI model (AtheroEdgeTM 4.0) that can predict the composite risk of CVD/Stroke using radiogenomics in the pharmaceutical paradigm. The following recommendations are: (i) requires a clinical evaluation and scientific validation for reliable detection and CVD risk stratification, and (ii) requires hyper-parameter optimization in CVD/Stroke risk stratification. (iii) balancing the risk classes (control, low-risk, and high-risk) is the most effective way to minimize DL bias; (iv) with proper pruning and compression, DL systems can be adapted to edge devices; (v) A DL system that relies on surrogate carotid imaging can be cost-effective without compromising precision in CVD risk stratification.

6.6 Strengths, Weakness, and Extensions

This pilot review’s ability to risk stratify CVD and stroke patients by integrating RBBM and GBBM was a major strength. The biomarkers generated from radiological, biochemical, and morphological complexity that demonstrated a link to CVD supported the first hypothesis. An XAI-based composite risk CVD/Stroke model using DL can predict, with precision, in a preventive and personalized (aiP3) framework benefiting the pharmaceutical paradigm. A DL approach was presented to evaluate CVD and stroke risk by integrating RBBM and GBBM. While the system is pretty straightforward, it requires optimization to eliminate the possibility of bias and generalization to account for comorbidities. Better comprehensive feature space can be tried for superior DL-based classification [210]. Ensemble-based solutions embedding PCA for best feature selection followed by recurrent neural networks are possible extensions for superior CVD/stroke risk solutions [211].

6.7 The Role Odds Ratio in Clinical Trials

In clinical trials, researchers often analyze the effectiveness of a new treatment or intervention compared to a control group. One of the essential statistical measures used in this context is the “Odds Ratio” (OR). The odds ratio helps to assess the association between an exposure (such as receiving a treatment) and an outcome (such as a positive response to the treatment) [212].

The odds ratio is calculated by taking the ratio of two odds: the odds of an event occurring in the treatment group and the odds of the same event occurring in the control group. Mathematically, it can be expressed as:

The odds ratio is calculated as follows:

(3) OR = ( a / b ) / ( c / d )

Where:

a is the number of people in the exposed group who have the outcome

b is the number of people in the exposed group who do not have the outcome

c is the number of people in the unexposed group who have the outcome

d is the number of people in the unexposed group who do not have the outcome

The odds ratio can be used to determine if there is a significant difference in the odds of the outcome between the treatment and control groups. If the odds ratio is equal to 1, it suggests that there is no association between the treatment and the outcome, meaning the treatment has no effect. If the odds ratio is greater than 1, it indicates that the treatment is associated with a higher likelihood of the outcome. Conversely, if the odds ratio is less than 1, it implies that the treatment is associated with a lower likelihood of the outcome [213].

Researchers typically calculate the odds ratio along with a confidence interval to assess the statistical significance of the results. If the confidence interval includes the value 1, the results are considered not statistically significant, while values above or below 1 indicate a statistically significant association [214]. The odds ratio is a useful tool in clinical trials as it provides valuable information about the relative impact of a treatment compared to a control group. However, it’s essential to interpret the odds ratio in the context of the study design and the specific clinical question being addressed [214]. In conclusion, the odds ratio is a critical statistical measure used in clinical trials to analyze the association between treatment and outcomes, providing valuable insights into the effectiveness of interventions.

7. Conclusions

The proposed research investigation presented two hypotheses: (i) an extensive investigation into several genomic biomarkers, namely, IL, CD31+, EPCs, and hs-CRP, which have been found to correlate most significantly with prognosis. The surrogate radiomic features such as plaque burden, plaque area, carotid intima thickness, maximum plaque height, and lumen stenosis/lumen diameter further improve the CVD risk prediction. (ii) The proposed review suggests a unique, unbiased, and XAI model (AtheroEdgeTM 4.0) that can predict the composite risk of CVD/Stroke using radio genomics in the pharmaceutical paradigm. There is a strong link between the genetic and radiomic biomarkers of atherosclerosis in the carotid, coronary, and aortic arteries. A DL-based system is critical in reducing nonlinearity among multi-covariate biomarkers and outcomes. During this pilot review, various issues were considered, such as AI bias, AI pruning, and a suggestion for a cloud-based cascaded system design.

Abbreviations

ARDS, Acute respiratory distress syndrome; LGE, Late gadolinium enhancement; ASCVD, Atherosclerotic cardiovascular disease; LIME, Local interpretable model-agnostic explanations; ANS, Autonomic nervous System; MRI, Magnetic resonance imaging; AUC, Area-under-the-curve; NR, Not reported; AI, Artificial intelligence; NPV, Negative predictive value; ACS, Acute coronary syndrome; NB, Naive bayes; BMI, Body mass index; Non-ML, Non-machine learning; CAD, Coronary artery disease; OBBM, Office-based biomarker; CAS, Coronary artery syndrome; OH, Orthostatic hypotension; CHD, Coronary heart disease; OxLDL, Oxidation of low-density lipoprotein; CT, Computed tomography; PE, Performance evaluation; CUSIP, Carotid ultrasound image phenotype; PPV, Positive predictive value; CV, Cross-validation; PCA, Principal component analysis; CVD, Cardiovascular disease; PBBM, Proteomics based bio-markers; CVE, Cardiovascular events; PRISMA, Preferred reporting items for systematic reviews and meta-analyses; CNN, Convolution neural network; PTC, Plaque tissue characterization; DL, Deep learning; RA, Rheumatoid arthritis; DM, Diabetes mellitus; RF, Random forest; DT, Decision tree; ROS, Reactive oxides stress; EC, Endothelial cell; RoB, Risk of bias; EBBM, Environment-based biomarkers; ROC, Receiver operating-characteristics; GT, Ground truth; RNN, Recurrent neural network; GBBM, Genetically based biomarkers; SCORE, Systematic coronary risk evaluation; HTN, Hypertension; SMOTE, Synthetic minority over-sampling technique; HDL, Hybrid deep learning; SVM, Support vector machine; ICAM, Intercellular adhesion molecule; SHAP, Shapley additive explanations; VCAM, Vascular cell adhesion molecule; TPA, Total plaque area; LBBM, Laboratory-based biomarker; TC, Tissue characterization; LA, left atrium; US, Ultrasound.

Author Contributions

MM: Design of the manuscript, proofreading many iterations, researching PubMed and other research sites for article search, AMJ, KIP, NNK, MM, JFF, JSS: Resources, imaging contribution and proofreading of the manuscript, MM, AMJ, LM, ERI, MMF: Design of the genomics and genetics component of the manuscript, proofreading many iterations, researching PubMed and other research sites for article search, JSS, VR, VV, MKK, NS: Proofreading and guidance of cardiology components of the manuscript, JSS, AN, NNK: The vision of cardiac risk assessment and proofreading the manuscript, final approval of the manuscript, MKK, LS: Design and support of radiology components such as CT and carotid ultrasound, JRL, SC, MMF, MAM: Proofreading and guidance of cardiology imaging components of the manuscript, JRL, VV, KIP ZR, ERI: Proofreading and guidance of cardiology and genomics components, MM, JSS: Design and solid proofreading of the manuscript, especially the Phramamode component, revising it critically for important intellectual content, and final approval of the manuscript, SC, VR, MKK ERI, MMF: clinical trial and proofreading of the manuscript, JSS: Principal Investigator-design, proofreding of the manuscript and management. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

Dr. Suri and Dr. Maindarkar is with AtheroPoint™ LLC, Roseville, CA, USA, which does cardiovascular and stroke imaging. The authors declare no conflict of interest.

References
[1]
Gunnarsson SI, Peppard PE, Korcarz CE, Barnet JH, Aeschlimann SE, Hagen EW, et al. Obstructive sleep apnea is associated with future subclinical carotid artery disease: thirteen-year follow-up from the Wisconsin sleep cohort. Arteriosclerosis, Thrombosis, and Vascular Biology. 2014; 34: 2338–2342.
[2]
Smith DF, Schuler CL, Hossain MM, Huang G, McConnell K, Urbina EM, et al. Early Atherosclerotic Inflammatory Pathways in Children with Obstructive Sleep Apnea. The Journal of Pediatrics. 2021; 239: 168–174.
[3]
Wong I, Swanson N. Approaches to managing work-related fatigue to meet the needs of American workers and employers. American Journal of Industrial Medicine. 2022; 65: 827–831.
[4]
Hirata T, Arai Y, Takayama M, Abe Y, Ohkuma K, Takebayashi T. Carotid Plaque Score and Risk of Cardiovascular Mortality in the Oldest Old: Results from the TOOTH Study. Journal of Atherosclerosis and Thrombosis. 2018; 25: 55–64.
[5]
Park HW, Kim WH, Kim KH, Yang DJ, Kim JH, Song IG, et al. Carotid plaque is associated with increased cardiac mortality in patients with coronary artery disease. International Journal of Cardiology. 2013; 166: 658–663.
[6]
Kim H, Lim DH, Kim Y. Classification and Prediction on the Effects of Nutritional Intake on Overweight/Obesity, Dyslipidemia, Hypertension and Type 2 Diabetes Mellitus Using Deep Learning Model: 4-7th Korea National Health and Nutrition Examination Survey. International Journal of Environmental Research and Public Health. 2021; 18: 5597.
[7]
Zhang B, Li G, Ma Y, Pan X. Projection of temperature-related mortality due to cardiovascular disease in beijing under different climate change, population, and adaptation scenarios. Environmental Research. 2018; 162: 152–159.
[8]
Khanna NN, Maindarkar MA, Viswanathan V, Puvvula A, Paul S, Bhagawati M, et al. Cardiovascular/Stroke Risk Stratification in Diabetic Foot Infection Patients Using Deep Learning-Based Artificial Intelligence: An Investigative Study. Journal of Clinical Medicine. 2022; 11: 6844.
[9]
Domingueti CP, Dusse LMS, Carvalho MDG, de Sousa LP, Gomes KB, Fernandes AP. Diabetes mellitus: The linkage between oxidative stress, inflammation, hypercoagulability and vascular complications. Journal of Diabetes and its Complications. 2016; 30: 738–745.
[10]
Rana JS, Nieuwdorp M, Jukema JW, Kastelein JJP. Cardiovascular metabolic syndrome - an interplay of, obesity, inflammation, diabetes and coronary heart disease. Diabetes, Obesity & Metabolism. 2007; 9: 218–232.
[11]
Jamthikar A, Gupta D, Cuadrado-Godia E, Puvvula A, Khanna NN, Saba L, et al. Ultrasound-based stroke/cardiovascular risk stratification using Framingham Risk Score and ASCVD Risk Score based on “Integrated Vascular Age” instead of “Chronological Age”: a multi-ethnic study of Asian Indian, Caucasian, and Japanese cohorts. Cardiovascular Diagnosis and Therapy. 2020; 10: 939–954.
[12]
Jamthikar AD, Gupta D, Mantella LE, Saba L, Laird JR, Johri AM, et al. Multiclass machine learning vs. conventional calculators for stroke/CVD risk assessment using carotid plaque predictors with coronary angiography scores as gold standard: a 500 participants study. The International Journal of Cardiovascular Imaging. 2021; 37: 1171–1187.
[13]
Littnerova S, Kala P, Jarkovsky J, Kubkova L, Prymusova K, Kubena P, et al. GRACE Score among Six Risk Scoring Systems (CADILLAC, PAMI, TIMI, Dynamic TIMI, Zwolle) Demonstrated the Best Predictive Value for Prediction of Long-Term Mortality in Patients with ST-Elevation Myocardial Infarction. PloS One. 2015; 10: e0123215.
[14]
Fabi M, Andreozzi L, Corinaldesi E, Bodnar T, Lami F, Cicero C, et al. Inability of Asian risk scoring systems to predict intravenous immunoglobulin resistance and coronary lesions in Kawasaki disease in an Italian cohort. European Journal of Pediatrics. 2019; 178: 315–322.
[15]
Biswas M, Kuppili V, Araki T, Edla DR, Godia EC, Saba L, et al. Deep learning strategy for accurate carotid intima-media thickness measurement: An ultrasound study on Japanese diabetic cohort. Computers in Biology and Medicine. 2018; 98: 100–117.
[16]
Yamagishi SI, Matsui T. Role of Hyperglycemia-Induced Advanced Glycation End Product (AGE) Accumulation in Atherosclerosis. Annals of Vascular Diseases. 2018; 11: 253–258.
[17]
Tavil Y, Kanbay A, Sen N, Ulukavak Ciftçi T, Abaci A, Yalçin MR, et al. The relationship between aortic stiffness and cardiac function in patients with obstructive sleep apnea, independently from systemic hypertension. Journal of the American Society of Echocardiography: Official Publication of the American Society of Echocardiography. 2007; 20: 366–372.
[18]
Oñatibia-Astibia A, Larrañaga B, Iribar J, Etxeberria A, Odriozola N, Montero-Muñoz A, et al. A communication procedure between community pharmacists and primary care professionals resolves medication errors and other administrative issues. The International Journal of Pharmacy Practice. 2022; 30: 235–240.
[19]
Bonkhoff AK, Grefkes C. Precision medicine in stroke: towards personalized outcome predictions using artificial intelligence. Brain: a Journal of Neurology. 2022; 145: 457–475.
[20]
Shah SH, Arnett D, Houser SR, Ginsburg GS, MacRae C, Mital S, et al. Opportunities for the Cardiovascular Community in the Precision Medicine Initiative. Circulation. 2016; 133: 226–231.
[21]
Jaffe S. Planning for US Precision Medicine Initiative underway. Lancet (London, England). 2015; 385: 2448–2449.
[22]
Rezayi S, R Niakan Kalhori S, Saeedi S. Effectiveness of Artificial Intelligence for Personalized Medicine in Neoplasms: A Systematic Review. BioMed Research International. 2022; 2022: 7842566.
[23]
Usova EI, Alieva AS, Yakovlev AN, Alieva MS, Prokhorikhin AA, Konradi AO, et al. Integrative Analysis of Multi-Omics and Genetic Approaches-A New Level in Atherosclerotic Cardiovascular Risk Prediction. Biomolecules. 2021; 11: 1597.
[24]
Doran S, Arif M, Lam S, Bayraktar A, Turkez H, Uhlen M, et al. Multi-omics approaches for revealing the complexity of cardiovascular disease. Briefings in Bioinformatics. 2021; 22: bbab061.
[25]
Wojeck BS, Inzucchi SE, Neeland IJ, Mancuso JP, Frederich R, Masiukiewicz U, et al. Ertugliflozin and incident obstructive sleep apnea: an analysis from the VERTIS CV trial. Sleep & Breathing. 2023; 27: 669–672.
[26]
Chopra H, Baig AA, Gautam RK, Kamal MA. Application of Artificial Intelligence in Drug Discovery. Current Pharmaceutical Design. 2022; 28: 2690–2703.
[27]
Krittanawong C, Zhang H, Wang Z, Aydar M, Kitai T. Artificial Intelligence in Precision Cardiovascular Medicine. Journal of the American College of Cardiology. 2017; 69: 2657–2664.
[28]
El-Baz A, Suri JS. Big data in multimodal medical imaging. CRC Press: CA, USA. 2019.
[29]
Narang M, Walia DR, Kaul DU, Sudhir DK. Evolving Paradigm of Precision Medicine in Cardiovascular Disease. Medical and Clinical Research: Open Access. 2021; 2: 1–8.
[30]
Biswas M, Saba L, Chakrabartty S, Khanna NN, Song H, Suri HS, et al. Two-stage artificial intelligence model for jointly measurement of atherosclerotic wall thickness and plaque burden in carotid ultrasound: A screening tool for cardiovascular/stroke risk assessment. Computers in Biology and Medicine. 2020; 123: 103847.
[31]
Kannel WB, McGee DL, Schatzkin A. An epidemiological perspective of sudden death. 26-year follow-up in the Framingham Study. Drugs. 1984; 28: 1–16.
[32]
Xie Y, Bowe B, Al-Aly Z. Burdens of post-acute sequelae of COVID-19 by severity of acute infection, demographics and health status. Nature Communications. 2021; 12: 6571.
[33]
Saxena S, Jena B, Gupta N, Das S, Sarmah D, Bhattacharya P, et al. Role of Artificial Intelligence in Radiogenomics for Cancers in the Era of Precision Medicine. Cancers. 2022; 14: 2860.
[34]
Shui L, Ren H, Yang X, Li J, Chen Z, Yi C, et al. The Era of Radiogenomics in Precision Medicine: An Emerging Approach to Support Diagnosis, Treatment Decisions, and Prognostication in Oncology. Frontiers in Oncology. 2021; 10: 570465.
[35]
Jamthikar A, Gupta D, Johri AM, Mantella LE, Saba L, Suri JS. A machine learning framework for risk prediction of multi-label cardiovascular events based on focused carotid plaque B-Mode ultrasound: A Canadian study. Computers in Biology and Medicine. 2022; 140: 105102.
[36]
Acharya UR, Sree SV, Krishnan MMR, Molinari F, Zieleźnik W, Bardales RH, et al. Computer-aided diagnostic system for detection of Hashimoto thyroiditis on ultrasound images from a Polish population. Journal of Ultrasound in Medicine: Official Journal of the American Institute of Ultrasound in Medicine. 2014; 33: 245–253.
[37]
Khanna NN, Maindarkar M, Saxena A, Ahluwalia P, Paul S, Srivastava SK, et al. Cardiovascular/Stroke Risk Assessment in Patients with Erectile Dysfunction-A Role of Carotid Wall Arterial Imaging and Plaque Tissue Characterization Using Artificial Intelligence Paradigm: A Narrative Review. Diagnostics (Basel, Switzerland). 2022; 12: 1249.
[38]
Jamthikar AD, Gupta D, Johri AM, Mantella LE, Saba L, Kolluri R, et al. Low-Cost Office-Based Cardiovascular Risk Stratification Using Machine Learning and Focused Carotid Ultrasound in an Asian-Indian Cohort. Journal of Medical Systems. 2020; 44: 208.
[39]
Saba L, Biswas M, Kuppili V, Cuadrado Godia E, Suri HS, Edla DR, et al. The present and future of deep learning in radiology. European Journal of Radiology. 2019; 114: 14–24.
[40]
Agarwal M, Agarwal S, Saba L, Chabert GL, Gupta S, Carriero A, et al. Eight pruning deep learning models for low storage and high-speed COVID-19 computed tomography lung segmentation and heatmap-based lesion localization: A multicenter study using COVLIAS 2.0. Computers in Biology and Medicine. 2022; 146: 105571.
[41]
Dainis AM, Ashley EA. Cardiovascular Precision Medicine in the Genomics Era. JACC. Basic to Translational Science. 2018; 3: 313–326.
[42]
Nillmani, Sharma N, Saba L, Khanna NN, Kalra MK, Fouda MM, et al. Segmentation-Based Classification Deep Learning Model Embedded with Explainable AI for COVID-19 Detection in Chest X-ray Scans. Diagnostics (Basel, Switzerland). 2022; 12: 2132.
[43]
Suri JS, Agarwal S, Chabert GL, Carriero A, Paschè A, Danna PSC, et al. COVLIAS 2.0-cXAI: Cloud-Based Explainable Deep Learning System for COVID-19 Lesion Localization in Computed Tomography Scans. Diagnostics (Basel, Switzerland). 2022; 12: 1482.
[44]
Brink-Kjaer A, Leary EB, Sun H, Westover MB, Stone KL, Peppard PE, et al. Age estimation from sleep studies using deep learning predicts life expectancy. NPJ Digital Medicine. 2022; 5: 103.
[45]
Araki T, Ikeda N, Molinari F, Dey N, Acharjee S, Saba L, et al. Link between automated coronary calcium volumes from intravascular ultrasound to automated carotid IMT from B-mode ultrasound in coronary artery disease population. International Angiology: a Journal of the International Union of Angiology. 2014; 33: 392–403.
[46]
Viswanathan V, Jamthikar AD, Gupta D, Puvvula A, Khanna NN, Saba L, et al. Integration of estimated glomerular filtration rate biomarker in image-based cardiovascular disease/stroke risk calculator: a south Asian-Indian diabetes cohort with moderate chronic kidney disease. International Angiology: a Journal of the International Union of Angiology. 2020; 39: 290–306.
[47]
Munjral S, Ahluwalia P, Jamthikar AD, Puvvula A, Saba L, Faa G, et al. Nutrition, atherosclerosis, arterial imaging, cardiovascular risk stratification, and manifestations in COVID-19 framework: a narrative review. Frontiers in Bioscience (Landmark Edition). 2021; 26: 1312–1339.
[48]
Araki T, Ikeda N, Shukla D, Londhe ND, Shrivastava VK, Banchhor SK, et al. A new method for IVUS-based coronary artery disease risk stratification: A link between coronary & carotid ultrasound plaque burdens. Computer Methods and Programs in Biomedicine. 2016; 124: 161–179.
[49]
Skeoch S, Cristinacce PLH, Williams H, Pemberton P, Xu D, Sun J, et al. Imaging atherosclerosis in rheumatoid arthritis: evidence for increased prevalence, altered phenotype and a link between systemic and localised plaque inflammation. Scientific Reports. 2017; 7: 827.
[50]
Kramer CM, Anderson JD. MRI of atherosclerosis: diagnosis and monitoring therapy. Expert Review of Cardiovascular Therapy. 2007; 5: 69–80.
[51]
Yuan C, Kerwin WS. MRI of atherosclerosis. Journal of Magnetic Resonance Imaging: JMRI. 2004; 19: 710–719.
[52]
Koelemay MJW, Nederkoorn PJ, Reitsma JB, Majoie CB. Systematic review of computed tomographic angiography for assessment of carotid artery disease. Stroke. 2004; 35: 2306–2312.
[53]
Grassi G, Laino ME, Fantini MC, Argiolas GM, Cherchi MV, Nicola R, et al. Advanced imaging and Crohn’s disease: An overview of clinical application and the added value of artificial intelligence. European Journal of Radiology. 2022; 157: 110551.
[54]
Corrias G, Micheletti G, Barberini L, Suri JS, Saba L. Texture analysis imaging “what a clinical radiologist needs to know”. European Journal of Radiology. 2022; 146: 110055.
[55]
Murgia A, Balestrieri A, Crivelli P, Suri JS, Conti M, Cademartiri F, et al. Cardiac computed tomography radiomics: an emerging tool for the non-invasive assessment of coronary atherosclerosis. Cardiovascular Diagnosis and Therapy. 2020; 10: 2005–2017.
[56]
Saba L, Sanfilippo R, Sannia S, Anzidei M, Montisci R, Mallarini G, et al. Association between carotid artery plaque volume, composition, and ulceration: a retrospective assessment with MDCT. AJR. American Journal of Roentgenology. 2012; 199: 151–156.
[57]
Boi A, Jamthikar AD, Saba L, Gupta D, Sharma A, Loi B, et al. A Survey on Coronary Atherosclerotic Plaque Tissue Characterization in Intravascular Optical Coherence Tomography. Current Atherosclerosis Reports. 2018; 20: 33.
[58]
Laine A, Sanches JM, Suri JS. Ultrasound imaging: advances and applications. Springer: USA. 2012.
[59]
Suri JS. Advances in diagnostic and therapeutic ultrasound imaging. Artech House: USA. 2008.
[60]
Beach KW. Principles of Ultrasonic Imaging and Instrumentation. In Andrew, N., Kirk, W.B., Efthivoulos, K., Constantinos, S.P. (eds) Ultrasound and Carotid Bifurcation Atherosclerosis (pp. 67–96). Springer: London. 2012.
[61]
Saba L, Jamthikar A, Gupta D, Khanna NN, Viskovic K, Suri HS, et al. Global perspective on carotid intima-media thickness and plaque: should the current measurement guidelines be revisited? International Angiology: a Journal of the International Union of Angiology. 2019; 38: 451–465.
[62]
Firincioglulari M, Aksoy S, Orhan K, Rasmussen F. Comparison of Intracranial and Extracranial Carotid Artery Calcifications between Obstructive Sleep Apnea Patients and Healthy Individuals: A Combined Cone-Beam Computed Tomography and Polysomnographic Study. Radiology Research and Practice. 2022; 2022: 1625779.
[63]
Ikeda N, Gupta A, Dey N, Bose S, Shafique S, Arak T, et al. Improved correlation between carotid and coronary atherosclerosis SYNTAX score using automated ultrasound carotid bulb plaque IMT measurement. Ultrasound in Medicine & Biology. 2015; 41: 1247–1262.
[64]
Munjral S, Maindarkar M, Ahluwalia P, Puvvula A, Jamthikar A, Jujaray T, et al. Cardiovascular Risk Stratification in Diabetic Retinopathy via Atherosclerotic Pathway in COVID-19/Non-COVID-19 Frameworks Using Artificial Intelligence Paradigm: A Narrative Review. Diagnostics (Basel, Switzerland). 2022; 12: 1234.
[65]
Kotsis V, Jamthikar AD, Araki T, Gupta D, Laird JR, Giannopoulos AA, et al. Echolucency-based phenotype in carotid atherosclerosis disease for risk stratification of diabetes patients. Diabetes Research and Clinical Practice. 2018; 143: 322–331.
[66]
Khanna NN, Jamthikar AD, Gupta D, Nicolaides A, Araki T, Saba L, et al. Performance evaluation of 10-year ultrasound image-based stroke/cardiovascular (CV) risk calculator by comparing against ten conventional CV risk calculators: A diabetic study. Computers in Biology and Medicine. 2019; 105: 125–143.
[67]
Khanna NN, Jamthikar AD, Araki T, Gupta D, Piga M, Saba L, et al. Nonlinear model for the carotid artery disease 10-year risk prediction by fusing conventional cardiovascular factors to carotid ultrasound image phenotypes: A Japanese diabetes cohort study. Echocardiography (Mount Kisco, N.Y.). 2019; 36: 345–361.
[68]
Khanna NN, Jamthikar AD, Gupta D, Araki T, Piga M, Saba L, et al. Effect of carotid image-based phenotypes on cardiovascular risk calculator: AECRS1.0. Medical & Biological Engineering & Computing. 2019; 57: 1553–1566.
[69]
Jamthikar A, Gupta D, Khanna NN, Saba L, Araki T, Viskovic K, et al. A low-cost machine learning-based cardiovascular/stroke risk assessment system: integration of conventional factors with image phenotypes. Cardiovascular Diagnosis and Therapy. 2019; 9: 420–430.
[70]
Liu K, Suri JS. Automatic vessel indentification for angiographic screening. U.S. Patent and Trademark Office: Washington, DC. 2005.
[71]
Biswas M, Saba L, Omerzu T, Johri AM, Khanna NN, Viskovic K, et al. A Review on Joint Carotid Intima-Media Thickness and Plaque Area Measurement in Ultrasound for Cardiovascular/Stroke Risk Monitoring: Artificial Intelligence Framework. Journal of Digital Imaging. 2021; 34: 581–604.
[72]
Saba L, Banchhor SK, Araki T, Viskovic K, Londhe ND, Laird JR, et al. Intra- and inter-operator reproducibility of automated cloud-based carotid lumen diameter ultrasound measurement. Indian Heart Journal. 2018; 70: 649–664.
[73]
Vila MDM, Remeseiro B, Grau M, Elosua R, Betriu À, Fernandez-Giraldez E, et al. Semantic segmentation with DenseNets for carotid artery ultrasound plaque segmentation and CIMT estimation. Artificial Intelligence in Medicine. 2020; 103: 101784.
[74]
Jain PK, Sharma N, Giannopoulos AA, Saba L, Nicolaides A, Suri JS. Hybrid deep learning segmentation models for atherosclerotic plaque in internal carotid artery B-mode ultrasound. Computers in Biology and Medicine. 2021; 136: 104721.
[75]
Jain PK, Sharma N, Kalra MK, Johri A, Saba L, Suri JS. Far wall plaque segmentation and area measurement in common and internal carotid artery ultrasound using U-series architectures: An unseen Artificial Intelligence paradigm for stroke risk assessment. Computers in Biology and Medicine. 2022; 149: 106017.
[76]
Yuan Y, Li C, Zhang K, Hua Y, Zhang J. HRU-Net: A Transfer Learning Method for Carotid Artery Plaque Segmentation in Ultrasound Images. Diagnostics (Basel, Switzerland). 2022; 12: 2852.
[77]
Molinari F, Meiburger KM, Saba L, Acharya UR, Ledda G, Zeng G, et al. Ultrasound IMT measurement on a multi-ethnic and multi-institutional database: our review and experience using four fully automated and one semi-automated methods. Computer Methods and Programs in Biomedicine. 2012; 108: 946–960.
[78]
Gago L, Vila MDM, Grau M, Remeseiro B, Igual L. An end-to-end framework for intima media measurement and atherosclerotic plaque detection in the carotid artery. Computer Methods and Programs in Biomedicine. 2022; 223: 106954.
[79]
Shin J, Choi EY, Kwon HM, Rhee K. Estimation of viscoelasticity of a carotid artery from ultrasound cine images and brachial pressure waveforms: Viscous parameters as a new index of detecting low plaque burden. Medical Engineering & Physics. 2022; 108: 103886.
[80]
Lainé N, Zahnd G, Liebgott H, Orkisz M. ‘Segmenting the carotid-artery wall in ultrasound image sequences with a dual-resolution U-net’ , 2022 IEEE International Ultrasonics Symposium (IUS). IEEE. 2022.
[81]
O’Sullivan JW, Raghavan S, Marquez-Luna C, Luzum JA, Damrauer SM, Ashley EA, et al. Polygenic Risk Scores for Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation. 2022; 146: e93–e118.
[82]
Collister JA, Liu X, Clifton L. Calculating Polygenic Risk Scores (PRS) in UK Biobank: A Practical Guide for Epidemiologists. Frontiers in Genetics. 2022; 13: 818574.
[83]
Gola D, Erdmann J, Müller-Myhsok B, Schunkert H, König IR. Polygenic risk scores outperform machine learning methods in predicting coronary artery disease status. Genetic Epidemiology. 2020; 44: 125–138.
[84]
Ho DSW, Schierding W, Wake M, Saffery R, O’Sullivan J. Machine Learning SNP Based Prediction for Precision Medicine. Frontiers in Genetics. 2019; 10: 267.
[85]
Li R, Chen Y, Ritchie MD, Moore JH. Electronic health records and polygenic risk scores for predicting disease risk. Nature Reviews. Genetics. 2020; 21: 493–502.
[86]
Ramírez J, van Duijvenboden S, Young WJ, Tinker A, Lambiase PD, Munroe PB, et al. ‘Interaction Between ECG and Genetic Markers of Coronary Artery Disease’, 2020 Computing in Cardiology. IEEE. 2020.
[87]
Cugliari G, Benevenuta S, Guarrera S, Sacerdote C, Panico S, Krogh V, et al. ‘Improving the prediction of cardiovascular risk with machine-learning and dna methylation data’, 2019 IEEE Conference on Computational Intelligence in Bioinformatics and Computational Biology (CIBCB). IEEE. 2019.
[88]
Krittanawong C, Johnson KW, Choi E, Kaplin S, Venner E, Murugan M, et al. Artificial Intelligence and Cardiovascular Genetics. Life (Basel, Switzerland). 2022; 12: 279.
[89]
Widen E, Raben TG, Lello L, Hsu SDH. Machine Learning Prediction of Biomarkers from SNPs and of Disease Risk from Biomarkers in the UK Biobank. Genes. 2021; 12: 991.
[90]
Lewis CM, Vassos E. Polygenic risk scores: from research tools to clinical instruments. Genome Medicine. 2020; 12: 44.
[91]
Axelrud LK, Santoro ML, Pine DS, Talarico F, Gadelha A, Manfro GG, et al. Polygenic Risk Score for Alzheimer’s Disease: Implications for Memory Performance and Hippocampal Volumes in Early Life. The American Journal of Psychiatry. 2018; 175: 555–563.
[92]
Aung N, Vargas JD, Yang C, Cabrera CP, Warren HR, Fung K, et al. Genome-Wide Analysis of Left Ventricular Image-Derived Phenotypes Identifies Fourteen Loci Associated With Cardiac Morphogenesis and Heart Failure Development. Circulation. 2019; 140: 1318–1330.
[93]
Thibaut A, Schiff N, Giacino J, Laureys S, Gosseries O. Therapeutic interventions in patients with prolonged disorders of consciousness. The Lancet. Neurology. 2019; 18: 600–614.
[94]
Padua L, Cuccagna C, Pazzaglia C. Novel sensory paradigms for neuromodulation in disorders of consciousness in traumatic brain injury. Current Opinion in Neurology. 2019; 32: 844–849.
[95]
Barwari T, Joshi A, Mayr M. MicroRNAs in Cardiovascular Disease. Journal of the American College of Cardiology. 2016; 68: 2577–2584.
[96]
Joshi A, Rienks M, Theofilatos K, Mayr M. Systems biology in cardiovascular disease: a multiomics approach. Nature Reviews. Cardiology. 2021; 18: 313–330.
[97]
Cristell N, Cianflone D, Durante A, Ammirati E, Vanuzzo D, Banfi M, et al. High-sensitivity C-reactive protein is within normal levels at the very onset of first ST-segment elevation acute myocardial infarction in 41% of cases: a multiethnic case-control study. Journal of the American College of Cardiology. 2011; 58: 2654–2661.
[98]
Pedicino D, Severino A, Ucci S, Bugli F, Flego D, Giglio AF, et al. Epicardial adipose tissue microbial colonization and inflammasome activation in acute coronary syndrome. International Journal of Cardiology. 2017; 236: 95–99.
[99]
Partida RA, Libby P, Crea F, Jang IK. Plaque erosion: a new in vivo diagnosis and a potential major shift in the management of patients with acute coronary syndromes. European Heart Journal. 2018; 39: 2070–2076.
[100]
Sato K, Kaikita K, Nakayama N, Horio E, Yoshimura H, Ono T, et al. Coronary vasomotor response to intracoronary acetylcholine injection, clinical features, and long-term prognosis in 873 consecutive patients with coronary spasm: analysis of a single-center study over 20 years. Journal of the American Heart Association. 2013; 2: e000227.
[101]
Montone RA, Niccoli G, Fracassi F, Russo M, Gurgoglione F, Cammà G, et al. Patients with acute myocardial infarction and non-obstructive coronary arteries: safety and prognostic relevance of invasive coronary provocative tests. European Heart Journal. 2018; 39: 91–98.
[102]
Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. International Journal of Molecular Sciences. 2019; 20: 3328.
[103]
Kreiner FF, Kraaijenhof JM, von Herrath M, Hovingh GKK, von Scholten BJ. Interleukin 6 in diabetes, chronic kidney disease, and cardiovascular disease: mechanisms and therapeutic perspectives. Expert Review of Clinical Immunology. 2022; 18: 377–389.
[104]
Toldo S, Mezzaroma E, Buckley LF, Potere N, Di Nisio M, Biondi-Zoccai G, et al. Targeting the NLRP3 inflammasome in cardiovascular diseases. Pharmacology & Therapeutics. 2022; 236: 108053.
[105]
Omerovic E, Citro R, Bossone E, Redfors B, Backs J, Bruns B, et al. Pathophysiology of Takotsubo syndrome - a joint scientific statement from the Heart Failure Association Takotsubo Syndrome Study Group and Myocardial Function Working Group of the European Society of Cardiology - Part 2: vascular pathophysiology, gender and sex hormones, genetics, chronic cardiovascular problems and clinical implications. European Journal of Heart Failure. 2022; 24: 274–286.
[106]
Repova K, Aziriova S, Krajcirovicova K, Simko F. Cardiovascular therapeutics: A new potential for anxiety treatment? Medicinal Research Reviews. 2022; 42: 1202–1245.
[107]
Dang G, Li T, Yang D, Yang G, Du X, Yang J, et al. T lymphocyte-derived extracellular vesicles aggravate abdominal aortic aneurysm by promoting macrophage lipid peroxidation and migration via pyruvate kinase muscle isozyme 2. Redox Biology. 2022; 50: 102257.
[108]
Thota LNR, Chignalia AZ. The role of the glypican and syndecan families of heparan sulfate proteoglycans in cardiovascular function and disease. American Journal of Physiology. Cell Physiology. 2022; 323: C1052–C1060.
[109]
Wang Q, Chi L. The Alterations and Roles of Glycosaminoglycans in Human Diseases. Polymers. 2022; 14: 5014.
[110]
Worssam MD, Lambert J, Oc S, Taylor JCK, Taylor AL, Dobnikar L, et al. Cellular mechanisms of oligoclonal vascular smooth muscle cell expansion in cardiovascular disease. Cardiovascular Research. 2023; 119: 1279–1294.
[111]
Romo M, López-Vicario C, Pérez-Romero N, Casulleras M, Martínez-Puchol AI, Sánchez B, et al. Small fragments of hyaluronan are increased in individuals with obesity and contribute to low-grade inflammation through TLR-mediated activation of innate immune cells. International Journal of Obesity (2005). 2022; 46: 1960–1969.
[112]
Pinheiro-de-Sousa I, Fonseca-Alaniz MH, Teixeira SK, Rodrigues MV, Krieger JE. Uncovering emergent phenotypes in endothelial cells by clustering of surrogates of cardiovascular risk factors. Scientific Reports. 2022; 12: 1372.
[113]
Chong JH, Ghosh AK. Coronary Artery Vasospasm Induced by 5-fluorouracil: Proposed Mechanisms, Existing Management Options and Future Directions. Interventional Cardiology (London, England). 2019; 14: 89–94.
[114]
Shimokawa H. Reactive oxygen species in cardiovascular health and disease: special references to nitric oxide, hydrogen peroxide, and Rho-kinase. Journal of Clinical Biochemistry and Nutrition. 2020; 66: 83–91.
[115]
Hlatky MA, Greenland P, Arnett DK, Ballantyne CM, Criqui MH, Elkind MSV, et al. Criteria for evaluation of novel markers of cardiovascular risk: a scientific statement from the American Heart Association. Circulation. 2009; 119: 2408–2416.
[116]
Collins GS, Moons KGM. Reporting of artificial intelligence prediction models. Lancet (London, England). 2019; 393: 1577–1579.
[117]
Nillmani, Jain PK, Sharma N, Kalra MK, Viskovic K, Saba L, et al. Four Types of Multiclass Frameworks for Pneumonia Classification and Its Validation in X-ray Scans Using Seven Types of Deep Learning Artificial Intelligence Models. Diagnostics (Basel, Switzerland). 2022; 12: 652.
[118]
Araki T, Jain PK, Suri HS, Londhe ND, Ikeda N, El-Baz A, et al. Stroke Risk Stratification and its Validation using Ultrasonic Echolucent Carotid Wall Plaque Morphology: A Machine Learning Paradigm. Computers in Biology and Medicine. 2017; 80: 77–96.
[119]
Jain PK, Sharma N, Saba L, Paraskevas KI, Kalra MK, Johri A, et al. Unseen Artificial Intelligence-Deep Learning Paradigm for Segmentation of Low Atherosclerotic Plaque in Carotid Ultrasound: A Multicenter Cardiovascular Study. Diagnostics (Basel, Switzerland). 2021; 11: 2257.
[120]
Blankenberg S, Rupprecht HJ, Bickel C, Peetz D, Hafner G, Tiret L, et al. Circulating cell adhesion molecules and death in patients with coronary artery disease. Circulation. 2001; 104: 1336–1342.
[121]
Fernández-Sanlés A, Sayols-Baixeras S, Subirana I, Degano IR, Elosua R. Association between DNA methylation and coronary heart disease or other atherosclerotic events: A systematic review. Atherosclerosis. 2017; 263: 325–333.
[122]
Rafieian-Kopaei M, Setorki M, Doudi M, Baradaran A, Nasri H. Atherosclerosis: process, indicators, risk factors and new hopes. International Journal of Preventive Medicine. 2014; 5: 927–946.
[123]
Tabas I. Apoptosis and plaque destabilization in atherosclerosis: the role of macrophage apoptosis induced by cholesterol. Cell Death and Differentiation. 2004; 11: S12–S16.
[124]
Frostegård J, Haegerstrand A, Gidlund M, Nilsson J. Biologically modified LDL increases the adhesive properties of endothelial cells. Atherosclerosis. 1991; 90: 119–126.
[125]
Winston PH. Artificial intelligence. Addison-Wesley Longman Publishing Co., Inc.: USA. 1992.
[126]
Ramesh AN, Kambhampati C, Monson JRT, Drew PJ. Artificial intelligence in medicine. Annals of the Royal College of Surgeons of England. 2004; 86: 334–338.
[127]
Biswas M, Kuppili V, Saba L, Edla DR, Suri HS, Cuadrado-Godia E, et al. State-of-the-art review on deep learning in medical imaging. Frontiers in Bioscience (Landmark Edition). 2019; 24: 392–426.
[128]
Khanna NN, Maindarkar M, Puvvula A, Paul S, Bhagawati M, Ahluwalia P, et al. Vascular Implications of COVID-19: Role of Radiological Imaging, Artificial Intelligence, and Tissue Characterization: A Special Report. Journal of Cardiovascular Development and Disease. 2022; 9: 268.
[129]
Saba L, Agarwal M, Patrick A, Puvvula A, Gupta SK, Carriero A, et al. Six artificial intelligence paradigms for tissue characterisation and classification of non-COVID-19 pneumonia against COVID-19 pneumonia in computed tomography lungs. International Journal of Computer Assisted Radiology and Surgery. 2021; 16: 423–434.
[130]
Suri JS, Puvvula A, Majhail M, Biswas M, Jamthikar AD, Saba L, et al. Integration of cardiovascular risk assessment with COVID-19 using artificial intelligence. Reviews in Cardiovascular Medicine. 2020; 21: 541–560.
[131]
Jamthikar A, Gupta D, Khanna NN, Araki T, Saba L, Nicolaides A, et al. A Special Report on Changing Trends in Preventive Stroke/Cardiovascular Risk Assessment Via B-Mode Ultrasonography. Current Atherosclerosis Reports. 2019; 21: 25.
[132]
Jamthikar A, Gupta D, Khanna NN, Saba L, Laird JR, Suri JS. Cardiovascular/stroke risk prevention: A new machine learning framework integrating carotid ultrasound image-based phenotypes and its harmonics with conventional risk factors. Indian Heart Journal. 2020; 72: 258–264.
[133]
Jamthikar AD, Gupta D, Saba L, Khanna NN, Viskovic K, Mavrogeni S, et al. Artificial intelligence framework for predictive cardiovascular and stroke risk assessment models: A narrative review of integrated approaches using carotid ultrasound. Computers in Biology and Medicine. 2020; 126: 104043.
[134]
Jamthikar A, Gupta D, Saba L, Khanna NN, Araki T, Viskovic K, et al. Cardiovascular/stroke risk predictive calculators: a comparison between statistical and machine learning models. Cardiovascular Diagnosis and Therapy. 2020; 10: 919–938.
[135]
Panayides AS, Pattichis MS, Leandrou S, Pitris C, Constantinidou A, Pattichis CS. Radiogenomics for Precision Medicine With a Big Data Analytics Perspective. IEEE Journal of Biomedical and Health Informatics. 2019; 23: 2063–2079.
[136]
Quazi S. Artificial intelligence and machine learning in precision and genomic medicine. Medical Oncology (Northwood, London, England). 2022; 39: 120.
[137]
Saba L, Dey N, Ashour AS, Samanta S, Nath SS, Chakraborty S, et al. Automated stratification of liver disease in ultrasound: An online accurate feature classification paradigm. Computer Methods and Programs in Biomedicine. 2016; 130: 118–134.
[138]
Skandha SS, Nicolaides A, Gupta SK, Koppula VK, Saba L, Johri AM, et al. A hybrid deep learning paradigm for carotid plaque tissue characterization and its validation in multicenter cohorts using a supercomputer framework. Computers in Biology and Medicine. 2022; 141: 105131.
[139]
Naseer A, Rani M, Naz S, Razzak MI, Imran M, Xu G. Refining Parkinson’s neurological disorder identification through deep transfer learning. Neural Computing and Applications. 2020; 32: 839–854.
[140]
Suri JS, Bhagawati M, Paul S, Protogeron A, Sfikakis PP, Kitas GD, et al. Understanding the bias in machine learning systems for cardiovascular disease risk assessment: The first of its kind review. Computers in Biology and Medicine. 2022; 142: 105204.
[141]
Banchhor SK, Araki T, Londhe ND, Ikeda N, Radeva P, Elbaz A, et al. Five multiresolution-based calcium volume measurement techniques from coronary IVUS videos: A comparative approach. Computer Methods and Programs in Biomedicine. 2016; 134: 237–258.
[142]
Banchhor SK, Londhe ND, Saba L, Radeva P, Laird JR, Suri JS. Relationship between Automated Coronary Calcium Volumes and a Set of Manual Coronary Lumen Volume, Vessel Volume and Atheroma Volume in Japanese Diabetic Cohort. Journal of Clinical and Diagnostic Research: JCDR. 2017; 11: TC09–TC14.
[143]
Tarkin JM, Dweck MR, Evans NR, Takx RA, Brown AJ, Tawakol A, et al. Imaging Atherosclerosis. Circulation Research. 2016; 118: 750–69.
[144]
Jo T, Nho K, Saykin AJ. Deep Learning in Alzheimer’s Disease: Diagnostic Classification and Prognostic Prediction Using Neuroimaging Data. Frontiers in Aging Neuroscience. 2019; 11: 220.
[145]
Su J, Hu J, Jiang J, Xie J, Yang Y, He B, et al. Extraction of risk factors for cardiovascular diseases from Chinese electronic medical records. Computer Methods and Programs in Biomedicine. 2019; 172: 1–10.
[146]
Goehring T, Keshavarzi M, Carlyon RP, Moore BCJ. Using recurrent neural networks to improve the perception of speech in non-stationary noise by people with cochlear implants. The Journal of the Acoustical Society of America. 2019; 146: 705.
[147]
Bandyopadhyay SK, Dutta S. Stacked bi-directional LSTM layer based model for prediction of possible heart disease during lockdown period of COVID-19: bidirectional LSTM. Journal of Advanced Research in Medical Science & Technology. 2020; 7: 10–14.
[148]
Ramaraj E. A novel deep learning based gated recurrent unit with extreme learning machine for electrocardiogram (ECG) signal recognition. Biomedical Signal Processing and Control. 2021; 68: 102779.
[149]
Zhang X, Li R, Dai H, Liu Y, Zhou B, Wang Z. Localization of myocardial infarction with multi-lead bidirectional gated recurrent unit neural network. IEEE Access. 2019; 7: 161152–161166.
[150]
Acharya UR, Joseph KP, Kannathal N, Min LC, Suri JS. Heart rate variability. Advances in Cardiac Signal Processing. 2007; 121–165.
[151]
Coto-Jiménez M. Improving Post-Filtering of Artificial Speech Using Pre-Trained LSTM Neural Networks. Biomimetics (Basel, Switzerland). 2019; 4: 39.
[152]
Graves A, Liwicki M, Fernández S, Bertolami R, Bunke H, Schmidhuber J. A novel connectionist system for unconstrained handwriting recognition. IEEE Transactions on Pattern Analysis and Machine Intelligence. 2009; 31: 855–868.
[153]
Suri JS, Paul S, Maindarkar MA, Puvvula A, Saxena S, Saba L, et al. Cardiovascular/Stroke Risk Stratification in Parkinson’s Disease Patients Using Atherosclerosis Pathway and Artificial Intelligence Paradigm: A Systematic Review. Metabolites. 2022; 12: 312.
[154]
Davies RW, Dandona S, Stewart AFR, Chen L, Ellis SG, Tang WHW, et al. Improved prediction of cardiovascular disease based on a panel of single nucleotide polymorphisms identified through genome-wide association studies. Circulation. Cardiovascular Genetics. 2010; 3: 468–474.
[155]
Valavanis IK, Mougiakakou SG, Grimaldi KA, Nikita KS. A multifactorial analysis of obesity as CVD risk factor: use of neural network based methods in a nutrigenetics context. BMC Bioinformatics. 2010; 11: 453.
[156]
Mohammad Beigi M, Behjati M, Mohabatkar H. Prediction of metalloproteinase family based on the concept of Chou’s pseudo amino acid composition using a machine learning approach. Journal of Structural and Functional Genomics. 2011; 12: 191–197.
[157]
Montañez CAC, Fergus P, Hussain A, Al-Jumeily D, Abdulaimma B, Hind J, et al. ‘Machine learning approaches for the prediction of obesity using publicly available genetic profiles’, 2017 International Joint Conference on Neural Networks (IJCNN). IEEE. 2017.
[158]
Zhao J, Feng Q, Wu P, Lupu RA, Wilke RA, Wells QS, et al. Learning from Longitudinal Data in Electronic Health Record and Genetic Data to Improve Cardiovascular Event Prediction. Scientific Reports. 2019; 9: 717.
[159]
Schiano C, Franzese M, Geraci F, Zanfardino M, Maiello C, Palmieri V, et al. Machine Learning and Bioinformatics Framework Integration to Potential Familial DCM-Related Markers Discovery. Genes. 2021; 12: 1946.
[160]
Yang NI, Yeh CH, Tsai TH, Chou YJ, Hsu PWC, Li CH, et al. Artificial Intelligence-Assisted Identification of Genetic Factors Predisposing High-Risk Individuals to Asymptomatic Heart Failure. Cells. 2021; 10: 2430.
[161]
Akbarzadeh M, Alipour N, Moheimani H, Zahedi AS, Hosseini-Esfahani F, Lanjanian H, et al. Evaluating machine learning-powered classification algorithms which utilize variants in the GCKR gene to predict metabolic syndrome: Tehran Cardio-metabolic Genetics Study. Journal of Translational Medicine. 2022; 20: 164.
[162]
Kesar A, Baluch A, Barber O, Hoffmann H, Jovanovic M, Renz D, et al. Actionable absolute risk prediction of atherosclerotic cardiovascular disease based on the UK Biobank. PloS One. 2022; 17: e0263940.
[163]
Pirruccello JP, Chaffin MD, Chou EL, Fleming SJ, Lin H, Nekoui M, et al. Deep learning enables genetic analysis of the human thoracic aorta. Nature Genetics. 2022; 54: 40–51.
[164]
Steinfeldt J, Buergel T, Loock L, Kittner P, Ruyoga G, Zu Belzen JU, et al. Neural network-based integration of polygenic and clinical information: development and validation of a prediction model for 10-year risk of major adverse cardiac events in the UK Biobank cohort. The Lancet. Digital Health. 2022; 4: e84–e94.
[165]
Lee T, Hwang S, Seo DM, Shin HC, Kim HS, Kim JY, et al. Identification of Cardiovascular Disease-Related Genes Based on the Co-Expression Network Analysis of Genome-Wide Blood Transcriptome. Cells. 2022; 11: 2867.
[166]
Libiseller-Egger J, Phelan JE, Attia ZI, Benavente ED, Campino S, Friedman PA, et al. Deep learning-derived cardiovascular age shares a genetic basis with other cardiac phenotypes. Scientific Reports. 2022; 12: 22625.
[167]
Kang L, Zhao Q, Jiang K, Yu X, Chao H, Yin L, et al. Uncovering potential diagnostic biomarkers of acute myocardial infarction based on machine learning and analyzing its relationship with immune cells. BMC Cardiovascular Disorders. 2023; 23: 2.
[168]
Venkat V, Abdelhalim H, DeGroat W, Zeeshan S, Ahmed Z. Investigating genes associated with heart failure, atrial fibrillation, and other cardiovascular diseases, and predicting disease using machine learning techniques for translational research and precision medicine. Genomics. 2023; 115: 110584.
[169]
Espejo PG, Ventura S, Herrera F. A survey on the application of genetic programming to classification. IEEE Transactions on Systems, Man, and Cybernetics, Part C (Applications and Reviews). 2009; 40: 121–144.
[170]
Rudin C. Stop Explaining Black Box Machine Learning Models for High Stakes Decisions and Use Interpretable Models Instead. Nature Machine Intelligence. 2019; 1: 206–215.
[171]
Yang G, Ye Q, Xia J. Unbox the black-box for the medical explainable AI via multi-modal and multi-centre data fusion: A mini-review, two showcases and beyond. An International Journal on Information Fusion. 2022; 77: 29–52.
[172]
Khanna NN, Maindarkar MA, Viswanathan V, Fernandes JFE, Paul S, Bhagawati M, et al. Economics of Artificial Intelligence in Healthcare: Diagnosis vs. Treatment. Healthcare (Basel, Switzerland). 2022; 10: 2493.
[173]
Samek W, Montavon G, Lapuschkin S, Anders CJ, Müller KR. Explaining deep neural networks and beyond: A review of methods and applications. Proceedings of the IEEE. 2021; 109: 247–278.
[174]
Frierson J, Si D. ‘Who’s next: Evaluating attrition with machine learning algorithms and survival analysis’, Big Data–BigData 2018: 7th International Congress, Held as Part of the Services Conference Federation, SCF 2018. Seattle, WA, USA, June 25–30, 2018. Springer International Publishing. 2018.
[175]
Guleria P, Naga Srinivasu P, Ahmed S, Almusallam N, Alarfaj FK. XAI framework for cardiovascular disease prediction using classification techniques. Electronics. 2022; 11: 4086.
[176]
Clough JR, Oksuz I, Puyol-Antón E, Ruijsink B, King AP, Schnabel JA. ‘Global and local interpretability for cardiac MRI classification’, International Conference on Medical Image Computing and Computer-Assisted Intervention. Springer International Publishing: Cham. 2019.
[177]
DeGrave AJ, Janizek JD, Lee SI. AI for radiographic COVID-19 detection selects shortcuts over signal. Nature Machine Intelligence. 2021; 3: 610–619.
[178]
Saba L, Banchhor SK, Suri HS, Londhe ND, Araki T, Ikeda N, et al. Accurate cloud-based smart IMT measurement, its validation and stroke risk stratification in carotid ultrasound: A web-based point-of-care tool for multicenter clinical trial. Computers in Biology and Medicine. 2016; 75: 217–234.
[179]
Saba L, Banchhor SK, Londhe ND, Araki T, Laird JR, Gupta A, et al. Web-based accurate measurements of carotid lumen diameter and stenosis severity: An ultrasound-based clinical tool for stroke risk assessment during multicenter clinical trials. Computers in Biology and Medicine. 2017; 91: 306–317.
[180]
Singh P. Deploy Machine Learning Models to Production. Springer: Cham, Switzerland. 2021.
[181]
Ivanovic M, Autexier S, Kokkonidis M. ‘AI approaches in processing and using data in personalized medicine’, European Conference on Advances in Databases and Information Systems. Springer International Publishing: Cham. 2022.
[182]
Hajiyan H, Ebrahimi M. ‘Multi-scale local explanation approach for image analysis using model-agnostic Explainable Artificial Intelligence (XAI)’, Medical Imaging 2023: Digital and Computational Pathology. SPIE. 2023.
[183]
Gulum MA, Trombley CM, Kantardzic M. A review of explainable deep learning cancer detection models in medical imaging. Applied Sciences. 2021; 11: 4573.
[184]
Alicioglu G, Sun B. A survey of visual analytics for Explainable Artificial Intelligence methods. Computers & Graphics. 2022; 102: 502–520.
[185]
Gozes Ophir, Maayan Frid-Adar, Hayit Greenspan, Patrick D. Browning, Huangqi Zhang, Wenbin Ji, et al. Rapid ai development cycle for the coronavirus (covid-19) pandemic: Initial results for automated detection & patient monitoring using deep learning ct image analysis. arXiv preprint arXiv:2003.05037 (2020). (preprint)
[186]
Sattar Y, Ullah W, Rauf H, Virk HUH, Yadav S, Chowdhury M, et al. COVID-19 cardiovascular epidemiology, cellular pathogenesis, clinical manifestations and management. International Journal of Cardiology. Heart & Vasculature. 2020; 29: 100589.
[187]
Agarwal M, Saba L, Gupta SK, Johri AM, Khanna NN, Mavrogeni S, et al. Wilson disease tissue classification and characterization using seven artificial intelligence models embedded with 3D optimization paradigm on a weak training brain magnetic resonance imaging datasets: a supercomputer application. Medical & Biological Engineering & Computing. 2021; 59: 511–533.
[188]
Molinari F, Zeng G, Suri JS. Intima-media thickness: setting a standard for a completely automated method of ultrasound measurement. IEEE Transactions on Ultrasonics, Ferroelectrics, and Frequency Control. 2010; 57: 1112–1124.
[189]
Jain PK, Sharma N, Saba L, Paraskevas KI, Kalra MK, Johri A, et al. Automated deep learning-based paradigm for high-risk plaque detection in B-mode common carotid ultrasound scans: an asymptomatic Japanese cohort study. International Angiology: a Journal of the International Union of Angiology. 2022; 41: 9–23.
[190]
Puzantian H, Akers SR, Oldland G, Javaid K, Miller R, Ge Y, et al. Circulating Dephospho-Uncarboxylated Matrix Gla-Protein Is Associated With Kidney Dysfunction and Arterial Stiffness. American Journal of Hypertension. 2018; 31: 988–994.
[191]
Suri JS, Bhagawati M, Paul S, Protogerou AD, Sfikakis PP, Kitas GD, et al. A Powerful Paradigm for Cardiovascular Risk Stratification Using Multiclass, Multi-Label, and Ensemble-Based Machine Learning Paradigms: A Narrative Review. Diagnostics (Basel, Switzerland). 2022; 12: 722.
[192]
Li, Qi, Fushun Liu, Bin Wang, D. Z. Liu, and Zhenghua Qian. A novel physics-informed framework for reconstruction of structural defects. Applied Mathematics and Mechanics. 2022; 43: 1717–1730.
[193]
Mantella LE, Colledanchise KN, Hétu MF, Feinstein SB, Abunassar J, Johri AM. Carotid intraplaque neovascularization predicts coronary artery disease and cardiovascular events. European Heart Journal. Cardiovascular Imaging. 2019; 20: 1239–1247.
[194]
Krittanawong C, Johnson KW, Hershman SG, Tang WW. Big data, artificial intelligence, and cardiovascular precision medicine. Expert Review of Precision Medicine and Drug Development. 2018; 3: 305–317.
[195]
Arena R, Ozemek C, Laddu D, Campbell T, Rouleau CR, Standley R, et al. Applying Precision Medicine to Healthy Living for the Prevention and Treatment of Cardiovascular Disease. Current Problems in Cardiology. 2018; 43: 448–483.
[196]
Westerlund AM, Hawe JS, Heinig M, Schunkert H. Risk Prediction of Cardiovascular Events by Exploration of Molecular Data with Explainable Artificial Intelligence. International Journal of Molecular Sciences. 2021; 22: 10291.
[197]
Alimadadi A, Manandhar I, Aryal S, Munroe PB, Joe B, Cheng X. Machine learning-based classification and diagnosis of clinical cardiomyopathies. Physiological Genomics. 2020; 52: 391–400.
[198]
Saba L, Sanagala SS, Gupta SK, Koppula VK, Johri AM, Sharma AM, et al. Ultrasound-based internal carotid artery plaque characterization using deep learning paradigm on a supercomputer: a cardiovascular disease/stroke risk assessment system. The International Journal of Cardiovascular Imaging. 2021; 37: 1511–1528.
[199]
Saba L, Sanagala SS, Gupta SK, Koppula VK, Johri AM, Khanna NN, et al. Multimodality carotid plaque tissue characterization and classification in the artificial intelligence paradigm: a narrative review for stroke application. Annals of Translational Medicine. 2021; 9: 1206.
[200]
Staub D, Patel MB, Tibrewala A, Ludden D, Johnson M, Espinosa P, et al. Vasa vasorum and plaque neovascularization on contrast-enhanced carotid ultrasound imaging correlates with cardiovascular disease and past cardiovascular events. Stroke. 2010; 41: 41–47.
[201]
Gruson D, Bernardini S, Dabla PK, Gouget B, Stankovic S. Collaborative AI and Laboratory Medicine integration in precision cardiovascular medicine. Clinica Chimica Acta; International Journal of Clinical Chemistry. 2020; 509: 67–71.
[202]
Parry, Helen M., Harshal Deshmukh, Daniel Levin, Natalie Van Zuydam, Douglas HJ Elder, Andrew D. Morris, et al. Both high and low HbA1c predict incident heart failure in type 2 diabetes mellitus. Circulation: Heart Failure. 2015; 8: 236–242.
[203]
R. M. Conroy, K. Pyörälä, A. P. Fitzgerald, S. Sans, A. Menotti, G. De Backer, et al. Estimation of ten-year risk of fatal cardiovascular disease in Europe: the SCORE project. European Heart Journal. 2003; 24: 987–1003.
[204]
Vlachopoulos C, Aznaouridis K, Ioakeimidis N, Rokkas K, Vasiliadou C, Alexopoulos N, et al. Unfavourable endothelial and inflammatory state in erectile dysfunction patients with or without coronary artery disease. European Heart Journal. 2006; 27: 2640–2648.
[205]
Gandaglia G, Briganti A, Jackson G, Kloner RA, Montorsi F, Montorsi P, et al. A systematic review of the association between erectile dysfunction and cardiovascular disease. European Urology. 2014; 65: 968–978.
[206]
Suri JS, Agarwal S, Gupta S, Puvvula A, Viskovic K, Suri N, et al. Systematic Review of Artificial Intelligence in Acute Respiratory Distress Syndrome for COVID-19 Lung Patients: A Biomedical Imaging Perspective. IEEE Journal of Biomedical and Health Informatics. 2021; 25: 4128–4139.
[207]
Paul S, Maindarkar M, Saxena S, Saba L, Turk M, Kalra M, et al. Bias Investigation in Artificial Intelligence Systems for Early Detection of Parkinson’s Disease: A Narrative Review. Diagnostics (Basel, Switzerland). 2022; 12: 166.
[208]
Suri J S, Agarwal S, Jena B, Saxena S, El-Baz A, Agarwal V, et al. Five strategies for bias estimation in artificial intelligence-based hybrid deep learning for acute respiratory distress syndrome COVID-19 lung infected patients using AP (ai) Bias 2.0: a systematic review. IEEE Transactions on Instrumentation and Measurement. 2022.
[209]
Acharya UR, Mookiah MRK, Vinitha Sree S, Yanti R, Martis RJ, Saba L, et al. Evolutionary algorithm-based classifier parameter tuning for automatic ovarian cancer tissue characterization and classification. Ultraschall in Der Medizin (Stuttgart, Germany: 1980). 2014; 35: 237–245.
[210]
Shrivastava VK, Londhe ND, Sonawane RS, Suri JS. Reliable and accurate psoriasis disease classification in dermatology images using comprehensive feature space in machine learning paradigm. Expert Systems with Applications. 2015; 42: 6184–6195.
[211]
Araki T, Ikeda N, Shukla D, Jain PK, Londhe ND, Shrivastava VK, et al. PCA-based polling strategy in machine learning framework for coronary artery disease risk assessment in intravascular ultrasound: A link between carotid and coronary grayscale plaque morphology. Computer Methods and Programs in Biomedicine. 2016; 128: 137–158.
[212]
Galbraith RF. A note on graphical presentation of estimated odds ratios from several clinical trials. Statistics in Medicine. 1988; 7: 889–894.
[213]
Johri AM, Lajkosz KA, Grubic N, Islam S, Li TY, Simpson CS, et al. Maximum plaque height in carotid ultrasound predicts cardiovascular disease outcomes: a population-based validation study of the American society of echocardiography’s grade II-III plaque characterization and protocol. The International Journal of Cardiovascular Imaging. 2021; 37: 1601–1610.
[214]
Cuadrado-Godia E, Jamthikar AD, Gupta D, Khanna NN, Araki T, Maniruzzaman M, et al. Ranking of stroke and cardiovascular risk factors for an optimal risk calculator design: Logistic regression approach. Computers in Biology and Medicine. 2019; 108: 182–195.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top