Next Article in Journal
Transoral Robotic Surgery for the Salvage of Primarily Irradiated Oropharyngeal Squamous Cell Carcinomas Recurring at the Base of the Tongue: A Small Monoinstitutional Series
Previous Article in Journal
Microsatellite Instability and Myometrial Infiltration in Low-Grade Endometrial Cancer: A Focus on MMR Heterodimer Dysfunction by a Retrospective Multicentric Italian Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Cardiovascular Complications Are Increased in Inflammatory Bowel Disease: A Path Toward Achievement of a Personalized Risk Estimation

by
Vito Annese
1,2,*,
Maria Laura Annunziata
1,
Guglielmo Albertini Petroni
1,
Emanuele Orlando
1,
Sofia Cinque
1,3,
Marzio Parisi
1,4,
Paolo Biamonte
1,2,
Giuseppe Dell’Anna
1,2,
Anna Latiano
5 and
Serenella Castelvecchio
6
1
Unit of Gastroenterology, I.R.C.C.S. San Donato Policlinic, San Donato Milanese, 20097 Milan, Italy
2
Department of Gastroenterology, University Vita & Salute, San Raffaele, Via Olgettina 60, 20132 Milan, Italy
3
Gastroenterology Unit, Department of Clinical Medicine and Surgery, University Federico II, Via Pansini 5, 80131 Naples, Italy
4
Department of Internal Medicine and Medical Therapy, University of Pavia, Via Adolfo Ferrata 9, 27100 Pavia, Italy
5
Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, 71013 Foggia, Italy
6
Unit of Cardiovascular Prevention, I.R.C.C.S. San Donato Policlinic, San Donato Milanese, 20097 Milan, Italy
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2025, 15(9), 418; https://doi.org/10.3390/jpm15090418
Submission received: 21 July 2025 / Revised: 19 August 2025 / Accepted: 26 August 2025 / Published: 2 September 2025
(This article belongs to the Section Disease Biomarkers)

Abstract

Background/Objectives: The global burden of inflammatory bowel diseases (IBDs) continues to rise, with up to 50% of patients experiencing extraintestinal manifestations. Cardiovascular diseases (CVDs) are of particular concern, ranking as the second leading cause of mortality in this population. Despite a comparatively lower prevalence of traditional cardiovascular (CV) risk factors, the persistent inflammatory milieu and immune dysregulation inherent to IBD may contribute to heightened CVD risk. In this study, following a review of the current literature, an ongoing prospective trial designed to clarify CV risk profiles in IBD patients is detailed. Methods: A cohort of patients with IBD is being enrolled for comprehensive baseline evaluation of CV risk factors, lifestyle metrics, and disease characteristics. The incidence of major adverse cardiovascular events (MACEs) will be tracked and contrasted with a gender- and age-matched non-IBD cohort over a 2-year follow-up period. In cases of MACE occurrence, a multi-omics analysis—including genomic, proteomic, transcriptomic, and microbiome profiling—will be performed, along with a parallel evaluation in matched IBD controls without MACE. An artificial intelligence (AI) framework will support the analysis of this complex dataset. Results: To date, over 150 patients with IBD have been enrolled, and detailed phenotypic data and biological samples have been collected. Conclusions: We aim to introduce an IBD-specific correction factor for existing CV risk scores upon study completion. This is particularly relevant for individuals under 40 years of age, who are often inadequately assessed by current risk stratification models

1. Introduction

Crohn’s disease (CD) and ulcerative colitis (UC), the principal forms of inflammatory bowel disease (IBD), are showing a rising global prevalence, affecting an estimated 7 million individuals and placing a substantial strain on healthcare systems [1,2]. Many patients experience progressive intestinal damage, with complications such as hospitalization, surgery, malignancy, and elevated mortality risk [3,4]. Up to 50% of patients also develop extraintestinal manifestations, and there is an increased predisposition toward other immune-mediated disorders [5,6]. Among these comorbidities, cardiovascular disease (CVD) stands out in significance, especially as the aging IBD population sees a growing incidence of CVD-related mortality—now the second leading cause of death [7,8,9].
Despite advances in therapy, approximately one-third of patients with IBD exhibit primary non-response, and nearly half experience secondary loss of response [10]. While CVD remains the foremost cause of death globally [11], patients with IBD are more susceptible to premature atherosclerosis and myocardial infarction, particularly during active disease phases [12]. Alarmingly, conventional CV risk models often fail to capture this elevated risk, suggesting an underestimation in this specific population [13]. A comprehensive systematic review of more than 2 million patients with IBD identified smoking (24.2%) and alcohol use (4.6%) as the leading lifestyle risk factors. The common comorbidities included hypertension (30%), diabetes (14.4%), dyslipidemia (18.4%), pre-existing CVD (22%), and renal impairment (10%) [14].

1.1. IBD and CVD: A Shared Pathogenesis

IBD arises from a multifaceted interaction of genetic predisposition, immune dysregulation, environmental exposures, and dysbiosis of the gut microbiota—culminating in heightened cytokine activity [3,4]. This sustained inflammatory and immune-activated state may foster cardiovascular complications even in the absence of prominent conventional CV risk factors [7,8,12].
Emerging evidence points to shared pathophysiological mechanisms between IBD and CVD, including genetic overlap (e.g., NOD2, CDKN2B, stromelysin, and ApoE polymorphisms), shared environmental triggers like smoking, and convergent immune pathways [15]. Disruption of both intestinal epithelial and vascular endothelial barriers through cytokines like TNF-α may provide a mechanistic link between gut inflammation and vascular pathology [16]. In addition, gut microbiota alterations commonly seen in IBD have been implicated in the pathogenesis of cardiovascular conditions such as atherosclerosis [17].
Although the SCORE2 model developed by the European Society of Cardiology remains the prevailing tool for CV risk estimation, it has not been extensively validated in IBD populations and does not account for the heightened inflammatory burden, unlike the 1.5 correction factor applied in rheumatoid arthritis [18].

1.2. Risk of Major Adverse Cardiovascular Events in Inflammatory Bowel Disease

Multiple studies have demonstrated a significantly higher incidence of MACEs in IBD populations, with increases ranging from 25% to 300% compared with matched controls. A pivotal study by Bernstein et al. involving over 88,000 participants (8072 with IBD) reported an elevated incidence rate ratio (IRR) for ischemic heart disease (IHD) in patients with IBD (IRR: 1.26; 95% CI: 1.11–1.44) [19]. Similarly, a Danish cohort study analyzing 4.5 million individuals, including 28,833 with IBD, found a sharply elevated IHD risk in the first year after diagnosis (IRR: 2.13; 95% CI: 1.91–2.38), with a continued increased risk over time (IRR: 1.22; 95% CI: 1.14–1.30) [20]. A U.S. database study of over 29 million individuals showed a 25% higher likelihood of myocardial infarction among patients with IBD (OR: 1.25; 95% CI: 1.24–1.27) [21]. Standard CV risk algorithms may underestimate this burden, as chronic systemic inflammation is central to pathogenesis of CVD in IBD [22,23]. Elevated risks of stroke and cerebrovascular events have also been documented [24,25], with C-reactive protein (CRP) serving as a potential inflammatory marker [25]. A meta-analysis encompassing cohort and case–control studies confirmed a 21% increase in stroke risk in patients with IBD (OR/RR: 1.21; 95% CI: 1.08–1.34) [26]. Young IBD patients are particularly vulnerable to ischemic strokes, especially during active disease phases [24,25,26]. Additionally, an increased incidence of atrial fibrillation may further amplify the cerebrovascular risk [27].

1.3. Impact of IBD Treatments on Cardiovascular Risk

Certain IBD treatments, particularly corticosteroids, may worsen cardiovascular profiles by inducing hypertension, insulin resistance, and dyslipidemia [28]. However, steroid use often reflects severe disease activity, a known risk factor for CVD itself. Conversely, Rungoe et al. reported that patients with IBD treated with 5-ASA exhibited a reduced risk of IHD (IRR: 1.16 vs. 1.36 for non-users; p = 0.02), with prolonged use linked to even lower risks (IRR: 1.08) [20].
TNF-α inhibitors may provide cardiovascular protection [29]. A large observational study found a lower incidence of acute arterial events (AAEs) among TNF inhibitor users (HR: 0.79), with a similar risk reduction for recurrent events (HR: 0.75) [30]. Thiopurines have shown similar protective effects.
Data on the cardiovascular safety of newer biological therapies available, such as vedolizumab, ustekinumab, mirikizumab, and risankizumab, remain limited; however, no concerning safety signals related to AAEs have been identified thus far.
Janus kinase (JAK) inhibitors, including tofacitinib, filgotinib, and upadacitinib, are among the most recent agents approved for moderate to severe ulcerative colitis and, in the case of upadacitinib, also Crohn’s disease. Clinical trials have noted reversible elevations in serum lipids, including total cholesterol, LDL, and HDL, during early treatment with tofacitinib [31]. Hypercholesterolemia is a known contributor to cardiovascular events, and concerns have been raised regarding MACEs associated with this drug class. The ORAL Surveillance trial, which enrolled rheumatoid arthritis (RA) patients aged ≥50 with pre-existing cardiovascular risk factors, found a higher incidence of MACEs in those treated with tofacitinib (5 or 10 mg twice daily) compared with those on TNF inhibitors (HR: 1.33; 95% CI: 0.91–1.94) [32].
Based on recommendations from the Pharmacovigilance Risk Assessment Committee (PRAC), the European Medicines Agency’s Committee for Medicinal Products for Human Use (CHMP), has advised limiting the use of JAK inhibitors in patients with elevated cardiovascular risk, reserving them for cases with no appropriate alternatives. Nonetheless, a meta-analysis reviewing the safety of JAK inhibitors across IBD and other immune-mediated inflammatory diseases (IMIDs) reported 30 studies assessing MACEs in 32,765 treated individuals (17 on tofacitinib, 6 on upadacitinib, 4 on baricitinib, and 3 on filgotinib), with a pooled incidence rate of 0.67 per 100 patient-years. The relative risk of MACEs was estimated at 1.07 (95% CI: 0.56–2.03) in the analysis of 22 controlled trials [33].
The following tables illustrate
Established cardiovascular risk factors (Table 1);
Shared biological mechanisms that may drive IBD and cardiovascular disease (Table 1);
Treatment-related cardiovascular risks (Table 2);
Summary of key studies conducted on this topic (Appendix A) [34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71].
Table 1. Risk factors for IBD and cardiovascular diseases.
Table 1. Risk factors for IBD and cardiovascular diseases.
Traditional CV Risk FactorsRisk Factors for IBDShared Risk Factors for
IBD and CVDs
Specific Risk Factors in IBD for CVDs
ObesityGenetic factorsDysbiosisInflammatory cytokines
HypertensionDysbiosisGenetic factorsOxidative stress
AgeSmoke (CD)Smoke (CD)Hypercoagulability
SmokeDiet? DietEndothelial dysfunction
Diabetes? No breast feeding? Stress
Dyslipidemia? Antibiotics abuse
Family history of IHD or strokeFamily history of IBD
? Stress
? Diet
IHD = ischemic heart disease; CD = Crohn’s disease. ? = conflicting data
Table 2. Impact of IBD therapies on main cardiovascular risk.
Table 2. Impact of IBD therapies on main cardiovascular risk.
IBD MedicationsPotential CV Effect
5-Aminosalicylic acid↓ Inflammation, ↓ platelet activation
↓ CV complications
Corticosteroids ↑ CV Risk (? proxy of more severe IBD)
Thiopurines↓ Inflammation, ↓ atherosclerosis and arterial events
Anti-TNFα↓ Inflammation, ↓ atherosclerosis and arterial events
Vedolizumab↓ Inflammation, limited data, no increased CV risk
Ustekinumab↓ Inflammation, limited data, no increased CV risk
IL-23 inhibitors↓ Inflammation, limited data, no increased CV risk
Anti-JAK↓ Inflammation, ↑ CV risk in RA
SP1 Modulators↓ Inflammation, limited data, no increased CV risk
RA = rheumatoid arthritis. ? = conflicting data. ↑ = increased. ↓ = reduced.

1.4. IBD and MACEs: Risk Prediction

There remains a critical need for robust assessment of MACE incidence in IBD, therapy-related risks and benefits, lifestyle influences, and disease activity (Appendix A) [34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71]. We have launched a prospective study involving a well-characterized IBD cohort, matched with controls by age and gender. Multi-omics profiling and AI-based analyses will be applied to refine traditional CV risk stratification and uncover novel biomarkers.

1.5. Dissecting the Complexity of CV Risk in IBD Patients

As summarized in the previous sections, there is still an unmet need to evaluate the real amount of CV risk in patients with IBD because of the lack of prospective trials, and the limitation of the available CV risk scores designed for an older background population compared to the patients with IBD. In addition, there is a lack of a systematic evaluation of other possible shared risk factors, such as genetic polymorphisms or dysbiosis, and more importantly, the need to evaluate the full systematic dataset with a hypothesis-free powerful methodology, such as AI-driven algorithms. For these reasons, an ongoing study designed to overcome these limitations is described here.

2. Materials and Methods

2.1. Study Population

Our first aim was to prospectively enroll a large cohort of IBD patients in the Unit of Gastroenterology of the Research Hospital San Donato Policlinic, San Donato Milanese (Mi), for 1 year to be compared with an age/gender-matched cohort under follow-up at the Unit of Cardiology of the same Institution. To this end, a set of specialized, privacy-preserving large language models (LLMs), coordinated by an agentic framework provided by Agora Labs [72], will be applied to different types of data sources containing records of patients under follow-up at the San Donato Policlinic to maximize enrollment. Specifically, the LLMs will extract clinical findings, diagnoses, disease activity indicators, family and social history items, past and current treatments, and other clinical data from all kinds of free-text notes. Under the clinicians’ supervision, such extracted data will drastically expand the underlying data pool and cohort size available for analysis. Data from both unstructured and structured sources will be harmonized to allow for multi-omics queries and model development, starting from descriptive statistics to the development of predictive algorithms. Following the signature of informed consent, all patients with confirmed IBD according with standard of care [3,4] will have a baseline evaluation of family and personal CVD risk factors with a standard questionnaire, including occurrence in family members of MACEs defined as ischemic heart disease (IHD), myocardial infarction (MI), stroke (ST), atrial fibrillation (AF), heart failure (HF), and thromboembolic events (TE). All patients will be evaluated with SCORE2 and the glyco-metabolic inflammatory axes. In case of the occurrence of a high CV risk score (>10 <20%), the carotid intima-media thickness (IMT) will be measured by Doppler ultrasound as a surrogate marker for subclinical atherosclerosis. In case of very high CV risk (more than 10% or more than 7.5% in subjects older than 69 years or younger than 50 years, respectively), the patient will be referred to the cardiologist (if not previously referred) for further investigations. To all patients, lifestyle modification measures will be given in written (i.e., stop smoking, minimize alcohol use, and follow a Mediterranean diet). Patients with IBD will undergo a careful baseline evaluation of disease activity and ongoing therapy, standard laboratory investigations, ileocolonoscopy, and biopsies as standard of care [3,4]. In addition, in all IBD patients, different biological materials (whole blood, plasma/serum, tissues, and stool samples) will be collected and stored in the Policlinic Biobank. All patients will then be followed for at least 2 years, and every year, a re-evaluation of IBD activity, ongoing therapy, lab tests, and monitoring of the occurrence of MACEs will be recorded. The enrollment of the study population will be performed according to the protocol approved by the local ethics committee (Milan 29/1/2025 CET 510-2024), which was recently also registered as NCT07095634.
The control cohort will be that of the CV-PREVITAL trial (a multicenter, prospective, randomized, controlled, open-label interventional trial—NCT05339841) designed to compare the effectiveness of an educational and motivational mobile health (mHealth) intervention versus usual care in reducing CV risk. The Cardiology Unit of IRCC Policlinico San Donato has already enrolled 1109 healthy individuals in this trial. These participants, who were free from known CVDs at baseline, have already completed a 1-year follow-up and are still under cardiological evaluation to monitor their CV health over time. The control group for the IBD cohort, matched for age and sex, will be extrapolated from this population.

2.2. Multi-Omics Evaluation

The second aim is to undertake an extensive multi-omics, metabolic, and histological evaluation.
Genomics: Germline genetic markers using DNA extracted from peripheral blood samples collected from patients with IBD will be evaluated. A panel of approximately 30 single-nucleotide polymorphisms (SNPs) will be analyzed using TaqMan allelic discrimination assays [73]. The list of SNPs to be evaluated will be selected by reviewing the available literature of genes involved in immune and inflammatory processes. Univariate and multivariate stepwise logistic regression models will be used to identify the variables associated with CVD risk and MACEs in patients with IBD.
Transcriptomics: Genome-wide expression profiling of mRNA will be conducted using RNAs extracted from tissue samples collected before and after treatment of selected patients to identify pivotal and critical genes associated with therapy response. The designed whole-transcript array used will provide the most accurate, sensitive, and comprehensive measurement of protein coding and long intergenic non-coding RNA transcripts [74]. Gene expression microarray analysis and functional enrichment analysis of the sample data will be performed to reveal abnormally expressed genes at the tissue level and to elucidate the biological significance of the differential expression pattern.
Metagenomics: A comprehensive sequencing of the gut microbiome will be performed. DNA will be extracted from stool samples at baseline and after 2 years of follow-up, and the variable V3 and V4 regions of the 16S rRNA gene will be sequenced. All procedures will follow extensively validated protocols to perform accurate taxonomic and functional profiling of the microbiome composition using newly developed custom bioinformatics and statistical tools [75].
Proteomics: To perform proteomic profiling of plasma samples at baseline and at the end of follow-up, cutting-edge technology will be applied to overcome the limitation of the outstanding dynamic range of plasma proteins [76]. With these targeted proteomic approaches, we will analyze the potential modulation of proteins involved in the most relevant physio-pathological pathways, including inflammation, immune response, and metabolic dysregulation.
Metabolic evaluation: In addition to the standard laboratory evaluation of the glyco-metabolic axis, patients with IBD will be assessed with Fibroscan at the time of enrollment and at the end of follow-up for the presence of hepatic steatosis.
Histology: The biopsy samples will be oriented on acetate cellulose filters, fixed in formalin, and embedded in paraffin. Sections stained with hematoxylin and eosin (H&E) will be used to evaluate the histological activity with standard validated scores. In addition, Claudin-2 immunohistochemistry will be performed on 4 um sections using the Claudin-2 antibody (Cell Signaling Technology, E1H90 clone, 1:100 dilution). The primary antibody will be detected using a biotin-free polymeric horseradish peroxidase (HRP)–linker antibody conjugate system with heat-induced epitope retrieval using the Bond III automated immune-stainer. Claudin-2 expression will be evaluated with a semi-quantified score along the cytoplasmatic cell membranes of the glandular epithelium and/or of the surface epithelium [77].

2.3. Prospective Evaluation

At the end of the 2-year follow-up, all patients in the IBD cohorts and the control population will be re-evaluated in terms of CVD risk using SCORE 2, the standard laboratory and cardiologic investigations will be repeated, and the occurrence of new MACEs will be reported. All patients with IBD will be regularly evaluated during the follow-up at 3–6 month intervals, and all will receive a clinical, endoscopic, and histologic evaluation at the end of follow-up. The subgroup of patients with the occurrence of MACEs and those with clinical and endoscopic remission will also repeat the transcriptomics and metagenomics evaluation.

2.4. Objectives of the Study

The primary objective of the study will be to evaluate the basal CVD risk in our IBD population compared with the age- and gender-matched cohort of non-IBD patients.
The secondary objectives will be as follows:
Correlation of the basal CVD risk with multi-omics biomarkers, disease activity, lifestyle (i.e., diet, smoking) and therapy;
To evaluate the occurrence of MACEs at the end of follow-up in the patients with IBD and control populations;
To investigate the possible correlation between high CVD risk and MACE occurrence with multi-omics findings;
To identify possible biomarkers of increased CVD risk compared with the control population;
To establish possible correction factors to apply to the standard traditional CVD risk score to incorporate the IBD inflammatory burden.

2.5. Statistical Analysis

In this study, a combination of descriptive and inferential statistical methods will be required to ensure robust analysis. For descriptive statistics of the key characteristics of the study (CD, UC, and matched controls), we used the mean and standard deviation (for continuous variables such as age and BMI), median and interquartile range (for skewed variables), and counts and percentages (for categorical variables, such as sex and smoking status). To check the balance between the groups, the standardized mean differences (SMDs) will assess the balance of covariates between the groups. An SMD < 0.1 typically indicates good balance. t-tests or Mann–Whitney U tests (depending on normality) for continuous variables will compare means or medians across groups. Chi-square tests or Fisher’s exact tests for categorical variables will compare proportions across groups. In view of the evaluation of the outcome of MACEs, we will use Kaplan–Meier survival analysis to estimate the time to the cardiovascular event, stratified by subgroups. In addition, a Cox proportional hazard regression will allow for evaluation of the hazard ratio (HR) of MACEs while adjusting for confounders. The log-rank test will compare the survival curves between the different groups. We will also apply a multivariate logistic regression evaluating the outcome as binary (e.g., MACEs occurring vs. not occurring during the study period). To ensure the robustness of our findings, sensitivity analyses, such as propensity score matching sensitivity may be performed. Because missing data is common in prospective studies, multiple imputation methods will be used to handle missing data in covariates or outcomes.
The power of our sample was estimated by Cohen’s h effect size for proportions based on different hypotheses about the annual incidence rate of main MACEs in Italy (ISTAT—National Institute of Statistics), a range from 1% to 5% per year, and the estimated increase in cardiovascular events in the Crohn’s disease and ulcerative colitis groups, a range from 1.25 (25% increase) to 3.5 (250% increase). Under the assumption of an incidence rate of 1% per year in the control population, a sample size of 300 patients with IBD (CD and UC) and 300 matched controls will have 80% power to detect an HR of 2.5 or greater. Under the assumption of an incidence rate of 5% per year, there will be approximately 80% power to detect a hazard ratio equal to 1.6. The recruitment period was set to 1 year with 4 years of follow-up (2 years retrospective and 2 years prospective) for a total duration of 5 years. The significance level was set at 5%. A dropout of 5% in both groups is assumed. Effect size parameters: Follow-up period of 3 years retrospectively and 2 years prospectively; alpha (α) of 0.05 (standard significance level); power (1—β) of 90%.
For genomics, logistic regression and generalized linear models will be used for association analyses between the identified genetic variants and clinical outcomes, adjusting for confounders, such as age, sex, and disease duration.
Transcriptome data will be analyzed for differential expression patterns, with statistical significance assessed through empirical Bayes methods, followed by pathway enrichment analysis using gene set enrichment analysis (GSEA) and functional network mapping through tools such as STRING (consortium 2024) or Cytoscape (ver. 3.10.3).
Microbiome data will undergo community profiling through alpha and beta diversity metrics, and associations with clinical endpoints will be tested via multivariate analysis methods, such as canonical correspondence analysis (CCA) and redundancy analysis (RDA). Furthermore, metagenomic functional profiles will be reconstructed using PICRUSt2 or HUMAnN3, allowing for functional comparison across patient subgroups. Integration across omics layers will be realized through data integration analysis for biomarker discovery using latent components (DIABLO) and complemented by machine learning-driven feature selection techniques (e.g., LASSO, elastic-net).
Targeted proteomics will be analyzed by ANOVA and Tukey’s for a multiple comparison test. To evaluate the concordance of histological evaluation (active/remission) of different scoring system and Claudin 2 immunostaining, Cohen’s kappa (k) will be evaluated.
In addition, to carry out the systematic evaluation of the large, multidimensional dataset, overcoming the limitation of sample size and length of follow-up, and to detect unexpected pathogenetic hypotheses and biomarkers, an artificial intelligence model will be implemented [78]. This will be performed with the use of the Agora Labs platform. Agora® Node supports the training of machine-learning and AI algorithms in a localized or federated architecture, thus allowing multivariate analysis to correlate clinical and multi-omics findings and other attributes included in the study, as well as deeper evaluation of multidimensional datasets by applying random forest (RF) and support vector machine (SVM) models, among others. Random forest (RF) and support vector machine (SVM) models, specifically, will be tested on feature selection tasks and on the identification of predictors of cardiovascular events from clinical and multi-omics data. RF will highlight influential factors (e.g., genetic markers, microbiome composition, and inflammatory markers), which will then be prioritized in subsequent modeling stages. SVMs, known for their performance with smaller datasets, will then refine classification based on selected features, creating a baseline model for cardiovascular risk. Neural networks, particularly deep neural networks (DNNs) or convolutional neural networks (CNNs), will also be tested in the handling of the study’s complex, high-dimensional dataset, especially in identifying non-linear relationships across genomics, transcriptomics, and proteomics data. After RF and SVM have highlighted the most predictive features, these will be integrated into neural networks to improve accuracy in predicting cardiovascular events.

3. Results

No solid results of the ongoing study are given; only the status of recruitment at the time of writing is reported. In total, 167 patients with IBD have been enrolled with detailed phenotyping and bio-sampling.

3.1. Characteristics of the IBD Study Population

The main characteristics of the IBD study population are depicted in Supplementary Table S1. Enrollment is still in progress, and the end of follow-up is estimated to be by the end of 2027. During the last 3–6 months of the project, multi-omics evaluation and analysis will be performed. At that time, the full data analysis will be shared.

3.2. Characteristics of the Control Population

Table 3 reports the basal characteristics of the control population enrolled in the study CV PREVITAL. The enrollment of this cohort is complete, and all subjects underwent evaluation of their CV risk score and provided blood samples at baseline to evaluate total cholesterol and its fractions, fasting glucose, glycate hemoglobin, triglycerides, homocysteine, and C reactive protein. In addition, bio-samples were collected for further possible multi-omics evaluation and carotid intima-media thickness (IMT) evaluation by Doppler ultrasound. By applying the propensity score from this cohort, an age- and gender-matched control population will be selected for comparison with the IBD cohort.

4. Discussion

The precise mechanisms responsible for the development of cardiovascular events in individuals with inflammatory bowel disease (IBD) remain incompletely defined [7,8,12]. Additional studies are needed to unravel the intricate relationship between systemic inflammation, immune system disturbances, and the onset of cardiovascular disease (CVD) in this population [8]. IBD represents a heterogeneous group of conditions with diverse clinical presentations and trajectories [3,4]. However, most existing research tends to treat IBD as a unified entity, often overlooking possible distinctions in CVD risk between ulcerative colitis (UC) and Crohn’s disease (CD), or across varying levels of disease severity. Further investigation is essential to better characterize these differences and to enable more tailored preventive and therapeutic strategies. Additionally, much of the current evidence is derived from observational or retrospective studies, limiting the ability to draw definitive causal inferences regarding CVD development in this setting (see Appendix A).
At present, there are no specific, validated guidelines for cardiovascular risk assessment in patients with IBD. Many IBD patients are younger adults who fall outside the demographic typically assessed by conventional cardiovascular risk calculators. Nevertheless, the 2019 guidelines from the European Society of Cardiology recognized IBD—alongside conditions such as cancer therapies, rheumatoid arthritis, and systemic lupus erythematosus—as requiring heightened vigilance in screening, counseling, and management for atherosclerotic disease [79]. Similarly, guidance from the European Alliance of Associations for Rheumatology highlights the importance of monitoring inflammatory activity using biomarkers, a principle that may be equally relevant to IBD populations [18].
A multidisciplinary approach is essential for appropriately addressing the interplay between IBD and cardiovascular risk. Beyond early detection and effective treatment, secondary prevention should be prioritized. In this context, recommendations from the American Heart Association support strategies to mitigate cardiovascular risk in patients with IBD, emphasizing the maintenance of disease remission as a key factor in reducing atherosclerotic cardiovascular disease (ASCVD) risk [11]. These guidelines also underscore the need for aggressive control of modifiable risk factors, including routine screening of blood pressure, blood glucose, lipid profiles, and promotion of healthier lifestyle choices. Moreover, there is a strong rationale for adopting personalized prevention strategies in IBD care. Clinicians can tailor interventions by identifying individuals at greater risk of cardiovascular complications, thereby facilitating informed, shared decision-making [11].
Several limitations still exist in fully understanding the IBD–CVD relationship, particularly with regard to therapeutic effects, biological pathways, and the availability of laboratory tools, such as biomarkers, that could help anticipate cardiovascular risk and pathogenesis.
Traditional risk calculators, such as SCORE2, likely underestimate CVD risk in patients with IBD. As seen in rheumatoid arthritis, a correction factor of 1.5 is recommended to account for the persistent inflammatory burden; a similar adjustment may be warranted in patients with IBD [18].
Our study will provide a comprehensive analysis of how disease activity, treatment regimens, and multi-omics profiles influence cardiovascular risk. This approach may help identify common risk pathways and open new prevention and management avenues. At the moment, we have only reported the status of recruitment in Section 3. The control population inclusion, derived from the PREVITAL study, is complete and will be used for gender- and age-matched comparison by applying the propensity score methodology. In contrast, the enrollment of the patients with IBD will be completed by the end of 2025, and after 2 years of follow-up, multi-omics evaluation will be performed and analyzed.
In Italy, there are currently an estimated 250,000 to 300,000 patients with IBD, with projections indicating a possible doubling over the next decade [1]. This trend will inevitably place greater pressure on the national healthcare system. A major strength of our study is its structured approach—applying standard care protocols and monitoring both clinical disease features and lifestyle data over a 5-year period (3 years retrospectively, 2 years prospectively). In addition, we will compare this cohort to a well-matched, rigorously followed control population. Multi-omics analysis will be used to identify potential biomarkers associated with increased cardiovascular risk.
Upon completion of the study, we aim to propose an IBD-specific correction factor to enhance the accuracy of existing CV risk scoring systems. This will be particularly valuable for younger patients—especially those under 40 years—who are not adequately captured by current tools.
Indeed, this subgroup appears to show a more pronounced disparity in MACE incidence compared to the general population. Notably, this will be the first prospective investigation in Italy specifically designed to evaluate CVD risk in patients with IBD and one of only a few worldwide [43]. The findings of this study will offer important insights to clinicians, researchers, and policymakers, contributing to more informed strategies to reduce cardiovascular morbidity and mortality in this vulnerable group.

5. Conclusions

The exact underlying mechanisms of cardiovascular events in IBD remain poorly understood. More research is required to elucidate the complex interplay between inflammation, immune dysregulation, and CVD pathogenesis in IBD, which encompasses a spectrum of diseases with varying clinical phenotypes and courses. More studies are needed to understand these nuances and to guide targeted interventions. Existing studies primarily rely on observational or retrospective data, making it difficult to establish definite causal relationships between specific factors and CVD development. In this study, all consecutive patients with IBD will be evaluated for their basal SCORE 2 and complete disease characteristics and will undergo screening cardiologic investigations to assess potential cardiovascular issues. Following at least 2 years of follow-up, the whole cardiologic and gastroenterological assessment will be repeated, and the occurrence of MACEs will be investigated and correlated with the multi-omics assessment and control cohort. This in-depth evaluation will potentially reveal unexpected and silent underlying high risks for MACEs and direct the subjects, under the control of a cardiologist, to make significant changes in their lifestyle, diet, and therapy. More importantly, the multi-omics evaluation may reveal biomarkers of increased CV risk and suggest corrective factors for the standard CV risk score in this patient population.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jpm15090418/s1, Table S1: Characteristics of the enrolled IBD study population so far (mean ± SD).

Author Contributions

Conceptualization, V.A., M.L.A. and A.L.; methodology, V.A., M.L.A. and S.C. (Sofia Cinque); formal analysis, M.P. and A.L.; investigation, M.L.A., S.C. (Serenella Castelvecchio), G.A.P., P.B., E.O. and G.D.; writing—original draft preparation, V.A.; writing—review and editing, V.A. and S.C. (Serenella Castelvecchio); funding acquisition, V.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was partially funded by an unrestricted grant from AG. Pharma, Via del casale della Marcigliana, 29, 00138 Rome, Italy.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Local Ethical Committee of the Lombardia region based in Milan (name of the study “PROVACI”, protocol code CET 510-2024 of 29 January 2025).

Informed Consent Statement

Informed consent was obtained from all the subjects involved in the study.

Acknowledgments

We sincerely thank Ns Orsola Raffaella Sociale (study coordinator and data manager IBD Outpatient Unit) and Ns Miriam Bottani (endoscopy coordinator) for the support in handling patient consultation and endoscopy. In addition, we thank Carmelina Chiariello, Vincenzo Frusciante, and Mattia Ricotti for their administrative support and communication with the ethics committee and funding sponsor. We also acknowledge the support that will be given by Vincenzo Villanacci for future histological evaluation of the biopsy samples, Cristina Banfi for future proteomics evaluation, Sara Boveri for the forthcoming statistical analysis, and Davide Zanichelli for the forthcoming evaluation using artificial intelligence tools. The authors have reviewed and edited the manuscript and take full responsibility for the content of this publication.

Conflicts of Interest

The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
IBDInflammatory bowel disease
CDCrohn’s disease
UCUlcerative colitis
CVCardiovascular
CVDCardiovascular disease
MACEMajor cardiovascular event
AIArtificial intelligence
IHDIschemic heart disease
MIMyocardial infarction
AFAtrial fibrillation
AAEAcute arterial events

Appendix A. List of More Relevant Studies Investigating the Risk of MACEs in IBD

Authors (Ref.)YearStudy DesignPopulationResults
Bernstein et al. [19]2008Population-based8060 IBD vs. 80,489 controlsRR IHD 1.26 (95% CI 1.11–1.44)
Rungoe et al. [20]2008Population-based28,833 IBD vs. 4,570,820 non-IBDRR IHD 2.13 (95% CI 1.91–2.38) in the first year after diagnosis
Andershon et al. [25]2010Population-based8054 CD and 161,078 controlsIncreased OR for IS in <50 years 2.93 (95% CI 1.44–5.98)
Osterman et al. [34]2011Retrospective15,498 UC and 9829 CD vs. 237,592 controls No increased risk of MI
Yarur et al. [35]2011Longitudinal356 IBD vs. 712 controls OR CAD 4.08 (95% CI 2.49–6.70)
Haapamaki et al. [36]2011Retrospective2831 IBD vs. 5662 controlsOR for CHD 1.88 (95% CI 1.29–2.73); active disease risk factor (p = 0.018)
Sridhar et al. [37]2011Cross-sectionalInpatient databaseOR for dysrhythmias in females aged 18–39 years 2.05 (95% CI 1.71–2.44)
Kristensen et al. [38]2013Population-based20,795 IBD vs. 199,978 controlsRR for MI 1.17 (95% CI 1.05–1.31); rate increased during flares (1.49; 95% CI 1.16–1.93)
Aggarwal et al. [39]2014Retrospective131 IBD with CAD vs. 524 controlsIBD younger, less active smokers, and lower BMI (p = 0.03)
Tsai et al. [40]2014Population-based11,822 IBD vs. 47,288 controlsHR for ACS 1.72 (95% CI 1.53–1.94)
Huang et al. [41]2014Retrospective18,392 IBD vs. 73,568 controlsRR for IS 1.12 (95% CI 1.02–1.230)
Kristensen et al. [27]2014Population-based24,499 IBD vs. 236,275 controlsIRR for stroke during active disease (1.57; 95% CI 1.32–2.21) and chronic activity (1.71; 95% CI 1.32–2.21)
Singh et al. [42] 2014Meta-analysis98,240 IBDOR for CVA 1.18
Kristensen et al. [43]2014Prospective23,681 IBD vs. 5,412,966 controlsIRR for hospitalization for HF 1.37
Xiao et al. [26]2015Meta-analysis124,493 IBD and 4748 strokeRR of stroke in IBD 1.29 (95% CI 1.16–1.43); increased risk in younger patients
Yuan et al. [44]2015Meta-analysis8 cohortsRR of stroke in IBD 1.32 (95% CI 1.20–1.44)
Keller et al. [41]2015Population-based3309 CD vs. 13,236 controlsHR for stroke 1.91 (95% CI 1.65–2.22)
Barnes et al. [45]2016Retrospective567,438 IBD with MI vs. 78,121,000 populationIBD had a lower rate of hospitalization OR 0.51 (95% CI 0.50–0.52)
Feng et al. [46]2017Meta-analysis177,330 IBD vs. 5,573,620 controlsRR IHD 1.24 (95% CI 1.14–1.35)
Sun et al. [47]2018Meta-analysis27 studiesRR stroke 1.25 (95% CI: 1.08, 1.44), RR IHD 1.12 (95% CI: 1.05, 1.21), RR MI 1.17 (95% CI: 1.05, 1.21)
Aniwan et al. [35]2018Longitudinal736 IBD vs. 1472 controls HR MI 2.82 (95% CI 1.98–4.04)
Kirchgesner et al. [48]2018Population-based210,162 IBD SIR for AAEs 1.19 (95% CI 1.16–1.22); SIR for IHD 1.17 (95% CI 1.13–1.21); highest risk < 55 years and disease active
Le Gall et al. [49]2018Case–control30 IBD with AAEs vs. 60 matched IBD withoutDisease activity associated with AAEs OR 10.4 (95% CI 2.1–49.9)
Panwhar et al. [21]2019Population-based290,430 IBD vs. 28,799,790 controlOR for MI 1.25 (95% CI 1.24–1.27)
Aarestrup et al. [50]2019Population-based1293 IBD vs. 107,496 controlsMore frequent CVD 13.2% vs. 10.9% (p = 0.009)
Choi et al. [51]2019Population-based37,477 IBD vs. 112,431 controlsHR for MI 1.8 (95% CI 1.47–2.21); age < 40 years HR 2.96 (95% CI 1.96–4.47)
Kumar et al. [52]2019Longitudinal2449 IBD, 271 IBD with HF vs. 20,444 HFHR for HF with complication in IBD 1.67; IRR of HF 2.54 (95% CI 2.13–3.04) during flare
Ghoneim et al. [53]2020Population-based261,890 IBD vs. 51,914,660 controlRR for CVA OR 8.07 (95% CI 7.9–8.2)
Zui et al. [54] 2020Meta-analysis62,287 IBD vs. 367,337 controlsOR for AF 2.26
Biondi et al. [23]2020Case–control52 IBD vs. 37 controlsCarotid intima–media thickness greater (0.690.12 mm vs. 0.630.12 mm, p = 0.031); atherosclerotic carotid plaque 25% vs. 5.4% p = 0.032
Li et al. [55]2021Meta-analysis710,520 IBD vs. 5,671,535 controlsRR IHD 1.26 (95% CI 1.2–1.32)
Gill et al. [56]2021Observational15,292 IBD vs. 30,584 controlsNo increased risk of MI (HR 1.05 (95% CI 0.89–1.23)
Card et al. [57]2021Retrospective31,175 IBD vs. 154,412 controlsHR for MI 1.83 (95% CI 1.28–2.62) for acute disease and chronic activity (HR 1.69; 95% CI 1.24–2.3)
Chen et al. [24]2021Meta-analysis149,908 patients with strokeRR for stroke in IBD 1.21 (95% CI 1.08–1.34)
Tanislav et al. [58]2021Retrospective11,497 IBD vs. 11,497 controlsHR stroke 1.5; HR for TIA 1.93
Lee et al. [59]2021Cross-sectional157,085 IBDPremature ASCVD OR 1.07; extremely premature OR 1.61
Pemmasani et al. [60]2021Cross-sectional24,220 IBD vs. 6,872,415 controlsMortality for ACS in IBD OR 0.81
Setyawan et al. [61]2022Retrospective34,687 IBD vs. 34,687 non-IBDNo increased risk of MI [IRR 0.62 (95% CI 0.44–0.88]
Nasir et al. [62] 2022Retrospective951 IBD vs. 165 ASCVDRR for ASCVD 1.58 (95% CI 1.17–2.13)
Fang et al. [63]2022Retrospective1425 IBD vs. 1588 controlsHigher risk for IHD (12.1% vs. 5.5%; p < 0.001); higher risk < 35 years (CD OR = 6.3; UC OR = 3.35)
Alayo et al. [22]2023Retrospective5094 IBD vs. 20,376 controlsHR for AAEs 1.19 (95% CI 1.08–1.31)
D’Ascenzo F et al. [64]2023Meta-analysis515,455 controls and 77,140 IBDHR for MI 1.36 [1.12–1.64], death 1.55 [1.27–1.90] other CV disease, such as stroke (HR 1.22 [1.01–1.49]
Sleutjes JAM et al. [65]2023Case–control235 IBD, 829 controlsOR 2.01, (95% CI 1.23–3.27) for CVDs
Pam HN et al. [66]2024NationwideCDC databaseAge-adjusted mortality rates (AAMR) decreased from 0.11 in 1999 to 0.07 in 2020
Sun J et al. [67]2024Nationwide81,749 IBD, 382,190 controlsHigher risk of HF HR 1.19, 95% CI 1.15–1.23
Gardezi SA et al. [68]2025Administrative data41,635 deaths due to CVDs in IBDIncreased age-adjusted mortality rates (AAMRs) from 1999 to 2023 (p = 0.0004)
Ebrahimi R et al. [69]2025Matched cohort987 IBD and 9571 controlsIncreased MACEs HR 1.37 (95% CI, 1.24–1.52).
Luo C et al. [70]2025Meta-analysis 2,802,955 Higher risk of stroke, HR of 1.30 [95% CI 1.21–1.39].
Cohen-Heyman et al. [71]2025Retrospective 14,768 IBD and 120,338 controls IHD associated with IBD in males (HR = 1.82; 95% CI: 1.52–2.17)
RR = relative risk; IRR = incidence rate ratio; OR = odds ratio; SIR = standardized incidence ratio; HR = hazard ratio; IHD = ischemic heart disease; IS = ischemic stroke; MI = myocardial infarction; CAD = coronary artery disease; CHD = coronary ischemic disease; ACS = acute coronary syndrome; CVA = cardiovascular atherosclerosis; AAE = acute artery events; HF = heart failure; AF = atrial fibrillation; MACEs = major cardiovascular events; AAMR = age-adjusted mortality rate; ASCVD = atherosclerotic cardiovascular disease; TIA = transient ischemic attack; CVA = cardiovascular accident (stroke).

References

  1. Ng, S.C.; Shi, H.Y.; Hamidi, N.; Underwood, F.E.; Tang, W.; Benchimol, E.I.; Panaccione, R.; Ghosh, S.; Wu, J.C.Y.; Chan, F.K.L.; et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: A systematic review of population-based study. Lancet 2018, 390, 2769–2778. [Google Scholar] [CrossRef]
  2. Burisch, J.; Zhao, M.; Odes, S.; De Cruz, S.; Vermeire, S.; Bernstein, C.N.; Kaplan, G.G.; Duriicova, D.; Greenberg, D.; Melberg, H.O.; et al. The cost of inflammatory bowel disease in high-income settings: A Lancet Gastroenterology & Hepatology Commission. Lancet Gastroenterol. Hepatol. 2023, 8, 458–492. [Google Scholar] [CrossRef] [PubMed]
  3. Gros, B.; Kaplan, G.G. Ulcerative Colitis in Adults: A Review. JAMA 2023, 330, 951–965. [Google Scholar] [CrossRef] [PubMed]
  4. Cushing, K.; Higgins, P.D.R. Management of Crohn Disease: A Review. JAMA 2021, 325, 69–80. [Google Scholar] [CrossRef] [PubMed]
  5. Harbord, M.; Annese, V.; Vavricka, S.; Allez, M.; Barreiro-de Acosta, M.; Boberg, K.M.; Bruisch, J.; De Vod, M.; De Vries, A.M.; Dick, A.D.; et al. The First European Evidence-based Consensus on Extra-intestinal Manifestations in Inflammatory Bowel Disease. J. Crohns Colitis 2016, 10, 239–254. [Google Scholar] [CrossRef]
  6. Jostins, L.; Ripke, S.; Weersma, R.K.; Duerr, R.H.; McGovern, D.P.; Hui, K.Y.; Lee, J.C.; Schumm, L.P.; Sharma, Y.; Anderson, C.A.; et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012, 491, 119–124. [Google Scholar] [CrossRef]
  7. Kumarapperuma, H.; Wang, R.; Little, P.J.; Kamato, D. Mechanistic insight: Linking cardiovascular complications of inflammatory bowel disease. Trends Cardiovasc. Med. 2024, 34, 203–211. [Google Scholar] [CrossRef]
  8. Lakhanpal, S.; Aggarwal, K.; Kaur, H.; Kanwar, K.; Gupta, V.; Bhavsar, J.; Jain, R. Cardiovascular disease: Extraintestinal manifestation of inflammatory bowel disease. Intest. Res. 2025, 23, 23–36. [Google Scholar] [CrossRef]
  9. Li, F.; Ramirez, Y.; Yano, Y.; Daniel, C.R.; Sharma, S.V.; Brown, E.L.; Li, R.; Moshiree, B.; Loftfield, E.; Lan, Q.; et al. The association between inflammatory bowel disease and all-cause and cause-specific mortality in the UK Biobank. Ann. Epidemiol. 2023, 88, 15–22. [Google Scholar] [CrossRef]
  10. Imbrizi, M.; Magro, S.; Rodrigues Coy, C.S. Pharmacological Therapy in Inflammatory Bowel Diseases: A Narrative Review of the Past 90 Years. Pharmaceuticals 2023, 16, 1272. [Google Scholar] [CrossRef]
  11. Arnett, D.K.; Blumenthal, R.S.; Albert, M.; Buroker, A.B.; Goldberger, Z.D.; Hahn, E.J.; Himmelfarb, C.D.; Khera, A.; Lloyd-Jones, D.; McEvoy, J.W.; et al. ACC/AHA guideline on the primary prevention of cardiovascular disease: Executive summary: A report of the American college of cardiology/American heart association task force on clinical practice guidelines. Circulation 2019, 140, e563-95. [Google Scholar] [CrossRef]
  12. Łykowska-Szuber, L.; Rychter, A.M.; Dudek, M.; Ratajczak, A.E.; Szymczak-Tomczak, A.; Zawada, A.; Eder, P.i.o.t.r.; Lesiak, M.; Dobrowolska, A.; Krela-Kazmierczak, I. What links an increased cardiovascular risk and inflammatory bowel disease? A narrative review. Nutrients 2021, 13, 266. [Google Scholar] [CrossRef] [PubMed]
  13. Alayo, Q.A.; Famutimi, D.; Ayoub, M.; De Las Fuentes, L.; Deepak, P. Performance of ASCVD Risk Prediction Models in Individuals With Inflammatory Bowel Disease: A UK Biobank Study. Inflamm. Bowel Dis. 2025, 31, 285–289. [Google Scholar] [CrossRef] [PubMed]
  14. Jaiswal, V.; Batra, N.; Dagar, M.; Butey, S.; Huang, H.; Chia, J.E.; Naz, S.; Endurance, E.O.; Raj, N.; Patel, S.; et al. Inflammatory bowel disease and associated cardiovascular disease outcomes: A systematic review. Medicine 2023, 102, e32775. [Google Scholar] [CrossRef] [PubMed]
  15. Kamperidis, N.; Kamperidis, V.; Zegkos, T.; Kostourou, J.; Nikolaidou, O.; Arebi, N.; Karvounis, H. Atherosclerosis and inflammatory bowel disease-shared pathogenesis and implication for treatment. Angiology 2021, 72, 303–314. [Google Scholar] [CrossRef]
  16. Kleinbongard, P.; Heusch, G.; Schilz, R. TNF-alpha in atherosclerosis, myocardial ischemia/perfusion and heart failure. Pharmacol. Ther. 2010, 127, 295–314. [Google Scholar] [CrossRef]
  17. Hasan, R.A.; Koh, A.Y.; Zia, A. The gut microbiome and thromboembolism. Thromb. Res. 2020, 189, 77–87. [Google Scholar] [CrossRef]
  18. Graham, I.M.; Angelantonio, E.; Huculeci, R. European Society of Cardiology’ Cardiovascular Risk Collaboration (CRC). New way to “SCORE” risk updates on the ESC scoring system and incorporation into ESC cardiovascular prevention guidelines. Curr. Cardiol. Rep. 2022, 24, 1679–1684. [Google Scholar] [CrossRef]
  19. Bernstein, C.N.; Wajda, A.; Blanchard, J.F. The incidence of arterial thromboembolic diseases in inflammatory bowel disease: A population-based study. Clin. Gastroenterol. Hepatol. 2008, 6, 41–45. [Google Scholar] [CrossRef]
  20. Rungoe, C.; Basit, S.; Ranthe, M.F.; Wohlfahrt, J.; Langholz, E.; Jess, T. Risk of ischaemic heart disease in patients with inflammatory bowel disease: A nationwide Danish cohort study. Gut 2013, 62, 689–694. [Google Scholar] [CrossRef]
  21. Panhwar, M.S.; Mansoor, E.; Al-Kindi, S.G.; Sinh, P.; Katz, J.; Oliveira, G.H.; Cooper, G.S.; Ginwalla, M. Risk of myocardial infarction in inflammatory bowel disease: A population-based national study. Inflamm. Bowel Dis. 2019, 25, 1080–1087. [Google Scholar] [CrossRef] [PubMed]
  22. Alayo, Q.; Loftus, E.V., Jr.; Yarur, A.; Alvarado, D.; Ciorba, M.A.; de las Fuentes, L.; Deepak, P. Inflammatory bowel disease is associated with an increased risk of incident acute arterial events: Analysis of the United Kingdom biobank. Clin. Gastroenterol. Hepatol. 2023, 21, 761. [Google Scholar] [CrossRef] [PubMed]
  23. Biondi, R.B.; Salmazo, P.S.; Sanati Bazan, S.G.; Hueb, J.C.; Rupp de Piava, S.A.; Sassaki, L.Y. Cardiovascular risk in individuals with inflammatory bowel disease. Clin. Exp. Gastroenterol. 2020, 13, 107–113. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, Y.; Wang, X. Increased risk of stroke among patients with inflammatory bowel disease: A PRISMA-compliant meta-analysis. Brain Behav. 2021, 11, e02159. [Google Scholar] [CrossRef]
  25. Andersohn, F.; Waring, M.; Garbe, E. Risk of ischemic stroke in patients with Crohn’s disease: A population-based nested case-control study. Inflamm. Bowel Dis. 2010, 16, 1387–1392. [Google Scholar] [CrossRef]
  26. Xiao, Z.; Pei, Z.; Yuan, M.; Li, X.; Chen, S.; Xu, L. Risk of stroke in patients with inflammatory bowel disease: A systematic review and meta-analysis. J. Stroke Cerebrovasc. Dis. 2015, 24, 2774–2780. [Google Scholar] [CrossRef]
  27. Kristensen, S.L.; Lindhardsen, J.; Ahlehoff, O.; Erichsen, R.; Lamberts, M.; Khalid, U.; Torp-Pedersen, C.; Nielsen, O.H.; Gislason, G.H.; Hansen, P.R. Increased risk of atrial fibrillation and stroke during active stages of inflammatory bowel disease: A nationwide study. Europace 2014, 16, 477–484. [Google Scholar] [CrossRef]
  28. Massironi, S.; Mulinacci, G.; Gallo, C.; Vigano, C.; Fichera, M.; Villatore, A.; Peretto, G.; Danese, S. The oft-overlooked cardiovascular complications of inflammatory bowel disease. Expert. Rev. Clin. Immunol. 2023, 8, 1–17. [Google Scholar] [CrossRef]
  29. Schinzari, F.; Armuzzi, A.; De Pascalis, B.; Mores, N.; Tesauro, M.; Melina, D.; Cardillo, C. Tumor necrosis factor-alpha antagonism improves endothelial dysfunction in patients with Crohn’s disease. Clin. Pharmacol. Ther. 2008, 83, 70–76. [Google Scholar] [CrossRef]
  30. Dheyriat, L.; Ward, D.; Beaugerie, L.; Jess, T.; Kirchgesner, J. Risk of recurrent acute arterial events associated with thiopurines and anti-tumor necrosis factor in inflammatory bowel diseases. Clin. Gastroenterol. Hepatol. 2023, 21, 164–172.e11. [Google Scholar] [CrossRef]
  31. Clarke, B.; Yates, M.; Adas, M.; Bechman, K.; Galloway, J. The safety of JAK-1 inhibitors. Rheumatology 2021, 60 (Suppl. S2), ii24–ii30. [Google Scholar] [CrossRef]
  32. Ytterberg, S.R.; Bhatt, D.L.; Mikuls, T.R.; Kock, G.G.; Fleischmann, R.; Rivas, J.L.; Germino, R.; Menon, S.; Sun, Y.; Wang, C.; et al. Cardiovascular and cancer risk with tofacitinib in rheumatoid arthritis. N. Engl. J. Med. 2022, 386, 316–326. [Google Scholar] [CrossRef]
  33. Olivera, P.A.; Lasa, J.S.; Bonovas, S.; Danese, S.; Peyrin-Biroulet, L. Safety of Janus kinase inhibitors in patients with inflammatory bowel diseases or other immune-mediated diseases: A systematic review and meta-analysis. Gastroenterology 2020, 158, 1554–1573. [Google Scholar] [CrossRef] [PubMed]
  34. Osterman, M.T.; Yang, Y.X.; Brensinger, C.; Forde, K.A.; Lichtenstein, G.R.; Lewis, J.D. No increased risk of myocardial infarction among patients with ulcerative colitis or no increased risk of myocardial infarction among patients with ulcerative colitis or Crohn’s disease. Clin. Gastroenterol. Hepatol. 2011, 9, 875–880. [Google Scholar] [CrossRef] [PubMed]
  35. Yarur, A.J.; Deshpande, A.R.; Pechman, D.M.; Tamariz, L.; Abreu, M.T.; Sussman, D.A. Inflammatory bowel disease is associated with an increased incidence of cardiovascular events. Am. J. Gastroenterol. 2011, 106, 741–747. [Google Scholar] [CrossRef] [PubMed]
  36. Haapamäki, J.; Roine, R.P.; Turunen, U.; Färkkilä, M.A.; Arkkila, P. Increased risk for coronary heart disease, asthma, and connective tissue diseases in inflammatory bowel disease. J. Crohns Colitis 2011, 5, 41–47. [Google Scholar] [CrossRef]
  37. Sridhar, A.R.M.; Parasa, S.; Navaneethan, U.; Crowell, M.D.; Olden, K. Comprehensive study of cardiovascular morbidity in hospitalized inflammatory bowel disease patients. J. Crohns Colitis 2011, 5, 287–294. [Google Scholar] [CrossRef]
  38. Kristensen, S.L.; Ahlehoff, O.; Lindhardsen, J.; Erichsen, R.; Jensen, G.V.; Torp-Pederson, C.; Nielsen, O.H.; Gislason, G.H.; Hansen, P.R. Disease activity in inflammatory bowel disease is associated with increased risk of myocardial infarction, stroke and cardiovascular death–a Danish nationwide cohort study. PLoS ONE 2013, 8, e56944. [Google Scholar] [CrossRef]
  39. Aggarwal, A.; Atreja, A.; Kapadia, S.; Lopez, R.; Achkar, J.P. Conventional risk and cardiovascular outcomes of patients with inflammatory bowel disease with confirmed coronary artery disease. Inflamm. Bowel Dis. 2014, 20, 1593–1601. [Google Scholar] [CrossRef]
  40. Tsai, M.S.; Lin, C.L.; Chen, H.P.; Lee, P.H.; Sung, F.C.; Kao, C.H. Long-term risk of acute coronary syndrome in patients with inflammatory bowel disease: A 13-year cohort study in an Asian population. Inflamm. Bowel Dis. 2014, 20, 502–507. [Google Scholar] [CrossRef]
  41. Huang, W.S.; Tseng, C.H.; Chen, P.C.; Tsai, C.H.; Lin, C.L.; Sung, F.C.; Kao, C.H. Inflammatory bowel diseases increase future ischemic stroke risk: A Taiwanese population-based retrospective cohort study. Eur. J. Intern. Med. 2014, 25, 561–565. [Google Scholar] [CrossRef] [PubMed]
  42. Singh, S.; Singh, H.; Loftus, E.V.J.; Pardi, D.S. Risk of cerebrovascular accidents and ischemic heart disease in patients with inflammatory bowel disease: A systematic review and meta-analysis. Clin. Gastroenterol. Hepatol. 2014, 12, 382–393.e1. [Google Scholar] [CrossRef] [PubMed]
  43. Kristensen, S.L.; Ahlehoff, E.; Lindharsen, J.; Erichsen, R.; Lamberts, M.; Khalid, U.; Mielsen, O.H.; Torp-Pedersen, C.; Gislason, G.H.; Hansen, P.R. Inflammatory bowel disease is associated with an increased risk of hospitalization for heart failure: A Danish Nationwide Cohort study. Circ. Heart Fail. 2014, 7, 717–722. [Google Scholar] [CrossRef]
  44. Yuan, M.; Zhou, H.Y.; Xiao, X.L.; Wang, Z.Q.; Yao-Zhi; Yin, X.P. Inflammatory bowel disease and risk of stroke: A meta-analysis of cohort studies. Int. J. Cardiol. 2016, 202, 106–109. [Google Scholar] [CrossRef]
  45. Barnes, E.L.; Beery, R.M.; Schulman, A.R.; McCaerthy, E.P.; Korzenij, J.R.; Winter, R.W. Hospitalizations for acute myocardial infarction are decreased among patients with inflammatory bowel disease using a nationwide inpatient database. Inflamm. Bowel Dis. 2016, 22, 2229–2237. [Google Scholar] [CrossRef]
  46. Feng, W.; Chen, G.; Cai, D.; Zhao, S.; Cheng, J.; Shen, H. Inflammatory bowel disease and risk of ischemic heart disease: An updated meta-analysis of cohort studies. J. Am. Heart Assoc. 2017, 6, e005892. [Google Scholar] [CrossRef]
  47. Sun, H.H.; Tian, F. Inflammatory bowel disease and cardiovascular disease incidence and mortality: A meta-analysis. Eur. J. Prev. Cardiol. 2018, 25, 1623–1631. [Google Scholar] [CrossRef]
  48. Kirchgesner, J.; Beaugerie, L.; Carrat, F.; Andersen, N.N.; Jess, T.; Schwarzinger, M. BERENICE study group. Increased risk of acute arterial events in young patients and severely active IBD: A nationwide French cohort study. Gut 2018, 67, 1261–1268. [Google Scholar] [CrossRef]
  49. Le Gall, G.; Kirchgesner, J.; Bejaoui, M.; Landman, C.; Nion-Larmurier, I.; Bourrier, A.; Sokol, H.; Seksik, P.; Beaugerie, L. Clinical activity is an independent risk factor of ischemic heart and cerebrovascular arterial disease in patients with inflammatory bowel disease. PLoS ONE 2018, 13, e0201991. [Google Scholar] [CrossRef]
  50. Aarestrup, J.; Jess, T.; Kobylecki, C.J.; Nordestgaard, B.G.; Allin, K.H. Cardiovascular study of more than 100 000 individuals. J. Crohns Colitis 2019, 13, 319–323. [Google Scholar] [CrossRef]
  51. Choi, Y.J.; Lee, D.H.; Shin, D.W.; Han, K.D.; Yoon, H.; Shin, C.M.; Park, Y.S.; Kin, N. Patients with inflammatory bowel disease have an increased risk of myocardial infarction: A nationwide study. Aliment. Pharmacol. Ther. 2019, 50, 769–779. [Google Scholar] [CrossRef] [PubMed]
  52. Kumar, A.; Lukin, D.J. Incident heart failure is a predictor of adverse outcomes in inflammatory bowel disease. Eur. J. Gastroenterol. Hepatol. 2020, 32, 205–215. [Google Scholar] [CrossRef] [PubMed]
  53. Ghoneim, S.; Shah, A.; Dhorepatil, A.; Butt, M.U.; Wagheay, N. The risk of cerebrovascular accidents in inflammatory bowel disease in the United States: A population-based national study. Clin. Exp. Gastroenterol. 2020, 13, 123–129. [Google Scholar] [CrossRef] [PubMed]
  54. Zuin, M.; Zuliani, G.; Rigatelli, G. Atrial fibrillation in patients with inflammatory bowel disease: A systematic review and meta-analysis. Eur. J. Intern. Med. 2020, 76, 120–122. [Google Scholar] [CrossRef]
  55. Li, Z.; Qiao, L.; Yun, X.; Du, F.; Xing, S.; Yang, M. Increased risk of ischemic heart disease and diabetes in inflammatory bowel disease. Z. Gastroenterol. 2021, 59, 117–124. [Google Scholar] [CrossRef]
  56. Gill, G.S.; Fernandez, S.J.; Malhotra, N.; Mete, M.; Garcia-Garcia, H.M. Major acute cardiovascular events in patients with inflammatory bowel disease. Coron. Artery Dis. 2021, 32, 73–77. [Google Scholar] [CrossRef]
  57. Card, T.R.; Zittan, E.; Nguyen, G.C.; Grainge, M.J. Disease activity in inflammatory bowel disease is associated with arterial vascular disease. Inflamm. Bowel Dis. 2021, 27, 629–638. [Google Scholar] [CrossRef]
  58. Tanislav, C.; Trommer, K.; Labenz, C.; Kostev, K. Inflammatory bowel disease as a precondition for stroke or TIA: A matter of Crohn’s disease rather than ulcerative colitis. J. Stroke Cerebrovasc. Dis. 2021, 30, 105787. [Google Scholar] [CrossRef]
  59. Lee, M.T.; Mahtta, D.; Chen, L.; Hussain, A.; Al Rifai, M.; Sinh, P.; Khalid, U.; Nasir, K.; Ballantyne, C.M.; Petersen, L.A.; et al. Premature Atherosclerotic Cardiovascular Disease Risk Among Patients with Inflammatory Bowel Disease. Am. J. Med. 2021, 134, 1047–1051. [Google Scholar] [CrossRef]
  60. Pemmasani, G.; Elgendy, I.; Mamas, M.A.; Leighton, J.A.; Aronow, W.S.; Tremaine, W.J. Epidemiology and Clinical Outcomes of Patients with Inflammatory Bowel Disease Presenting with Acute Coronary Syndrome. Inflamm. Bowel Dis. 2021, 27, 1017–1025. [Google Scholar] [CrossRef]
  61. Setyawan, J.; Mu, F.; Zichlin, M.L.; Billnyer, E.; Downes, N.; Yang, H.; Azimi, N.; Strand, V.; Yarur, A. Risk of thromboembolic events and associated healthcare costs in patients with inflammatory bowel disease. Adv. Ther. 2022, 39, 738–753. [Google Scholar] [CrossRef]
  62. Nasir, K.; Acquah, I.; Dey, A.K.; Agrawal, T.; Hassan, S.Z.; Glassner, K.; Abraham, B.; Quigley, E.M.M.; Blankstein, R.; Virani, S.S.; et al. Inflammatory bowel disease and atherosclerotic cardiovascular disease in U.S. adults—A population-level analysis in the national health interview survey. Am. J. Prev. Cardiol. 2022, 9, 100316. [Google Scholar] [CrossRef]
  63. Fang, L.; Gao, H.; Gao, X.; Wu, W.; Miao, Y.; Zhang, H.; Guleng, B.; Zhang, H.; Wang, Y.; Li, M.; et al. Risks of cardiovascular events in patients with inflammatory bowel disease in China: A retrospective multicenter cohort study. Inflamm. Bowel Dis. 2022, 28, S52–S58. [Google Scholar] [CrossRef] [PubMed]
  64. D’Ascenzo, F.; Bruno, F.; Iannaccone, M.; Testa, G.; De Filippo, O.; Giannino, G.; Caviglia, G.P.; Bernsteing, C.N.; De Ferrari, G.M.; Bugianesi, E.; et al. Patients with inflammatory bowel disease are at increased risk of atherothrombotic disease: A systematic review with meta-analysis. Int. J. Cardiol. 2023, 378, 96–104. [Google Scholar] [CrossRef] [PubMed]
  65. Sleutjes, J.A.M.; van der Woude, C.J.; Verploegh, P.J.P.; Aribas, E.; Kavousi, M.; van Lennep, J.E.R.; de Vries, A.C. Cardiovascular risk profiles in patients with inflammatory bowel disease differ from matched controls from the general population. Eur. J. Prev. Cardiol. 2023, 30, 1615–1622. [Google Scholar] [CrossRef] [PubMed]
  66. Pham, H.N.; Ibrahim, R.; Sainbayar, E.; Aiti, D.; Mouhaffel, R.; Shahid, M.; Ozturk, N.B.; Olson, A.; Ferreira, J.P.; Lee, K. Ischemic heart disease mortality in individuals with inflammatory bowel disease: A nationwide analysis of disparities in the United States. Cardiovasc. Revasc Med. 2024, 65, 46–51. [Google Scholar] [CrossRef]
  67. Sun, J.; Yao, J.; Olén, O.; Halfvarson, J.; Bergman, D.; Ebrahimi, F.; Rosengren, A.; Sundstrom, J.; Ludvigsson, J.F. Risk of heart failure in inflammatory bowel disease: A Swedish population-based study. Eur. Heart J. 2024, 45, 2493–2504. [Google Scholar] [CrossRef]
  68. Gardezi, S.A.; Sachdeva, N.; Rampurawala, I.M.; Ranasinghe, A.; Shehzad, M.U.; Gill, K.; Qureshi, R.; Gupta, A.; Hasan, A.; Farhan, M.; et al. Trends and disparities in inflammatory bowel disease and cardiovascular disease-related mortality in the United States from 1999 to 2023, a CDC WONDER analysis. Int. J. Cardiol. Cardiovasc. Risk Prev. 2025, 26, 200438. [Google Scholar] [CrossRef]
  69. Ebrahimi, R.; Ebrahimi, F.; Niess, J.H.; Mahdi, A.; Chen, S.; Di Vece, D.; Bian, W.; Kutz, A.; Forss, A. Increased Risk of Cardiovascular Events After Coronary Interventions in Inflammatory Bowel Disease: A Nationwide Matched Cohort Study. Aliment. Pharmacol. Ther. 2025, 61, 1904–1912. [Google Scholar] [CrossRef]
  70. Luo, C.; Liu, L.; Zhu, D.; Ge, Z.; Chen, Y.; Chen, F. Risk of stroke in patients with inflammatory bowel disease: A systematic review and meta-analysis. BMC Gastroenterol. 2025, 25, 114. [Google Scholar] [CrossRef]
  71. Cohen-Heyman, N.; Chodick, G. Inflammatory bowel disease is associated with an increased risk of cardiovascular events in a sex and age-dependent manner: A historical cohort study. Int. J. Cardiol. Cardiovasc. Risk Prev. 2025, 24, 200363. [Google Scholar] [CrossRef]
  72. Available online: https://www.agoralabs.eu/ (accessed on 1 June 2025).
  73. Latiano, A.; Palmieri, O.; Latiano, T.; Corritore, G.; Bossa, F.; Martino, G.; Biscaglia, G.; Scimeca, D.; Valvano, M.R.; Pastore, M.; et al. Investigation of multiple susceptibility loci for inflammatory bowel disease in an Italian cohort of patients. PLoS ONE 2011, 6, e22688. [Google Scholar] [CrossRef]
  74. Palmieri, O.; Creanza, T.M.; Bossa, F.; Latiano, T.; Corritore, G.; Palumbo, O.; Martino, G.; Buscaglia, G.; Scimeca, D.; Carella, M.; et al. Functional Implications of MicroRNAs in Crohn’s Disease Revealed by Integrating MicroRNA and Messenger RNA Expression Profiling. Int. J. Mol. Sci. 2017, 18, 1580. [Google Scholar] [CrossRef]
  75. Palmieri, O.; Bossa, F.; Castellana, S.; Latiano, T.; Carparelli, S.; Martino, G.; Mangoni, M.; Corritore, G.; Nardella, M.; Guerra, M.; et al. Deciphering Microbial Composition in Patients with Inflammatory Bowel Disease: Implications for Therapeutic Response to Biologic Agents. Microorganisms 2024, 12, 1260. [Google Scholar] [CrossRef] [PubMed]
  76. Sartore, G.; Piarulli, F.; Ragazzi, E.; Mallia, A.; Ghilardi, S.; Carollo, M.; Lapolla, A.; Banfi, C. Circulating Factors as Potential Biomarkers of Cardiovascular Damage Progression Associated with Type 2 Diabetes. Proteomes 2024, 12, 29. [Google Scholar] [CrossRef] [PubMed]
  77. Villanacci, V.; Del Sordo, R.; Lanzarotto, F.; Ricci, C.; Sidoni, A.; Manenti, S.; Mino, S.; Bugatti, M.; Bassotti, G. Claudin-2, A marker for a better evaluation of histological mucosal healing in inflammatory bowel diseases. Dig. Liver Dis. 2025, 57, 827–832. [Google Scholar] [CrossRef] [PubMed]
  78. Gao, S.; Fang, A.; Huang, Y.; Giunchiglia, V.; Noori, A.; Schwarz, J.R.; Ektefaie, Y.; Kondic, J.; Zitnik, M. Empowering biomedical discovery with AI agents. Cell 2024, 187, 6125–6151. [Google Scholar] [CrossRef]
  79. Knuuti, J.; Wijns, W.; Saraste, A.; Capodanno, D.; Barbato, E.; Funck-Brentano, C.; Prescott, E.; Storey, R.F.; Deaton, C.; Cuisset, T.; et al. 2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes. Eur. Heart J. 2020, 41, 407–477. [Google Scholar] [CrossRef]
Table 3. Basal characteristics of the control population (mean ± SD).
Table 3. Basal characteristics of the control population (mean ± SD).
Overall (N = 1109)Male (N = 492)Female (N = 617)
Age (yrs)55.9 ± 6.556.9 ± 6.555.1 ± 6.2
BMI25.7 ± 4.526.8 ± 3.9 24.9 ± 4.8
Waist (cm)90.3 ± 13.498.1 ± 11.484.5 ± 12.9
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Annese, V.; Annunziata, M.L.; Petroni, G.A.; Orlando, E.; Cinque, S.; Parisi, M.; Biamonte, P.; Dell’Anna, G.; Latiano, A.; Castelvecchio, S. Cardiovascular Complications Are Increased in Inflammatory Bowel Disease: A Path Toward Achievement of a Personalized Risk Estimation. J. Pers. Med. 2025, 15, 418. https://doi.org/10.3390/jpm15090418

AMA Style

Annese V, Annunziata ML, Petroni GA, Orlando E, Cinque S, Parisi M, Biamonte P, Dell’Anna G, Latiano A, Castelvecchio S. Cardiovascular Complications Are Increased in Inflammatory Bowel Disease: A Path Toward Achievement of a Personalized Risk Estimation. Journal of Personalized Medicine. 2025; 15(9):418. https://doi.org/10.3390/jpm15090418

Chicago/Turabian Style

Annese, Vito, Maria Laura Annunziata, Guglielmo Albertini Petroni, Emanuele Orlando, Sofia Cinque, Marzio Parisi, Paolo Biamonte, Giuseppe Dell’Anna, Anna Latiano, and Serenella Castelvecchio. 2025. "Cardiovascular Complications Are Increased in Inflammatory Bowel Disease: A Path Toward Achievement of a Personalized Risk Estimation" Journal of Personalized Medicine 15, no. 9: 418. https://doi.org/10.3390/jpm15090418

APA Style

Annese, V., Annunziata, M. L., Petroni, G. A., Orlando, E., Cinque, S., Parisi, M., Biamonte, P., Dell’Anna, G., Latiano, A., & Castelvecchio, S. (2025). Cardiovascular Complications Are Increased in Inflammatory Bowel Disease: A Path Toward Achievement of a Personalized Risk Estimation. Journal of Personalized Medicine, 15(9), 418. https://doi.org/10.3390/jpm15090418

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

Article Metrics

Back to TopTop