Dental and Medical Problems

Dent Med Probl
Impact Factor (IF 2024) – 3.9
Journal Citation Indicator (JCI 2024) - 1.36
Scopus CiteScore (2024) – 5.0
Index Copernicus Value (ICV 2023) – 181.00
MNiSW – 70 pts
ISSN 1644-387X (print)
ISSN 2300-9020 (online)
Periodicity – bimonthly


 

Download original text (EN)

Dental and Medical Problems

Ahead of print

doi: 10.17219/dmp/210102

Publication type: review

Language: English

License: Creative Commons Attribution 3.0 Unported (CC BY 3.0)

Download citation:

  • BIBTEX (JabRef, Mendeley)
  • RIS (Papers, Reference Manager, RefWorks, Zotero)

Cite as:


Wilk MM, Wilk J, Florek K, Zimoch WJ. Smoldering inflammation: The silent flame driving heart failure [published online as ahead of print on October 31, 2025]. Dent Med Probl. doi:10.17219/dmp/210102

Smoldering inflammation: The silent flame driving heart failure

Michał Maksymilian Wilk1,2,A,B,C,D,E,F, Jakub Wilk1,2,A,B,C,D,E,F, Kamila Florek1,3,A,B,C,D,E,F, Wojciech Jan Zimoch1,4,C,E,F

1 Institute of Heart Diseases, Wroclaw Medical University, Poland

2 Student Scientific Organization, Institute of Heart Diseases, Wroclaw Medical University, Poland

3 Students’ Scientific Group of Invasive Cardiology, Institute of Heart Diseases, Wroclaw Medical University, Poland

4 Institute of Heart Diseases, Wroclaw University Hospital, Poland

Graphical abstract


Graphical abstracts

Highlights


  • Smoldering inflammation (SI) is a key factor in the pathophysiology of heart failure (HF).
  • Understanding SI may enhance risk stratification and guide personalized management strategies in HF.
  • Both established and emerging anti-inflammatory therapies, used alone or in combination, may effectively target SI to improve HF outcomes.

Abstract

In recent years, significant advancements in the understanding of the processes underlying heart failure (HF) have been made, particularly regarding the role of chronic low-intensity inflammation or smoldering inflammation (SI). This review consolidates findings from the available literature and illustrates the relationships between inflammation, neurohormonal activation, metabolic derangements, and comorbidities in HF, with a focus on heart failure with preserved ejection fraction (HFpEF).

A comprehensive literature search was conducted using PubMed®, Wiley Online Library, Scopus, and Web of Science (limited to 2025). The search terms included “heart failure”, “HFpEF”, “inflammation”, “smoldering inflammation”, “biomarkers”, “cytokines”, “fibrosis”, and “comorbidities”. Peer-reviewed articles, reviews, as well as clinical and observational studies describing the mechanistic, prognostic and therapeutic aspects of SI in HF were included. Studies limited to acute coronary syndrome (ACS) were excluded.

Structural changes leading to hemodynamic perturbations in HFpEF are correlated with processes mediated by SI. Several biomarkers measure inflammation and provide diagnostic and prognostic value, including C-reactive protein (CRP), interleukin-6 (IL-6), soluble suppression of tumorigenicity 2 (sST2), galectin-3 (Gal-3), and iron homeostasis. Clinical trials demonstrate the efficacy of sodium–glucose cotransporter-2 (SGLT-2) inhibitors, glucagon-like peptide 1 (GLP-1) receptor agonists, and other targeted interventions in the modulation of SI.

Smoldering inflammation is a key mechanism in the pathogenesis of HFpEF and the progression of comorbidities. Understanding SI may improve risk stratification and management strategies. Both established and emerging anti-inflammatory therapies, when administered alone or in combination, may target SI in order to enhance HF management.

Keywords: inflammation, heart failure, cardiac remodeling, neurohormonal interactions

Introduction

Heart failure (HF) is a cardiac condition that has emerged as a global health concern, affecting approx. 64 million individuals worldwide.1 Recent advancements in pharmacotherapy for one of the HF phenotypes, HF with reduced ejection fraction (HFrEF), have shifted the structure of patients presenting with a specific HF phenotype toward HFpEF. Nowadays, HFpEF accounts for more than half of the patients presenting with HF.2 Due to its heterogeneous etiology and complexity of development, managing this condition is still a significant challenge. One of the potential factors contributing to the pathophysiology of HF, and especially to HFpEF, is smoldering inflammation (SI), which can be described as chronic low-grade inflammation, promoting maladaptive responses and triggering maladaptive mechanisms.3

Inflammation is associated with neurohormonal activation, fibroblast activation, and subsequent fibrosis, oxidative stress, vascular endothelial dysfunction, and ischemia.3 These processes are the fundamental core of SI. The chronic nature of this condition complicates its observation and analysis. However, in recent decades, the complex relationship between metabolic pathways and the onset and development of HF has been revealed.4 Consequently, we have identified specific inflammation-related biomarkers that might assist in predicting the future onset of HF that remains clinically silent.5 Addition­ally, inflammatory indicators may prove useful in risk stratification and outcome assessment.2, 3, 5, 6, 7, 8, 9, 10, 11, 12

Comorbidities that are prevalent in patients with HF should be perceived not only as additional conditions, but rather as a common result of a shared pathophysiological, inflammation-dependent process or even a direct cause of HF development.13, 14, 15, 16, 17, 18, 19, 20, 21, 22 It is important to distinguish the connection between HF and comorbidities because effective treatment of concomitant diseases is a promising way of preventing HF onset or progression. In the sub­set of HFpEF patients, SI appears to play a pivotal role in the pathogenesis process, simultaneously promoting the development of multimorbidity and exacerbating its severity.5, 23, 24 Therefore, the role of SI in the etiology of the disease requires further evaluation.

Lastly, pioneering discoveries related to the role of inflammation in the exacerbation of HF provided a ratio­nale for the evaluation of novel drugs for SI alleviation, as well as for the identification of effective responses to the growing number of patients presenting with HFpEF who require advanced treatment.16, 25, 26, 27 Numerous therapeutic avenues related to SI research offer optimism for the identification of efficacious, life-extending therapeutic modalities, but the results of these studies require careful analysis.

Even though numerous studies and reviews have explored the role of inflammation in cardiovascular disease (CVD) and HF, the majority of these studies have focused on either HFpEF or responses to acute inflamma­tion. Smoldering inflammation has not been explored as a mechanism that connects comorbidities, metabolic dysfunction and structural cardiac remodeling in the context of HFpEF. Importantly, none of the reviews has systematically linked biomarkers, comorbidities or therapies into a single construct. Current studies describe biomarkers, comorbidities or therapies separately, and do not provide a comprehensive framework of how chronic, low-grade inflammation contributes to the presentation, progression and therapeutic response of HFpEF. By using available evidence regarding molecular pathways, biomarkers, comorbidities, and novel anti-inflammatory interventions, our objective is to identify and address this knowledge gap by offering a comprehensive view of SI in the context of HFpEF. We hope that our unique approach will compel others to emulate this model in clinical research and alternative therapeutic approaches.

Therefore, the aim of this review was to consolidate the current evidence regarding the role of SI in HF, with a particular focus on HFpEF. Particular attention has been placed on biomarkers, outcomes, comorbidities, and novel therapies to elucidate the associations between SI and the development, exacerbation and treatment of HFpEF.

Material and methods

In this review, a structured literature search was performed for studies assessing the role of SI in the field of HF, with a focus on HFpEF. The core databases used for the search were PubMed® and Scopus, with supplemen­tary searches conducted in Web of Science and Wiley Online Library, if appropriate. The following Medical Subject Headings (MeSH) terms were used: “heart failure”, “HFpEF”, “inflammation”, “smoldering inflammation”, “biomarkers”, “cytokines”, “fibrosis”, and “comorbidities”. These keywords were utilized to assess studies evaluating a range of pathophysiological mechanisms, as well as diagnostic and prognostic aspects of molecular pathways for SI and therapeutic measures implemented in HF.

The search encompassed articles published until 2025, with no other limitations regarding study design. To assess the multifactorial source of inflammation in HF, the study analyzed clinical trials, systematic reviews and observational cohort studies.

The inclusion criteria for the study encompassed peer-reviewed original articles, reviews, meta-analyses, and clinical trials that discussed inflammation, inflammatory biomarkers and inflammation-targeted therapies within the context of HF, with a particular focus on HFpEF. The exclusion criteria were studies that focused solely on acute coronary syndrome (ACS) or non-inflammatory origin of HF. The selection of articles was based on relevance, scientific quality and novelty.

Pathophysiology

The pathophysiology of HF is complex, with inflammation playing a pivotal role in this process. Nevertheless, the contribution of inflammation varies depending on the HF phenotype, with myocardial injury being the leading cause of HFrEF and inflammation in HFpEF.3 Notably, these phenotypes are not distinct entities, with heart failure with mildly reduced ejection fraction (HFmrEF) serving as a buffer between them that can progress to either HFpEF or HFrEF.4, 28 The phenotypes of HF share some common pathways, some of which are more pronounced in HFpEF compared to HFrEF and vice versa.3

Heart failure with preserved ejection fraction is mainly preceded by metabolic diseases, including obesity and type 2 diabetes mellitus (T2DM). These conditions induce chronic low-grade inflammation, also referred to as metainflammation.29 Metainflammation leads to coronary microvascular endothelial inflammation, which contributes to cardiac remodeling through fibrosis and hypertrophy.30, 31 The aforementioned mechanisms result in left ventricular (LV) stiffness and increased filling pressure at rest and during exercise.32, 33, 34, 35

A prominent contributor to this comorbidity-driven inflammation is the nucleotide oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome.36, 37 This cascade is triggered by interleukin (IL)-1 and tumor necrosis factor alpha (TNF-α), or by reactive oxygen species (ROS) with mitochondrial damage, both leading to the increased activation of NLRP3. Consequently, NLRP3, via adaptor protein, binds to the caspase-1, which cleaves pro-IL-1β and pro-IL-18 to their active forms.38 An effect of IL-1β is seen as a further enhancement of nuclear factor kappa B (NF-κB) production and an increase in IL-6 level, directly stimulating the liver to synthesize highly sensitive C-reactive protein (hs-CRP), which is a widely available biomarker of inflammation.39 On the other hand, higher oxidative stress observed in HFpEF contributes to the enhancement in ROS production with concomitant endothelial dysfunction, causing a drop in nitric oxide (NO) synthesis, which is a direct inhibitor of NLRP3. Nevertheless, NO is not only involved in the cross-talk between these pathways; its impaired production results in the suppression of the NO–soluble guanylate cyclase (cGMP)–protein kinase G (PKG) cascade, leading to vasoconstriction and fibrosis.40 Moreover, NO–cGMP–PKG knockdown results in the hypophosphorylation of titin, which is a key sarcomeric protein involved in diastolic cardiomyocyte relaxation, consequently inducing cardiomyocyte stiffness.41 In addition, PKG downregulation results in the hypophosphorylation of the protein RhoA, which reduces its protective role and leads to the hypertrophic and fibrotic activation of vascular smooth muscle cells (VSMCs).41

Another factor that exerts an antifibrotic and anti­hypertrophic effect on the heart is IL-33, which is released in response to cardiac mechanical stress and injury.42, 43 Importantly, the cardioprotective signaling pathway activated by IL-33 involves suppression of tumorigenicity 2 (ST2) with its 2 variants – transmembrane (suppression of tumorgenicity 2 ligand (ST2L)) located at myocytes, fibroblasts and inflammatory cells, which is activated by IL-33, inducing antihypertrophic and antifibrotic mechanisms that promote adaptive remodeling, and a soluble form (soluble suppression of tumorigenicity 2 (sST2)), which plays the role of the decoy receptor, sequestering IL-33.43 On the other hand, in conditions of cardiac stress or damage, an increased level of sST2 is released, preventing the formation of the ST2–IL-33 complex and suppressing its cardioprotective effect.43 Of note, IL-33 activates NF-κB, which has a dual role in acute hypoxia and cardiac injury and exerts a protective role by inhibiting NF-κB activation induced by angiotensin II, a hypertrophic stimulus.44 However, its chronic activation promotes HF by enhancing the effects of IL-1, TNF-α and IL-6.44

Galectin-3 (Gal-3) is another molecule with a dual role. It is secreted by activated macrophages and plays apoptotic and antinecrotic roles. However, its long-term over­expression observed in HF enhances pro-inflammatory and pro-fibrotic processes.45 Galectin-3, via fibroblast activa­tion, stimulates extracellular matrix (ECM) components like collagen I and collagen III and the synthesis of cyto­skeletal proteins, simultaneously inhibiting matrix metallo­proteinase (MMP)-induced degradation.45 It is worth mentioning that the disturbance between MMPs and their endogenous inhibitor may lead to excessive accumulation of collagen in the myocardium, which, in turn, further promotes fibrosis and secondary stiffness, ultimately impairing the diastolic function of the ventricles, especially in HFpEF.46 Other factors contributing to collagen degradation are angiotensin II, aldosterone, TNF-α, and transforming growth factor beta (TGF-β).47 Moreover, Gal-3 induces aortic valve calcifications through NF-κB and, by the TGF-β1/Smad pathway, promotes atrial fibrillation (AF), subsequently leading to impaired diastolic filling.48

The cumulative effect of these molecular alterations — inflammation, fibrosis and endothelial dysfunction — leads to structural and functional impairment, character­ized by myocardial stiffening and impaired ventricular relaxation, accompanied by increased filling pressures and atrial remodeling. As a result, the clinical manifestation of HF presents as dyspnea, exercise intolerance and systemic congestion. A complex metabolic interplay is demonstrated in Figure 1, and key findings regarding biomarkers and pathways related to SI are summarized in Table 1.

Clinical role of inflammatory biomarkers in HFpEF

Heart failure is a progressive disease that develops through stages, as defined by the American College of Cardiology (ACC) and the American Heart Association (AHA).49

The presence of risk factors contributing to HF development, e.g., T2DM, corresponds to stage A, followed by asymptomatic structural heart diseases (stage B), the presence of clinical symptoms of HF (stage C), and end-stage or refractory HF described as stage D.50 Even though the risk factors of HFpEF are well-defined, the determinants contributing to the progression of HFpEF from stage A to B, as well as more advanced stages, are under investigation.51 Several biomarkers involved in the inflammatory processes underlying HFpEF development are considered to be predictors of disease progression and patients’ outcomes, as well as therapeutic targets.52

A recent meta-analysis emphasized that high levels of CRP were associated with a 9% increase in the risk of HFpEF development.6 In addition, a cohort study revealed the significant role of other NLRP3 inflamma­some molecules, particularly IL-6 in HFpEF and TNF-α in the entire HF group.53 Importantly, a prospective study within a PREVEND cohort demonstrated strong evidence of IL-6’s predictive role in new-onset HFpEF, underscoring its significance in HFpEF, but not in HFrEF incidence prognosis.54

However, among patients diagnosed with HFpEF, significantly higher CRP levels were noted in those with a greater comorbidity burden, whereas CRP was within a normal range in 40% of individuals. These findings underscore the need to broaden the spectrum of biomarkers involved in the assessment of patients with HFpEF.55 Even though current HF phenotyping relies primarily on the echocardiographic examination, a meta-analysis involving proteomic studies that compared HFpEF and HFrEF biomarker profiles suggested that they can be distinguished based on higher levels of IL-6 and lower levels of syndecan-1 (SDC-1) and NO in HFpEF patients, highlighting the difference in prevailing pathomechanisms underlying HF presentation.56

Moreover, a meta-analysis on the role of CRP in HF progression confirmed its predictive value regarding cardiovascular and all-cause mortality. Of note, the significance of CRP regarding long-term adverse cardiovascular outcomes was inconclusive, depending on its presentation in the included studies. A significant association was found for categorical variables, while no such effect was observed for continuous variables.6 Nevertheless, the LUCRIC study has analyzed the difference in the inflammatory biomarkers comparing patients with HFpEF and HFrEF, and exhibited the role of IL-6 in the mortality prognosis in a mean 9.9-year follow-up period among HFpEF patients.5 Similar results were observed for hs-CRP, whereas they failed to be significant after full adjustment.5 Of note, neither IL-6 nor hs-CRP predicted cardiovascular mortality in HFrEF patients, hence highlighting the role of subclinical inflammation in the HFpEF population.5 However, recent results from the TOPCAT trial revealed that hs-CRP ≥ 2 mg/L is an appropriate tool to identify HFpEF patients at a higher risk of HF events and cardiovascular mortality.57

Other relevant biomarkers, whose role in the progression to clinically overt HF among patients with risk factors was evaluated in the retrospective analysis of the STOP-HF longitudinal study, are sST2 and Gal-3.58 This study confirmed the role of Gal-3 in the predictive profile, along with B-type natriuretic peptide (BNP) and high-sensitivity troponin I, whereas it rejected sST2 and IL-6. The results in terms of IL-6 were contrary to those of larger, prospective studies.58 The role of Gal-3 was further confirmed by a meta-analysis, which considered it an appropriate biomarker to indicate new-onset HFpEF, as well as a valuable predictor of adverse outcomes and severity of left ventricular diastolic dysfunction (LVDD) in HFpEF.59 Furthermore, Gal-3 is involved in the activation of MMP.46 Among patients with hypertension, MMP-9 and tissue inhibitor of metalloproteinase 1 (TIMP-1) indicated significantly greater degrees of asymptomatic LVDD.46 Galectin-3 also induces the upregulation of TGF-β, whereas its role as a biomarker is modest.46 In contrast to another TGF-β family molecule, substantially increased inflammation and oxidative stress, known as growth differentiation factor 15 (GDF-15), was shown to be an independent risk factor of adverse outcomes across the entire HF spectrum, with its incremental prognostic role in the HFpEF subgroup.60 In another study, the GDF-15 level measured within 48 h of hospital admission in patients with HFpEF was a better predictor of 1-year rehospitalization than N-terminal pro-B-type natriuretic peptide (NT-proBNP).61

Nevertheless, while focusing on sST2 among individuals hospitalized due to HF, its baseline levels are associated with further rehospitalizations and all-cause mortality, independently of ejection fraction. Furthermore, sST2 combined with NT-proBNP, especially in HFpEF patients, is suggested to improve the predictive value of the test.62 Another clinical trial analyzing the role of sST2 noted its role as a significant biomarker in both HF phenotypes, whereas in HFpEF, sST2 revealed a stronger association with patients’ outcomes.63 Other molecules involved in the pathogenesis of HFpEF are mechanistically crucial and play key roles as therapeutic targets, whereas their clinical predictive role is limited and currently of lower importance than the aforementioned parameters.

It is also important to note that SI and the activation of various inflammatory biomarkers or processes can contribute to congestion development, and vice versa.64, 65, 66, 67 Moreover, several mechanisms of both congestion and inflammation contribute to fibrosis and impaired sodium handling, which are part of the disease’s pathophysiology. However, clinical trial data indicates that these pathways are independent. For example, interventions targeting the natriuretic peptide axis do not reduce inflammatory processes.68 Importantly, the role of the lymphatic system has recently been demonstrated in both HF and decongestion, further linking the immune system to congestion development.69, 70, 71, 72, 73, 74

Iron deficiency in patients with HF is also an important biomarker for predicting outcomes.75, 76, 77 Patients with HF are more likely to develop an iron shortage, which can lead to poorer therapeutic results than in patients without such conditions. Jankowska et al. defined iron deficiency as a high (≥1.59 mg/L) level of serum soluble transferrin receptor (sTfR), indicating unmet cellular iron needs, and a low (<14.5 ng/mL) hepcidin level, reflecting low iron storage.75 Patients with acute HF (AHF) who met both criteria had the highest mortality rate of 41%. In comparison, a mortality rate of 15% was noted in individuals with an isolated high sTfR level, and a rate of 7% was observed in those with an isolated low hepcidin level.75 Another trial has connected lactate levels with sTfR. Biegus et al. assessed the relationship between elevated markers (>2 mmol/L and >1.59 mg/L, respectively) in patients with AHF and the occurrence of adverse outcomes.76 It showed that with both markers present, the prognoses were poorer than in patients with one or none of the criteria met.76 The solution to this issue may be iron supplementation. Ahmed et al. conducted a meta-analysis on intravenous (IV) iron supplementation in patients with HF.77 The results indicated that such therapy can reduce CV mortal­ity, 1-year all-cause mortality, first HF hospitalization, and even improve left ventricle ejection fraction (LVEF).77 These studies underscore the importance of assessing iron status in patients with HF, especially those with AHF, as it may be considered a valuable predictive factor of future outcomes. Another reason for its importance is the possibility of reversing it by IV iron supplementation. Although the relationship between iron metabolism and inflammation is well-documented, with evidence pointing to the modulatory role of hepcidin in regulating iron availabil­ity, current state of knowledge does not allow us to understand the impact of inflammation on the effectiveness of IV iron therapy. Therefore, additional long-term studies are needed in this area.78

Comorbidities associated with HFpEF and inflammation

During the past decades, medical attention has concentrated on the management of HFrEF, which resulted in clinical success and a decline in mortality rates within this particular phenotype. The described progress can be attributed to the implementation of a well-established quadruple therapy, consisting of sodium–glucose cotransporter-2 (SGLT-2) inhibitors, mineralocorticoid receptor antagonists (MRAs), beta-blockers, and angiotensin converting enzyme (ACE) inhibitors.2, 79, 80, 81 Despite the improvement in HFrEF survivability, another issue has emerged. Currently, the predominant form of HF is HFpEF. This HF subtype is a surging threat, accounting for more than half of all HF hospitalizations.2, 82 It is a complex condition that creates various diagnostic and therapeutic difficulties due to its multifactorial and hetero­geneous etiology, which is still not well understood.2, 14 Thereupon, specific attention should be given to established risk factors that contribute to the development of HFpEF, with a strong focus on the widespread comorbidities associated with this condition.2, 5, 13, 14, 15, 16, 20, 21, 22, 24, 79, 82, 83, 84, 85 Distinguishing each coexisting disorder and a thorough evaluation are essential to assess whether the condition is a complication of HF or an independent disease that can be addressed through an individualized therapeutic strategy.13 Accordingly, it is imperative to investigate the correlation between the onset of HFpEF and the preva­lence of comorbidities. We hereby propose a focused examination of low-grade chronic inflammation, which not only contributes to HFpEF evolution but also worsens its course by the promotion of comorbidities (Figure 2).5, 13, 24, 82, 86

Atrial fibrillation

Atrial fibrillation is a common form of arrhythmia, widely diagnosed in patients with HFpEF due to the presence of shared pathophysiological mechanisms. It has been associated with left atrial (LA) wall remodeling and initiated by increased LA pressure, as a consequence of impaired diastolic function of the left ventricle.13, 86, 87 The etiological co-occurrence between these 2 conditions is associated with the activation of maladaptive processes, such as chronic inflammation, oxidative stress and endothelial dysfunction. These risk factors are induced by highly prevalent diseases, mostly hypertension, obesity, T2DM, and chronic kidney disease (CKD). The conditions are manageable in many cases, therefore, they represent an important aspect of curtailing HFpEF progression as well as implementing suitable treatment.87, 88 On a molecular
level, inflammation plays a pivotal role in disease evolution since pro-inflammatory cytokines are responsible for activating fibroblasts that promote remodeling via collagen deposition within an ECM inside the LA myocardium. In patients suffering from HFpEF, AF might occur after HF diagnosis, concurrently, or even prior to HF identification, especially if HFpEF is subclinical or has been stable.86 Regardless of the coincidence of these 2 conditions, AF has been identified as a significant risk fac­tor for mortality when compared to HFpEF patients with sinus rhythm.86 Therefore, it is important to perceive AF not only as a coexisting disease, but also as a substantial stratification factor that may influence therapeutic interventions, including the mitigation of risk factors, proper pharmacotherapy, and the performance of electrophysio­logical procedures.2, 86

Obesity

Obesity is a condition characterized by excessive fat accumulation.15 The global prevalence of this disease has increased drastically over the past few decades, reaching pandemic levels.15, 89 In 2024, it was estimated that 13% of the adult world population suffered from obesity, while 39% were overweight.15 Accordingly, HF and obesity are two conditions that increasingly coexist each year, and impact each other via various pathophysiological interactions, including structural, functional, metabolic, and neurohormonal abnormalities.13, 14, 15, 16, 79, 82, 90, 91 Evidence suggests a strong interconnection between all phenotypes of HF and obesity; however, the association is remarkable and most significant in terms of HFpEF, where increased body mass is believed to be not only a comorbidity but, most importantly, a direct cause of HFpEF pathogenesis.2, 16, 90

It has been established that adipose tissue is an abundant source of biologically active molecules. Fat accumulation is associated with the dysregulation of various compounds, including leptin, resistin, adiponectin, and apelin.92 These molecules have multidirectional effects, such as appetite regulation, vasodilatation, insulin sensitization, the suppression of inflammation, and the production of ROS.92 The impaired function of these adipokines can contribute to the development of metabolic disorders, such as poor glucose tolerance, increased synthesis of fatty acids with pro-inflammatory potential, elevated production of reactive oxygen and nitrogen species, heightened sympathetic nervous system (SNS) activity, and, above all, the promotion of inflammation mediated by macrophages through the secretion of cytokines.15, 92, 93 This particular situation is a starting point at which meta­bolic and neurohormonal abnormalities contribute to the development of functional and structural alterations that manifest in the course of HFpEF. It is important to acknowledge the chronic nature of this process, which may remain in the subclinical phase for many years. However, the presence of additional contributing factors, such as AF, can precipitate the development of full-blown HFpEF and result in a condition that may be difficult or even impossible to reverse.13, 14, 15, 16, 79, 90, 93 The impact of inflamma­tion on cardiac remodeling in the HFpEF–obesity subset is extremely complex, but some common structural observations include perivascular and interstitial fibrosis, VSMC proliferation, LV and RV remodeling, LA dilation, and epicardial fat accumulation.15 Lastly, these findings contribute to functional impairment that is characterized by volume overload, coronary microvascular dysfunction, AF, a decline in systolic and diastolic reserve, and an increase in LV filling pressure.15 On this account, obesity plays a crucial role in the pathogenesis of HFpEF, as well as in its management and therapy. Given its reversible nature and the potential for early intervention, especially in patients with an extreme risk of adverse cardiovascular events, it should receive proper attention. Pharmacotherapy plays an essential role in the treatment of obesity, primarily due to the ineffectiveness of lifestyle modification in many cases.

Type 2 diabetes mellitus

Type 2 diabetes mellitus is another example of comor­bidity associated with inflammation-driven promotion of HFpEF. During periods of uncontrolled hyperglyce­mia, the spontaneous formation of advanced glycation end products (AGEs) has been observed.79, 93, 94 These compounds can promote an inflammatory response by activating the receptors for advanced glycation end prod­ucts (RAGEs), commonly expressed on the surface of endothelial cells, smooth muscle cells and macrophages.95 The binding of AGEs to their receptor triggers a signaling cascade that activates the NF-κB pathway.95 This, in turn, leads to the production and release of pro-inflammatory cytokines, such as TNF-α, IL-1β and IL-6,95 which promote inflammation, cause vascular dysfunction and contribute to HF pathogenesis. A vicious cycle ensues: chronic hyperglycemia leads to increased accumulation of AGEs, resulting in a heightened inflammatory response and cardiovascular damage, which causes deterioration of glucose tolerance.95 Chronic subclinical inflammation results in oxidative stress, endothelial injury, NO signaling deficiency, and altered calcium handling.94, 96 Subsequently, a compensatory response occurs in the form of neurohormonal activation, myocyte hypertrophy and fibrosis. These processes lead to ventricular remodeling, thereby contributing to the onset of HFpEF.79, 87, 94, 96 On account of sympathetic hyperactivity, symptoms such as tachycardia and/or arrhythmias may manifest.96 Therefore, it is important to evaluate the role of T2DM-driven inflammatory changes in the progression of HF, given its reversible character and the potential to counteract the development of HFpEF.2, 14, 87, 97 The significance of SGLT-2 cotransporter inhibitors must be emphasized. The STRIDE trial substantiated the efficacy of semaglutide, demonstrating a 50% reduction in all-cause mortality.27

Treatment

Guideline-directed medical therapy (GDMT) has revolutionized the management of HFrEF, with high-level evidence supporting the use of ACE inhibitors and angiotensin receptor/neprilysin inhibitors (ARNIs), beta-blockers, MRAs, and, more recently, SGLT-2 inhibitors to reduce mortality.98 Emerging studies suggest a potential therapeutic benefit of medication usage in the HFmrEF population.98 In contrast, the treatment options for HFpEF remain limited, with SGLT-2 inhibitors being the only drugs that have demonstrated a clinical effect. Thus, there is a great unmet need for novel therapies that target the pathophysiological mechanisms of HFpEF. The new data concerning systemic inflammation and its potential origins in other systemic diseases may facilitate the identification of the phenotype of HF in patients and the provision of the most effective treatment.

Sodium–glucose cotransporter-2 inhibitors have emerged as promising therapeutic agents for the treatment of HF.33 Their unique anti-inflammatory properties are crucial in the inflammation-based pathophysiology of HFpEF. Moreover, they lower the mortality rate in patients with T2DM.99 In a trial on endothelial cells, it was demonstrated that SGLT-2 inhibitors reduce the expression of NF-κB and MMP-9, thereby lowering the inflammatory pathways.100 Simultaneously, they increase the expression of SIRT6, which plays a role in DNA repair. The utilization of these pharmaceutical agents has been associated with a reduced likelihood of major adverse cardiac events.100 Other clinical trials have noted that SGLT-2 inhibitors lower the hs-CRP levels by over 54% after 1 year of usage in patients with T2DM.101 In light of the EMPEROR-Preserved study findings, special attention should be placed on the implementation of empagliflozin in all HFpEF patients, regardless of the pres­ence or absence of T2DM, given its established efficacy in mitigating HFpEF symptoms and improving prognoses.25

Several studies have demonstrated the efficacy of glucagon-like peptide 1 (GLP-1) receptor agonists, particu­larly semaglutide, in obese HFpEF patients. The administration of these agents resulted in weight loss, enhanced quality of life, and, most importantly, reduced mortality.2, 16, 27, 79, 102, 103 This establishes an important direction in the comprehensive treatment of patients with HF, emphasizing an approach in which obesity is treated as an independent co-occurring disease requiring treatment rather than placing the entire responsibility for alleviating obesity on the patient, which frequently proves unfeasible.

Another area of investigation aimed at finding a more effective method to phenotype patients is myeloperoxidase (MPO) inhibition. During the SATELLITE trial, AZD4831, the MPO inhibitor, has been administered to patients.104 Afterward, the biomarker pathways most related to clinical outcomes in individuals with HFpEF were found to be downregulated. Nevertheless, the cohorts included in this trial were small; therefore, the results must be investigated in a larger group.104

The NLRP3 inflammasome is the next target of anti-inflammatory therapy. Many molecules, including colchicine, GDC-2394 and dapansutrile, are in the stage of clinical development.26, 104, 105, 106 Despite this, colchicine exhibits a strong anti-inflammatory action by targeting the NLRP3 inflammasome and inhibiting the production of pro-inflammatory cytokines, such as IL-1 and IL-6.88, 105 In HFpEF, this property has the potential to reduce SI and possibly regulate maladaptive myocardial remodeling, along with enhancing cardiac function.88 Nevertheless, pro-inflammatory cytokines (IL-1β or IL-6), which are associated with the NLRP3 pathway, have undergone more advanced trials concerning their inhibitors. Anakinra and canakinumab, the inhibitors of IL-1β, have been found to reduce inflammation in patients with CVD. Anakinra has significantly decreased CRP levels in patients with HFpEF.107 The IL-6 inhibitors, namely tocili­zumab (ASSAIL-MI) and ziltivekimab (RESCUE) have reduced systemic inflammation, as evidenced by decreased CRP levels in patients with myocardial infarction and CKD, respectively.108, 109 Lastly, the recent CORTAHF trial has shown that the use of burst steroid therapy resulted in lower inflammation, more effective decongestion, and improvements in quality of life in the AHF population.68, 110, 111, 112, 113 Specifically, this study has demonstrated that 7-day therapy with prednisone at a dose of 40 mg in patients with AHF and elevated hs-CRP led to a reduction in hs-CRP levels on day 7 of therapy, and additionally, on day 90, patients who received burst therapy showed a significantly lower risk of rehospitalization or HF decompensation.68, 110, 111, 112, 113 The graphical summary of therapeutic modalities is demonstrated in Figure 3 and summarized in Table 2.

Smoldering inflammation and oral connections in cardiovascular disease

Recent findings support a correlation between SI and oral diseases.114, 115, 116, 117, 118, 119, 120, 121, 122 Numerous studies have demonstrated that chronic, low-grade inflammation accompanies periodontal disease and is associated with oral microbiome imbalances. These imbalances can contribute to a systemic inflammatory burden, directly increasing cardiovascular risk.114, 115, 116, 117, 118, 119, 120, 121, 122 Smoldering inflammation may be a potential mechanistic link between oral health and CVDs, including HF.119, 120 Periodontal diseases, especially periodontitis, contribute to endothelial dysfunction, oxidative stress and vascular remodeling by mediating inflammation, thereby promoting CVD progression.116 Additionally, oral microbiome dysbiosis, characterized by the entry of pathogenic bacteria and their metabolic products into the circulation, can trigger systemic immune pathway responses and induce SI of distant tissues, which amplifies the negative health outcomes associated with SI.122 The translocation of pathological bacteria may occur during processes such as toothbrushing, flossing and chewing, resulting in minor mechanical injuries in the oral cavity, which are the gateway for bacterial dissemination throughout the vascular system.115 Specific oral bacterial pathogens, such as Porphyromonas gingivalis, have been directly implicated in the initiation and progression of atherosclerotic lesions, further substantiating definitive microbial contribution to cardiovascular risk.115 What is more, overlapping risk factors, such as smoking, obesity and unhealthy diets, sustain persistent inflammatory pathways by establishing optimal conditions for the development of pathological microbiota that promote shared risk between the oral cavity and cardiovascular systems.114 Clinical studies demonstrate that the treatment of oral health conditions (e.g., periodontal disease) may lead to a decreased systemic inflammatory burden and a reduced incidence or progression of CVD.117, 120, 121 Therefore, the maintenance of proper oral health should be prioritized, especially among patients at risk of developing CVD.

Limitations

This review is subject to several limitations. The narrative style of the review, rather than a systematic or meta-analysis approach, raises the possibility of selection bias for the evaluated studies. The second limitation is the heterogeneity of HFpEF phenotypes and variations in study design, study population and biomarker assessment methodology that limit interpretations and generalizability of the findings. The third limitation pertains to the fact that, despite our efforts to provide a comprehensive overview of novel anti-inflammatory therapies, many of the interventions encompassed by this review are based on early-phase trials or relatively small cohorts. Therefore, the long-term efficacy and safety profiles of several interventions remain unknown. The final limitation regards the evolving nature of the field, which may have resulted in the omission of studies that were published at the time of the literature search. In summary, despite these limitations, this review provides context for SI in HFpEF and has identified important considerations for subsequent research.

Clinical implications and future research

Smoldering inflammation plays a crucial role in the pathogenesis of HFpEF, and it should not be overlooked during the diagnosis. The findings summarized in this article can convince clinicians to adopt a more holistic approach to patient care, rather than a narrow focus on one particular disease. Treating the underlying causes of inflammation can positively influence the course of HFpEF. The assessment of the levels of inflammatory markers is a crucial step in the diagnostic process for patients with various medical conditions. Subsequent research on inflammation in HFpEF may result in an update of the guidelines concerning its treatment.

Conclusions

Despite the presence of many known risk factors, recent research has examined the correlation between low-grade chronic inflammation, known as SI, and HF, from its onset to the end stage. A correlation between proinflamma­tory cytokines, such as IL-1β, IL-6, TNF-α, the NF-κB inflammatory pathway, and serum level of hs-CRP, and its impact on the remodeling of heart tissue has been proven. These factors, along with ROS and other proinflammatory molecules and pathways, lead to hypertrophy and fibrosis, therefore impairing heart muscle function and resulting in HF. Other comorbidities related to inflammation, such as AF, T2DM and obesity, have also been associated with the onset of HF, mainly HFpEF. Therefore, treating these conditions with anti-inflammatory medications may result in the improvement during the course of HF. More­over, trials on the efficacy of other inflammatory pathway inhibitors have provided promising outcomes in small clinical groups, suggesting the potential of these agents in HF treatment. The described pathophysiological mechanisms of SI demonstrate the complexity of the subject but also emphasize the importance of a thorough understanding of the topic in order to implement proper treatment. Smoldering inflammation begins as subclinical myocarditis and can remain unnoticed for a long time, finally progressing to post-inflammatory dilated cardiomyo­pathy. Therefore, anti-inflammatory interventions, when administered in conjunction with standard HF treatment, may contribute to the modulation of this progression. Further studies are necessary to optimize timing, dosage and patient selection for maximal benefit.

Ethics approval and consent to participate

Not applicable.

Data availability

Not applicable.

Consent for publication

Not applicable.

Use of AI and AI-assisted technologies

AI-assisted technology (Grammarly; Grammarly Inc., San Francisco, USA) was used for language editing and text refinement. The authors have reviewed and approved all content.

Tables


Table 1. Key findings on biomarkers and pathways of smoldering inflammation (SI) in heart failure (HF)

Biomarker/pathway

Role in HFpEF

Clinical relevance

CRP/hs-CRP

downstream of IL-6; marker of systemic inflammation

predicts HF onset and progression; hsCRP ≥ 2 mg/L identifies HFpEF patients at a higher risk of HF events/mortality

IL-6

central cytokine driving inflammation, fibrosis and endothelial dysfunction

strong predictor of new-onset HFpEF (PREVEND cohort); prognostic for mortality in HFpEF (LUCRIC)

TNF-α

pro-fibrotic, hypertrophic and pro-inflammatory mediator

associated with HF progression; significant in global HF cohorts, less specific in HFpEF

NLRP3 inflammasome

activates caspase-1, leading to IL-1β and IL-18 release; amplifies chronic inflammation

experimental/early clinical target linked to endothelial dysfunction and fibrosis

IL-1β

enhances NF-κB, upregulates IL-6, propagates inflammation

anakinra reduces CRP in HFpEF; canakinumab is effective in CVDs

sST2/IL-33 pathway

cardioprotective; sST2 acts as a decoy, blocking this effect

sST2 predicts rehospitalization and mortality; strong prognostic role in HFpEF

Gal-3

secreted by macrophages; induces fibroblast activation, collagen synthesis and ECM remodeling

predicts new-onset HFpEF, adverse outcomes and the severity of LVDD

GDF-15

marker of inflammation and oxidative stress

independent predictor of adverse outcomes, particularly strong in the HFpEF subgroup

Iron deficiency
(sTfR, hepcidin)

causes impaired oxygen delivery, worsens exercise tolerance and outcomes

predicts mortality; IV iron supplementation improves prognosis

Congestion–inflammation crosstalk

congestion activates inflammation and contributes to lymphatic dysfunction

inflammation and congestion are partly independent pathways; therapeutic implications

CRP – C-reactive protein; CVD – cardiovascular disease; ECM – extracellular matrix; Gal-3 – galectin-3; GDF-15 – growth differentiation factor 15; HF – heart failure; HFpEF – heart failure with preserved ejection fraction; hs-CRP – highly sensitive C-reactive protein; IL – interleukin; IV – intravenous; LVDD – left ventricular diastolic dysfunction; NF-κB – nuclear factor kappa B; sST2 – soluble suppression of tumorgenicity 2; ST2L – suppression of tumorgenicity 2 ligand; sTfR – serum soluble transferrin receptor; TNF-α – tumor necrosis factor alpha.
Table 2. Anti-inflammatory and related therapeutic strategies in heart failure (HF)

Therapy/drug class

Mechanism of action

Clinical relevance in HF

SGLT-2 inhibitors
(e.g., empagliflozin, dapagliflozin)

indirect anti-inflammatory effects: oxidative stress, cytokine activation, improved endothelial function

reduction in HF hospitalization and CV mortality in HFpEF (EMPEROR-Preserved, DELIVER); anti-inflammatory effect

GLP-1 receptor agonists
(e.g., semaglutide)

weight reduction, metabolic improvement, systemic inflammation (IL-6, CRP)

improvement in exercise capacity and symptoms in patients with obesity and HFpEF; ongoing evaluation for CV outcomes

IL-1β inhibitors
(anakinra, canakinumab)

blockade of IL-1β signaling downstream IL-6 and CRP

anakinra: improved CRP, NT-proBNP and symptoms in HFpEF pilot trials; canakinumab: reduced CV events (CANTOS), no HF-specific data

IL-6 pathway inhibitors
(e.g., tocilizumab, ziltivekimab)

blockade of IL-6 signaling

phase 2 data (RESCUE trial) showed CRP reduction; ongoing outcome trials

MPO inhibitors
(e.g., AZD5904)

reduction of oxidative stress and downstream inflammation

preclinical and early clinical development; potential for HFpEF therapy

Corticosteroids

broad suppression of inflammation

limited use; potential benefits in selected HFpEF patients with high SI; long-term safety concerns

Iron supplementation
(IV ferric carboxymaltose)

reversion of iron deficiency, reduction in oxidative stress and inflammation

improvement in exercise capacity and a reduction in HF hospitalizations; benefits extend to HFpEF

Other emerging targets
(e.g., NLRP3 inhibitors, inflammasome modulators)

blockade of IL-1β/IL-18 via the inflammasome pathway

promising preclinical data

GLP-1 – glucagon-like peptide 1; SGLT-2 – sodium–glucose cotransporter-2; MPO – myeloperoxidase; CV – cardiovascular; NLRP3 – nucleotide oligomerization domain-like receptor family pyrin domain containing 3; NT-proBNP – N-terminal pro-B-type natriuretic peptide.

Figures


Fig. 1. Pathophysiological mechanisms of smoldering inflammation (SI) in the development and progression of heart failure (HF)
cGMP – soluble guanylate cyclase; CRP – C-reactive protein; ECM – extracellular matrix; Gal-3 – galectin-3; IFN-γ – interferon gamma; IL – interleukin; NF-κB – nuclear factor kappa B; NLRP3 – nucleotide oligomerization domain-like receptor family pyrin domain containing 3; NO – nitric oxide; PKG – protein kinase G; ROS – reactive oxygen species; sST2 – soluble suppression of tumorgenicity 2; ST2 – suppression of tumorgenicity 2; ST2L – suppression of tumorgenicity 2 ligand; TGF-β – transforming growth factor beta; TNF-α – tumor necrosis factor alpha.
Fig. 2. Multifactorial pathogenesis of heart failure with preserved ejection fraction (HFpEF)
LA – left atrial; LV – left ventricular; NP – natriuretic peptide; RAAS – renin–angiotensin–aldosterone system; RV – right ventricular; SNS – sympathetic nervous system; VSCM – vascular smooth muscle cell.
Fig. 3. Therapeutic modalities in heart failure (HF) with the potential to attenuate inflammatory responses
GLP-1 – glucagon-like peptide 1; HFrEF – heart failure with reduced ejection fraction; SGLT-2 – sodium–glucose cotransporter-2.

References (122)

  1. Savarese G, Becher PM, Lund LH, Seferovic P, Rosano GMC, Coats AJS. Global burden of heart failure: A comprehensive and updated review of epidemiology. Cardiovasc Res. 2023;118(17):3272–3287. doi:10.1093/cvr/cvac013
  2. Beghini A, Sammartino AM, Papp Z, et al. 2024 update in heart failure. ESC Heart Fail. 2025;12(1):8–42. doi:10.1002/ehf2.14857
  3. Sanders‐van Wijk S, van Empel V, Davarzani N, et al.; TIME-CHF investigators. Circulating biomarkers of distinct pathophysiological pathways in heart failure with preserved vs. reduced left ventricular ejection fraction. Eur J Heart Fail. 2015;17(10):1006–1014. doi:10.1002/ejhf.414
  4. Simmonds SJ, Cuijpers I, Heymans S, Jones EAV. Cellular and molecular differences between HFpEF and HFrEF: A step ahead in an improved pathological understanding. Cells. 2020;9(1):242. doi:10.3390/cells9010242
  5. Berger M, März W, Niessner A, et al. IL‐6 and hsCRP predict cardiovascular mortality in patients with heart failure with preserved ejection fraction. ESC Heart Fail. 2024;11(6):3607–3615. doi:10.1002/ehf2.14959
  6. Lakhani I, Wong MV, Hung JKF, et al. Diagnostic and prognostic value of serum C-reactive protein in heart failure with preserved ejection fraction: A systematic review and meta-analysis. Heart Fail Rev. 2021;26(5):1141–1150. doi:10.1007/s10741-020-09927-x
  7. Michou E, Wussler D, Belkin M, et al. Quantifying inflammation using interleukin‐6 for improved phenotyping and risk stratification in acute heart failure. Eur J Heart Fail. 2023;25(2):174–184. doi:10.1002/ejhf.2767
  8. Ren J, Zhao J, Yang S, et al. Transcoronary study of biomarkers in patients with heart failure: Insights into intracardiac production. ESC Heart Fail. 2025;12(3):1640–1651. doi:10.1002/ehf2.15175
  9. Zhu XG, Liu GQ, Peng YP, et al. Causal correlations between inflammatory proteins and heart failure: A two‐sample Mendelian randomization analysis. ESC Heart Fail. 2025;12(2):1374–1385. doi:10.1002/ehf2.15151
  10. Qiu J, Huang X, Kuang M, et al. Evaluating the prognostic value of systemic immune‐inflammatory index in patients with acute decompensated heart failure. ESC Heart Fail. 2024;11(5):3133–3145. doi:10.1002/ehf2.14904
  11. Chen Y, Guan J, Qi C, et al. Association of point‐of‐care testing for sST2 with clinical outcomes in patients hospitalized with heart failure. ESC Heart Fail. 2024;11(5):2857–2868. doi:10.1002/ehf2.14860
  12. Teramoto K, Nochioka K, Sakata Y, Nishimura K, Shimokawa H, Yasuda S; SUPPORT Trial Investigators. Prognostic significance of growth differentiation factor‐15 across age in chronic heart failure. ESC Heart Fail. 2024;11(3):1666–1676. doi:10.1002/ehf2.14738
  13. Gajewski P, Zymlinski R, Biegus J. The critical role of comorbidities in managing heart failure with preserved ejection fraction (HFpEF). ESC Heart Fail. 2025;12(3):1541–1543. doi:10.1002/ehf2.15169
  14. Riccardi M, Sammartino AM, Piepoli M, et al. Heart failure: An update from the last years and a look at the near future. ESC Heart Fail. 2022;9(6):3667–3693. doi:10.1002/ehf2.14257
  15. Lembo M, Strisciuglio T, Fonderico C, et al. Obesity: The perfect storm for heart failure. ESC Heart Fail. 2024;11(4):1841–1860. doi:10.1002/ehf2.14641
  16. Cimino G, Vaduganathan M, Lombardi CM, et al. Obesity, heart failure with preserved ejection fraction, and the role of glucagon‐like peptide‐1 receptor agonists. ESC Heart Fail. 2024;11(2):649–661. doi:10.1002/ehf2.14560
  17. Baumert M, Linz D, Pfeifer M, et al. Hypoxaemic burden in heart failure patients receiving adaptive servo‐ventilation. ESC Heart Fail. 2023;10(6):3725–3728. doi:10.1002/ehf2.14556
  18. Cao Y, Guo S, Dong Y, Liu C, Zhu W. Comparison of liver fibrosis scores for predicting mortality and morbidity in heart failure with preserved ejection fraction. ESC Heart Fail. 2023;10(3):1771–1780. doi:10.1002/ehf2.14336
  19. Huang B, Huang Y, Zhai M, et al. Association of hypoxic burden metrics with cardiovascular outcomes in heart failure and sleep‐disordered breathing. ESC Heart Fail. 2023;10(6):3504–3514. doi:10.1002/ehf2.14526
  20. Mathew D, Kosuru B, Agarwal S, Shrestha U, Sherif A. Impact of sleep apnoea on 30 day hospital readmission rate and cost in heart failure with reduced ejection fraction. ESC Heart Fail. 2023;10(4):2534–2540. doi:10.1002/ehf2.14430
  21. Österman J, Al‐Sodany E, Haugen Löfman I, Barany P, Evans M. Heart failure: The grim reaper of the cardio‐renal‐metabolic triad. ESC Heart Fail. 2024;11(4):2334–2343. doi:10.1002/ehf2.14810
  22. Sharma A, Inzucchi SE, Testani JM, et al. Kidney and heart failure events are bidirectionally associated in patients with type 2 diabetes and cardiovascular disease. ESC Heart Fail. 2024;11(2):737–747. doi:10.1002/ehf2.14601
  23. Baumhove L, van Essen BJ, Dokter MM, et al. IL‐17 is associated with disease severity and targetable inflammatory processes in heart failure. ESC Heart Fail. 2024;11(6):3530–3538. doi:10.1002/ehf2.14968
  24. Eidizadeh A, Schnelle M, Leha A, et al. Biomarker profiles in heart failure with preserved vs. reduced ejection fraction: Results from the DIAST‐CHF study. ESC Heart Fail. 2023;10(1):200–210. doi:10.1002/ehf2.14167
  25. Anker SD, Butler J, Filippatos G, et al.; EMPEROR-Preserved Trial Investigators. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med. 2021;385(16):1451–1461. doi:10.1056/NEJMoa2107038
  26. Tang F, Kunder R, Chu T, et al. First‐in‐human phase 1 trial evaluating safety, pharmacokinetics, and pharmacodynamics of NLRP3 inflammasome inhibitor, GDC‐2394, in healthy volunteers. Clin Transl Sci. 2023;16(9):1653–1666. doi:10.1111/cts.13576
  27. Bonaca MP, Catarig AM, Houlind K, et al.; STRIDE Trial Investigators. Semaglutide and walking capacity in people with symptomatic peripheral artery disease and type 2 diabetes (STRIDE): A phase 3b, double-blind, randomised, placebo-controlled trial. Lancet. 2025;405(10489):1580–1593. doi:10.1016/S0140-6736(25)00509-4
  28. Momomura SI, Ito M. Heart failure mid-range ejection fraction – heart failure with multiple personalities. Circ J. 2019;83(2):274–276. doi:10.1253/circj.CJ-18-1305
  29. Schiattarella GG, Rodolico D, Hill JA. Metabolic inflammation in heart failure with preserved ejection fraction. Cardiovasc Res. 2021;117(2):423–434. doi:10.1093/cvr/cvaa217
  30. Saavedra-Alvarez A, Pereyra KV, Toledo C, Iturriaga R, Del Rio R. Vascular dysfunction in HFpEF: Potential role in the development, maintenance, and progression of the disease. Front Cardiovasc Med. 2022;9:1070935. doi:10.3389/fcvm.2022.1070935
  31. Singh A, Ashraf S, Irfan H, et al. Heart failure and microvascular dysfunction: An in-depth review of mechanisms, diagnostic strategies, and innovative therapies. Ann Med Surg (Lond). 2025;87(2):616–626. doi:10.1097/MS9.0000000000002971
  32. Paulus WJ, Tschöpe C. A novel paradigm for heart failure with preserved ejection fraction: Comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation. J Am Coll Cardiol. 2013;62(4):263–271. doi:10.1016/j.jacc.2013.02.092
  33. Oh JK, Miranda WR, Kane GC. Diagnosis of heart failure with preserved ejection fraction relies on detection of increased diastolic filling pressure, but how? J Am Heart Assoc. 2023;12(6):e028867. doi:10.1161/JAHA.122.028867
  34. Wattanachayakul P, Kittipibul V, Salah HM, et al. Invasive haemodynamic assessment in heart failure with preserved ejection fraction. ESC Heart Fail. 2025;12(3):1558–1570. doi:10.1002/ehf2.15163
  35. Jiang H, Wattanachayakul P, Kittipibul V, et al. Pulmonary artery pressure trajectories in heart failure patients treated with GLP‐1 receptor agonists. ESC Heart Fail. 2025;12(4):2578–2582. doi:10.1002/ehf2.15308
  36. Pugliese NR, Pellicori P, Filidei F, et al. Inflammatory pathways in heart failure with preserved left ventricular ejection fraction: Implications for future interventions. Cardiovasc Res. 2023;118(18):3536–3555. doi:10.1093/cvr/cvac133
  37. Murphy SP, Kakkar R, McCarthy CP, Januzzi JL. Inflammation in heart failure: JACC State-of-the-art review. J Am Coll Cardiol. 2020;75(11):1324–1340. doi:10.1016/j.jacc.2020.01.014
  38. Blevins HM, Xu Y, Biby S, Zhang S. The NLRP3 inflammasome pathway: A review of mechanisms and inhibitors for the treatment of inflammatory diseases. Front Aging Neurosci. 2022;14:879021. doi:10.3389/fnagi.2022.879021
  39. Kramer F, Torzewski J, Kamenz J, et al. Interleukin-1β stimulates acute phase response and C-reactive protein synthesis by inducing an NFκB- and C/EBPβ-dependent autocrine interleukin-6 loop. Mol Immunol. 2008;45(9):2678–2689. doi:10.1016/j.molimm.2007.12.017
  40. Liao K, Lv DY, Yu HL, Chen H, Luo SX. iNOS regulates activation of the NLRP3 inflammasome through the sGC/cGMP/PKG/TACE/TNF-α axis in response to cigarette smoke resulting in aortic endothelial pyroptosis and vascular dysfunction. Int Immunopharmacol. 2021;101(Pt B):108334. doi:10.1016/j.intimp.2021.108334
  41. Cai Z, Wu C, Xu Y, Cai J, Zhao M, Zu L. The NO-cGMP-PKG axis in HFpEF: From pathological mechanisms to potential therapies. Aging Dis. 2023;14(1):46–62. doi:10.14336/AD.2022.0523
  42. Mehrabi Nasab E, Hassanzadeh Makoei R, Aghajani H, Athari SS. IL‐33/ST2 pathway as upper‐hand of inflammation in allergic asthma contributes as predictive biomarker in heart failure. ESC Heart Fail. 2022;9(6):3785–3790. doi:10.1002/ehf2.14111
  43. Zach V, Bähr FL, Edelmann F. Suppression of tumourigenicity 2 in heart failure with preserved ejection fraction. Card Fail Rev. 2020;6:1–7. doi:10.15420/cfr.2019.10
  44. Gordon JW, Shaw JA, Kirshenbaum LA. Multiple facets of NF-κB in the heart: To be or not to NF-κB. Circ Res. 2011;108(9):1122–1132. doi:10.1161/CIRCRESAHA.110.226928
  45. Zaborska B, Sikora-Frąc M, Smarż K, et al. The role of galectin-3 in heart failure – the diagnostic, prognostic and therapeutic potential – where do we stand? Int J Mol Sci. 2023;24(17):13111. doi:10.3390/ijms241713111
  46. Collier P, Watson CJ, Voon V, et al. Can emerging biomarkers of myocardial remodelling identify asymptomatic hypertensive patients at risk for diastolic dysfunction and diastolic heart failure? Eur J Heart Fail. 2011;13(10):1087–1095. doi:10.1093/eurjhf/hfr079
  47. Karolko B, Przewłocka-Kosmala M. Fibrosis markers in heart failure [in Polish]. Folia Cardiol. 2017;12(3):245–253. doi:10.5603/FC.a2016.0072
  48. Hao WR, Cheng CH, Liu JC, Chen HY, Chen JJ, Cheng TH. Understanding galectin-3’s role in diastolic dysfunction: A contemporary perspective. Life (Basel). 2024;14(7):906. doi:10.3390/life14070906
  49. Hunt SA, Baker DW, Chin MH, et al.; American College of Cardiology/American Heart Association. ACC/AHA guidelines for the evaluation and management of chronic heart failure in the adult: Executive summary. A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Committee to revise the 1995 Guidelines for the Evaluation and Management of Heart Failure). J Am Coll Cardiol. 2001;38(7):2101–2113. doi:10.1016/S0735-1097(01)01683-7
  50. Lam CS. Diabetic cardiomyopathy: An expression of stage B heart failure with preserved ejection fraction. Diab Vasc Dis Res. 2015;12(4):234–238. doi:10.1177/1479164115579006
  51. Borlaug BA, Sharma K, Shah SJ, Ho JE. Heart failure with preserved ejection fraction: JACC scientific statement. J Am Coll Cardiol. 2023;81(18):1810–1834. doi:10.1016/j.jacc.2023.01.049
  52. Nawrocka-Millward S, Biegus J, Hurkacz M, et al. Differences in the biomarker profile of de novo acute heart failure versus decompensation of chronic heart failure. Biomolecules. 2021;11(11):1701. doi:10.3390/biom11111701
  53. Albar Z, Albakri M, Hajjari J, Karnib M, Janus SE, Al-Kindi SG. Inflammatory markers and risk of heart failure with reduced to preserved ejection fraction. Am J Cardiol. 2022;167:68–75. doi:10.1016/j.amjcard.2021.11.045
  54. Chia YC, Kieneker LM, van Hassel G, et al. Interleukin 6 and development of heart failure with preserved ejection fraction in the general population. J Am Heart Assoc. 2021;10(11):e018549. doi:10.1161/JAHA.120.018549
  55. DuBrock HM, AbouEzzeddine OF, Redfield MM. High-sensitivity C-reactive protein in heart failure with preserved ejection fraction. PLoS One. 2018;13(8):e0201836. doi:10.1371/journal.pone.0201836
  56. Gui XY, Rabkin SW. C-reactive protein, interleukin-6, trimethylamine-N-oxide, syndecan-1, nitric oxide, and tumor necrosis factor receptor-1 in heart failure with preserved versus reduced ejection fraction: A meta-analysis. Curr Heart Fail Rep. 2023;20(1):1–11. doi:10.1007/s11897-022-00584-9
  57. Ferreira JP, Claggett BL, Liu J, et al. High-sensitivity C-reactive protein in heart failure with preserved ejection fraction: Findings from TOPCAT. Int J Cardiol. 2024;402:131818. doi:10.1016/j.ijcard.2024.131818
  58. Watson CJ, Gallagher J, Wilkinson M, et al. Biomarker profiling for risk of future heart failure (HFpEF) development. J Transl Med. 2021;19(1):61. doi:10.1186/s12967-021-02735-3
  59. Shi Y, Dong G, Liu J, et al. Clinical implications of plasma galectin-3 in heart failure with preserved ejection fraction: A meta-analysis. Front Cardiovasc Med. 2022;9:854501. doi:10.3389/fcvm.2022.854501
  60. Otaki Y, Watanabe T, Shimizu M, et al. Growth differentiation factor‐15 and N‐terminal pro‐BNP in acute heart failure with preserved ejection fraction. ESC Heart Fail. 2025;12(2):888–899. doi:10.1002/ehf2.15068
  61. Yin D, Yan X, Bai X, Tian A, Gao Y, Li J. Prognostic value of growth differentiation factors 15 in acute heart failure patients with preserved ejection fraction. ESC Heart Fail. 2023;10(2):1025–1034. doi:10.1002/ehf2.14271
  62. Song Y, Li F, Xu Y, et al. Prognostic value of sST2 in patients with heart failure with reduced, mid-range and preserved ejection fraction. Int J Cardiol. 2020;304:95–100. doi:10.1016/j.ijcard.2020.01.039
  63. Najjar E, Faxén UL, Hage C, et al. ST2 in heart failure with preserved and reduced ejection fraction. Scand Cardiovasc J. 2019;53(1):21–27. doi:10.1080/14017431.2019.1583363
  64. Zymliński R, Sierpiński R, Metra M, et al. Elevated plasma endothelin‐1 is related to low natriuresis, clinical signs of congestion, and poor outcome in acute heart failure. ESC Heart Fail. 2020;7(6):3536–3544. doi:10.1002/ehf2.13064
  65. Núñez J, de la Espriella R, Rossignol P, et al. Congestion in heart failure: A circulating biomarker‐based perspective. A review from the Biomarkers Working Group of the Heart Failure Association, European Society of Cardiology. Eur J Heart Fail. 2022;24(10):1751–1766. doi:10.1002/ejhf.2664
  66. Zymliński R, Biegus J, Sokolski M, et al. Increased blood lactate is prevalent and identifies poor prognosis in patients with acute heart failure without overt peripheral hypoperfusion. Eur J Heart Fail. 2018;20(6):1011–1018. doi:10.1002/ejhf.1156
  67. Biegus J, Nawrocka-Millward S, Zymliński R, et al. Distinct renin/aldosterone activity profiles correlate with renal function, natriuretic response, decongestive ability and prognosis in acute heart failure. Int J Cardiol. 2021;345:54–60. doi:10.1016/j.ijcard.2021.10.149
  68. Cotter G, Davison BA, Freund Y, et al. Burst steroid therapy for acute heart failure: The CORTAHF randomized, open‐label, pilot trial. Eur J Heart Fail. 2024;26(10):2282–2292. doi:10.1002/ejhf.3452
  69. Salah HM, Biegus J, Fudim M. Role of the renal lymphatic system in heart failure. Curr Heart Fail Rep. 2023;20(2):113–120. doi:10.1007/s11897-023-00595-0
  70. Biegus J, Lindenfeld J, Felker GM, et al. Design and rationale of the eLymTM system for decompensation of excess lymphatic fluid via the thoracic duct in acute heart failure (DELTA‐HF). ESC Heart Fail. 2025;12(3):1719–1726. doi:10.1002/ehf2.15192
  71. Ponikowska B, Zymliński R, Fudim M, et al. Lower extremity lymphatic flow is associated with diuretic response in acute heart failure. Eur J Heart Fail. 2025;27(6):1136–1144. doi:10.1002/ejhf.3655
  72. Ponikowska B, Fudim M, Iwanek G, Zymliński R, Biegus J. Harnessing the lymphatic system. Heart Fail Rev. 2025;30(4):673–683. doi:10.1007/s10741-024-10449-z
  73. Iwanek G, Ponikowska B, Zdanowicz A, et al. Relationship of vascular endothelial growth factor C, a lymphangiogenesis modulator, with edema formation, congestion and outcomes in acute heart failure. J Card Fail. 2023;29(12):1629–1638. doi:10.1016/j.cardfail.2023.04.006
  74. Ponikowska B, Biegus J, Fudim M, et al. Lower extremity lymphatic flow/drainage assessment by indocyanine green fluorescent lymphography in heart failure patients. JACC Basic Transl Sci. 2024;9(7):906–917. doi:10.1016/j.jacbts.2024.02.016
  75. Jankowska EA, Kasztura M, Sokolski M, et al. Iron deficiency defined as depleted iron stores accompanied by unmet cellular iron requirements identifies patients at the highest risk of death after an episode of acute heart failure. Eur Heart J. 2014;35(36):2468–2476. doi:10.1093/eurheartj/ehu235
  76. Biegus J, Zymliński R, Sokolski M, Jankowska EA, Banasiak W, Ponikowski P. Elevated lactate in acute heart failure patients with intracellular iron deficiency as identifier of poor outcome. Kardiol Pol. 2019;77(3):347–354. doi:10.5603/KP.a2019.0014
  77. Ahmed M, Shafiq A, Javaid H, et al. Intravenous iron therapy for heart failure and iron deficiency: An updated meta‐analysis of randomized clinical trials. ESC Heart Fail. 2025;12(1):43–53. doi:10.1002/ehf2.14905
  78. Nemeth E, Ganz T. Hepcidin and iron in health and disease. Annu Rev Med. 2023;74:261–277. doi:10.1146/annurev-med-043021-032816
  79. Anker SD, Usman MS, Anker MS, et al. Patient phenotype profiling in heart failure with preserved ejection fraction to guide therapeutic decision making. A scientific statement of the Heart Failure Association, the European Heart Rhythm Association of the European Society of Cardiology, and the European Society of Hypertension. Eur J Heart Fail. 2023;25(7):936–955. doi:10.1002/ejhf.2894
  80. Chioncel O, Čelutkienė J, Bělohlávek J, et al. Heart failure care in the Central and Eastern Europe and Baltic region: Status, barriers, and routes to improvement. ESC Heart Fail. 2024;11(4):1861–1874. doi:10.1002/ehf2.14687
  81. Volterrani M, Seferovic P, Savarese G, et al. Implementation of guideline‐recommended medical therapy for patients with heart failure in Europe. ESC Heart Fail. 2025;12(2):790–798. doi:10.1002/ehf2.15105
  82. Mesquita T, Lin YN, Ibrahim A. Chronic low‐grade inflammation in heart failure with preserved ejection fraction. Aging Cell. 2021;20(9):e13453. doi:10.1111/acel.13453
  83. Sunaga A, Hikoso S, Yamada T, et al.; OCVC-Heart Failure Investigators. Prognostic impact of Clinical Frailty Scale in patients with heart failure with preserved ejection fraction. ESC Heart Fail. 2021;8(4):3316–3326. doi:10.1002/ehf2.13482
  84. Zahir Anjum D, Bonde AN, Fosbol E, et al. Incidence of clinical outcomes in heart failure patients with and without advanced chronic kidney disease. ESC Heart Fail. 2024;11(5):3406–3415. doi:10.1002/ehf2.14933
  85. Otaki Y, Watanabe T, Shimizu M, et al. Renal tubular damage and clinical outcome in heart failure with preserved ejection fraction and chronic kidney disease. ESC Heart Fail. 2023;10(4):2458–2468. doi:10.1002/ehf2.14378
  86. Gierula J, Cole CA, Drozd M, et al. Atrial fibrillation and risk of progressive heart failure in patients with preserved ejection fraction heart failure. ESC Heart Fail. 2022;9(5):3254–3263. doi:10.1002/ehf2.14004
  87. Colombo G, Biering‐Sorensen T, Ferreira JP, et al. Cardiac remodelling in the era of the recommended four pillars heart failure medical therapy. ESC Heart Fail. 2025;12(2):1029–1044. doi:10.1002/ehf2.15095
  88. Shchendrygina A, Rachina S, Cherkasova N, et al. Colchicine in patients with heart failure and preserved left ventricular ejection fraction: Rationale and design of a prospective, randomised, open-label, crossover clinical trial. Open Heart. 2023;10(2):e002360. doi:10.1136/openhrt-2023-002360
  89. Blüher M. Obesity: Global epidemiology and pathogenesis. Nat Rev Endocrinol. 2019;15(5):288–298. doi:10.1038/s41574-019-0176-8
  90. Litwin SE, Komtebedde J, Seidler T, et al.; REDUCE LAP-HF Investigators and Research Staff. Obesity in heart failure with preserved ejection fraction: Insights from the REDUCE LAP‐HF II trial. Eur J Heart Fail. 2024;26(1):177–189. doi:10.1002/ejhf.3092
  91. Halade GV, Mat Y, Gowda SGB, et al. Sleep deprivation in obesogenic setting alters lipidome and microbiome toward suboptimal inflammation in acute heart failure. FASEB J. 2023;37(5):e22899. doi:10.1096/fj.202300184R
  92. Rafaqat S. Adipokines and their role in heart failure: A literature review. J Innov Card Rhythm Manag. 2023;14(11):5657–5669. doi:10.19102/icrm.2023.14111
  93. Fang Z, Raza U, Song J, et al. Systemic aging fuels heart failure: Molecular mechanisms and therapeutic avenues. ESC Heart Fail. 2025;12(2):1059–1080. doi:10.1002/ehf2.14947
  94. Kaze AD, Yuyun MF, Erqou S, Fonarow GC, Echouffo‐Tcheugui JB. Cardiac autonomic neuropathy and risk of incident heart failure among adults with type 2 diabetes. Eur J Heart Fail. 2022;24(4):634–641. doi:10.1002/ejhf.2432
  95. Yue Q, Song Y, Liu Z, Zhang L, Yang L, Li J. Receptor for advanced glycation end products (RAGE): A pivotal hub in immune diseases. Molecules. 2022;27(15):4922. doi:10.3390/molecules27154922
  96. Konstam MA. Autonomic dysregulation in diabetes: CAN we prevent heart failure? Eur J Heart Fail. 2022;24(4):642–644. doi:10.1002/ejhf.2476
  97. Dihoum A, Brown AJ, McCrimmon RJ, Lang CC, Mordi IR. Dapagliflozin, inflammation and left ventricular remodelling in patients with type 2 diabetes and left ventricular hypertrophy. BMC Cardiovasc Disord. 2024;24(1):356. doi:10.1186/s12872-024-04022-7
  98. McDonagh TA, Metra M, Adamo M, et al.; ESC Scientific Document Group. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur Heart J. 2021;42(36):3599–3726. doi:10.1093/eurheartj/ehab368
  99. Real J, Vlacho B, Ortega E, et al. Cardiovascular and mortality benefits of sodium–glucose co-transporter-2 inhibitors in patients with type 2 diabetes mellitus: CVD-Real Catalonia. Cardiovasc Diabetol. 2021;20(1):139. doi:10.1186/s12933-021-01323-5
  100. D’Onofrio N, Sardu C, Trotta MC, et al. Sodium-glucose co-transporter2 expression and inflammatory activity in diabetic atherosclerotic plaques: Effects of sodium-glucose co-transporter2 inhibitor treatment. Mol Metab. 2021;54:101337. doi:10.1016/j.molmet.2021.101337
  101. Hattori S. Anti-inflammatory effects of empagliflozin in patients with type 2 diabetes and insulin resistance. Diabetol Metab Syndr. 2018;10:93. doi:10.1186/s13098-018-0395-5
  102. Lincoff AM, Brown-Frandsen K, Colhoun HM, et al.; SELECT Trial Investigators. Semaglutide and cardiovascular outcomes in obesity without diabetes. N Engl J Med. 2023;389(24):2221–2232. doi:10.1056/NEJMoa2307563
  103. Kosiborod MN, Abildstrøm SZ, Borlaug BA, et al.; STEP-HFpEF Trial Committees and Investigators. Semaglutide in patients with heart failure with preserved ejection fraction and obesity. N Engl J Med. 2023;389(12):1069–1084. doi:10.1056/NEJMoa2306963
  104. Michaëlsson E, Lund LH, Hage C, et al. Myeloperoxidase inhibition reverses biomarker profiles associated with clinical outcomes in HFpEF. JACC Heart Fail. 2023;11(7):775–787. doi:10.1016/j.jchf.2023.03.002
  105. Vaidya K, Arnott C, Martínez GJ, et al. Colchicine therapy and plaque stabilization in patients with acute coronary syndrome: A CT coronary angiography study. JACC Cardiovasc Imaging. 2018;11(2 Pt 2):305–316. doi:10.1016/j.jcmg.2017.08.013
  106. Wohlford GF, van Tassell BW, Billingsley HE, et al. Phase 1B, randomized, double-blinded, dose escalation, single-center, repeat dose safety and pharmacodynamics study of the oral NLRP3 inhibitor dapansutrile in subjects with NYHA II–III systolic heart failure. J Cardiovasc Pharmacol. 2021;77(1):49–60. doi:10.1097/FJC.0000000000000931
  107. van Tassell BW, Arena R, Biondi-Zoccai G, et al. Effects of interleukin-1 blockade with anakinra on aerobic exercise capacity in patients with heart failure and preserved ejection fraction (from the D-HART pilot study). Am J Cardiol. 2014;113(2):321–327. doi:10.1016/j.amjcard.2013.08.047
  108. Ridker PM, Devalaraja M, Baeres FMM, et al.; RESCUE Investigators. IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): A double-blind, randomised, placebo-controlled, phase 2 trial. Lancet. 2021;397(10289):2060–2069. doi:10.1016/S0140-6736(21)00520-1
  109. Broch K, Anstensrud AK, Woxholt S, et al. Randomized trial of interleukin-6 receptor inhibition in patients with acute ST-segment elevation myocardial infarction. J Am Coll Cardiol. 2021;77(15):1845–1855. doi:10.1016/j.jacc.2021.02.049
  110. Cotter G, Davison B, Freund Y, et al. Corticosteroid burst therapy in patients with acute heart failure: Design of the CORTAHF pilot study. ESC Heart Fail. 2024;11(5):2672–2680. doi:10.1002/ehf2.14930
  111. Cotter G, Davison BA, Mann DL, et al. Acute heart failure: Transitioning from symptom-based care to remission. J Card Fail. 2025:S1071-9164(25)00046-6. doi:10.1016/j.cardfail.2024.12.016
  112. Biegus J, Cotter G, Davison BA, et al. The effects of burst steroid therapy on short-term decongestion in acute heart failure patients with pro-inflammatory activation: A post hoc analysis of the CORTAHF randomized, open-label, pilot trial. J Card Fail. 2025;31(2):354–366. doi:10.1016/j.cardfail.2024.09.002
  113. Pagnesi M, Cotter G, Davison BA, et al. Burst steroid therapy and quality of life in patients with acute heart failure: Insights from the CORTAHF trial. ESC Heart Fail. 2025;12(3):1620–1629. doi:10.1002/ehf2.15235
  114. Deraz O, Rangé H, Boutouyrie P, et al. Oral condition and incident coronary heart disease: A clustering analysis. J Dent Res. 2022;101(5):526–533. doi:10.1177/00220345211052507
  115. Dewan M, Pandit AK, Goyal L. Association of periodontitis and gingivitis with stroke: A systematic review and meta-analysis. Dent Med Probl. 2024;61(3):407–415. doi:10.17219/dmp/158793
  116. Ghorbani P, Rezaei Esfahrood Z, Foroughi M. Paraoxonase-1, a novel link between periodontitis and ischemic heart disease: A case–control study. Dent Med Probl. 2023;60(1):55–59. doi:10.17219/dmp/152181
  117. Hopkins S, Gajagowni S, Qadeer Y, et al. Oral health and cardiovascular disease. Am J Med. 2024;137(4):304–307. doi:10.1016/j.amjmed.2023.11.022
  118. Isola G, Santonocito S, Lupi SM, et al. Periodontal health and disease in the context of systemic diseases. Mediators Inflamm. 2023;2023:9720947. doi:10.1155/2023/9720947
  119. Narendran N, Shenoy S, Kodangala S, Raghavendra Vamsi A, Kamath V. Assessment of myocardial strain in hypertensive patients with periodontitis. Dent Med Probl. 2023;60(1):61–69. doi:10.17219/dmp/152707
  120. Sanz M, Marco Del Castillo A, Jepsen S, et al. Periodontitis and cardiovascular diseases: Consensus report. J Clin Periodontol. 2020;47(3):268–288. doi:10.1111/jcpe.13189
  121. Sumayin Ngamdu K, Mallawaarachchi I, Dunipace EA, et al. Association between periodontal disease and cardiovascular disease (from the NHANES). Am J Cardiol. 2022;178:163–168. doi:10.1016/j.amjcard.2022.05.028
  122. Tonelli A, Lumngwena EN, Ntusi NAB. The oral microbiome in the pathophysiology of cardiovascular disease. Nat Rev Cardiol. 2023;20(6):386–403. doi:10.1038/s41569-022-00825-3