1
|
Morrissey SM, Kirkland LG, Phillips TK, Levit RD, Hopke A, Jensen BC. Multifaceted roles of neutrophils in cardiac disease. J Leukoc Biol 2025; 117:qiaf017. [PMID: 39936506 DOI: 10.1093/jleuko/qiaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/15/2025] [Accepted: 02/11/2025] [Indexed: 02/13/2025] Open
Abstract
Neutrophils, the most abundant leukocytes in human blood, have long been recognized as critical first responders in the innate immune system's defense against pathogens. Some of the more notable innate antimicrobial properties of neutrophils include generation of superoxide free radicals like myeloperoxidase, production of proteases that reshape the extracellular matrix allowing for easier access to infected tissues, and release of neutrophil extracellular traps, extruded pieces of DNA that ensnare bacterial and fungi. These mechanisms developed to provide neutrophils with a vast array of specialized functions to provide the host defense against infection in an acute setting. However, emerging evidence over the past few decades has revealed a far more complex and nuanced role for these neutrophil-driven processes in various chronic conditions, particularly in cardiovascular diseases. The pathophysiology of cardiac diseases involves a complex interplay of hemodynamic, neurohumoral, and inflammatory factors. Neutrophils, as key mediators of inflammation, contribute significantly to this intricate network. Their involvement extends far beyond their classical role in pathogen clearance, encompassing diverse functions that can both exacerbate tissue damage and contribute to repair processes. Here, we consider the contributions of neutrophils to myocardial infarction, heart failure, cardiac arrhythmias, and nonischemic cardiomyopathies. Understanding these complex interactions is crucial for developing novel therapeutic strategies aimed at modulating neutrophil functions in these highly morbid cardiac diseases.
Collapse
Affiliation(s)
- Samantha M Morrissey
- Department of Medicine, University of North Carolina School of Medicine, 125 MacNider Hall, Chapel Hill, NC 27599-7005, United States
| | - Logan G Kirkland
- McAllister Heart Institute, University of North Carolina School of Medicine, 111 Mason Farm Rd., Chapel Hill, NC 27599-7126, United States
| | - Tasha K Phillips
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, PO Box 70577, Johnson City, TN 37614, United States
| | - Rebecca D Levit
- Division of Cardiology, Department of Medicine, Emory University, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Alex Hopke
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, PO Box 70577, Johnson City, TN 37614, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, PO Box 70300, Johnson City, TN 37614, United States
| | - Brian C Jensen
- Department of Medicine, University of North Carolina School of Medicine, 125 MacNider Hall, Chapel Hill, NC 27599-7005, United States
- McAllister Heart Institute, University of North Carolina School of Medicine, 111 Mason Farm Rd., Chapel Hill, NC 27599-7126, United States
- Department of Pharmacology, University of North Carolina School of Medicine, 120 Mason Farm Rd., Chapel Hill, NC 27599-7365, United States
| |
Collapse
|
2
|
Haim IR, Gruber A, Kazma N, Bashai C, Lichtig Kinsbruner H, Caspi O. Modeling Heart Failure With Preserved Ejection Fraction Using Human Induced Pluripotent Stem Cell-Derived Cardiac Organoids. Circ Heart Fail 2025; 18:e011690. [PMID: 39873109 DOI: 10.1161/circheartfailure.124.011690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND The therapeutic armamentarium for heart failure with preserved ejection fraction (HFpEF) remains notably constrained. A factor contributing to this problem could be the scarcity of in vitro models for HFpEF, which hinders progress in developing new therapeutic strategies. Here, we aimed at developing a novel, comorbidity-inspired, human, in vitro model for HFpEF. METHODS Human induced pluripotent stem cells-derived cardiomyocytes were used to produce cardiac organoids. The generated organoids were then subjected to HFpEF-associated, comorbidity-inspired conditions, such as hypertension, diabetes, and obesity-related inflammation. To assess the development of HFpEF pathophysiological features, organoids were thoroughly evaluated for their structural, functional, electrophysiological, and metabolic properties. RESULTS Exposure to the combination of all comorbidity-mimicking conditions resulted in the largest cellular volume of 1692±52 versus 1346±84 µm3 in RPMI (Roswell Park Memorial Institute medium) control group (P=0.003), while lower in obesity, hypertension, and diabetes groups: 1059±40 µm3 (P=0.014), 1276±35 µm3 (P=0.940), and 1575±70 µm3 (P=0.146), respectively. Similarly, ultrastructural fibrosis was most significantly observed after exposure to the combination of all HFpEF-inducing conditions 14.6±1.2% compared with single condition exposure 5.2±1.3% (obesity), 6.7±3.5% (hypertension), and 9.0±1.1% (diabetes; P<0.001). Moreover, HFpEF-related conditions led to an increase in passive force compared with control (7.52±1.08 versus 2.33±0.46 mN/mm, P<0.001), whereas no significant alterations were noted in active contractile forces. Relaxation constant τ was significantly prolonged after exposure to HFpEF conditions showing a prolongation of 95.9 ms (36.4-106.4; P=0.028) compared with a shortening of 35.6 ms (43.3-67.3; P=0.80) in the control. Finally, organoid exposure to HFpEF conditions led to a significant increase in oxidative stress levels and a significant decline in oxygen consumption rate. CONCLUSIONS We established a novel, human, in vitro model for HFpEF, based on comorbidity-inspired conditions. The model faithfully recapitulated the structural, functional, and mechanistic features of HFpEF. This model holds the potential to provide mechanistic insights and facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Idan Refael Haim
- Bruce Rapport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel (I.R.H., N.K., C.B., O.C.)
- The Clinical Research Institute at Rambam, Haifa, Israel (I.R.H., A.G., N.K., C.B., H.L.K., O.C.)
| | - Amit Gruber
- The Clinical Research Institute at Rambam, Haifa, Israel (I.R.H., A.G., N.K., C.B., H.L.K., O.C.)
- The Heart Failure Unit, Department of Cardiology, Rambam Health Care Campus, Haifa, Israel (A.G., H.L.K., O.C.)
| | - Noam Kazma
- Bruce Rapport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel (I.R.H., N.K., C.B., O.C.)
- The Clinical Research Institute at Rambam, Haifa, Israel (I.R.H., A.G., N.K., C.B., H.L.K., O.C.)
| | - Caroline Bashai
- Bruce Rapport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel (I.R.H., N.K., C.B., O.C.)
- The Clinical Research Institute at Rambam, Haifa, Israel (I.R.H., A.G., N.K., C.B., H.L.K., O.C.)
| | - Hava Lichtig Kinsbruner
- The Clinical Research Institute at Rambam, Haifa, Israel (I.R.H., A.G., N.K., C.B., H.L.K., O.C.)
- The Heart Failure Unit, Department of Cardiology, Rambam Health Care Campus, Haifa, Israel (A.G., H.L.K., O.C.)
| | - Oren Caspi
- Bruce Rapport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel (I.R.H., N.K., C.B., O.C.)
- The Clinical Research Institute at Rambam, Haifa, Israel (I.R.H., A.G., N.K., C.B., H.L.K., O.C.)
- The Heart Failure Unit, Department of Cardiology, Rambam Health Care Campus, Haifa, Israel (A.G., H.L.K., O.C.)
| |
Collapse
|
3
|
Mo B, Ding Y, Ji Q. NLRP3 inflammasome in cardiovascular diseases: an update. Front Immunol 2025; 16:1550226. [PMID: 40079000 PMCID: PMC11896874 DOI: 10.3389/fimmu.2025.1550226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular disease (CVD) continues to be the leading cause of mortality worldwide. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome is involved in numerous types of CVD. As part of innate immunity, the NLRP3 inflammasome plays a vital role, requiring priming and activation signals to trigger inflammation. The NLRP3 inflammasome leads both to the release of IL-1 family cytokines and to a distinct form of programmed cell death called pyroptosis. Inflammation related to CVD has been extensively investigated in relation to the NLRP3 inflammasome. In this review, we describe the pathways triggering NLRP3 priming and activation and discuss its pathogenic effects on CVD. This study also provides an overview of potential therapeutic approaches targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Binhai Mo
- People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yudi Ding
- First People’s Hospital of Nanning, Nanning, Guangxi, China
| | - Qingwei Ji
- People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
4
|
Fayyaz AU, Eltony M, Prokop LJ, Koepp KE, Borlaug BA, Dasari S, Bois MC, Margulies KB, Maleszewski JJ, Wang Y, Redfield MM. Pathophysiological insights into HFpEF from studies of human cardiac tissue. Nat Rev Cardiol 2025; 22:90-104. [PMID: 39198624 PMCID: PMC11750620 DOI: 10.1038/s41569-024-01067-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2024] [Indexed: 09/01/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major, worldwide health-care problem. Few therapies for HFpEF exist because the pathophysiology of this condition is poorly defined and, increasingly, postulated to be diverse. Although perturbations in other organs contribute to the clinical profile in HFpEF, altered cardiac structure, function or both are the primary causes of this heart failure syndrome. Therefore, studying myocardial tissue is fundamental to improve pathophysiological insights and therapeutic discovery in HFpEF. Most studies of myocardial changes in HFpEF have relied on cardiac tissue from animal models without (or with limited) confirmatory studies in human cardiac tissue. Animal models of HFpEF have evolved based on theoretical HFpEF aetiologies, but these models might not reflect the complex pathophysiology of human HFpEF. The focus of this Review is the pathophysiological insights gained from studies of human HFpEF myocardium. We outline the rationale for these studies, the challenges and opportunities in obtaining myocardial tissue from patients with HFpEF and relevant comparator groups, the analytical approaches, the pathophysiological insights gained to date and the remaining knowledge gaps. Our objective is to provide a roadmap for future studies of cardiac tissue from diverse cohorts of patients with HFpEF, coupling discovery biology with measures to account for pathophysiological diversity.
Collapse
Affiliation(s)
- Ahmed U Fayyaz
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Muhammad Eltony
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Larry J Prokop
- Mayo Clinic College of Medicine and Science, Library Reference Service, Rochester, MN, USA
| | - Katlyn E Koepp
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Barry A Borlaug
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Surendra Dasari
- Mayo Clinic College of Medicine and Science, Computational Biology, Rochester, MN, USA
| | - Melanie C Bois
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joesph J Maleszewski
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ying Wang
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Margaret M Redfield
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
5
|
Fuerlinger A, Stockner A, Sedej S, Abdellatif M. Caloric restriction and its mimetics in heart failure with preserved ejection fraction: mechanisms and therapeutic potential. Cardiovasc Diabetol 2025; 24:21. [PMID: 39827109 PMCID: PMC11742808 DOI: 10.1186/s12933-024-02566-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025] Open
Abstract
The global increase in human life expectancy, coupled with an unprecedented rise in the prevalence of obesity, has led to a growing clinical and socioeconomic burden of heart failure with preserved ejection fraction (HFpEF). Mechanistically, the molecular and cellular hallmarks of aging are omnipresent in HFpEF and are further exacerbated by obesity and associated metabolic diseases. Conversely, weight loss strategies, particularly caloric restriction, have shown promise in improving health status in patients with HFpEF and are considered the gold standard for promoting longevity and healthspan (disease-free lifetime) in model organisms. In this review, we implicate fundamental mechanisms of aging in driving HFpEF and elucidate how caloric restriction mitigates the disease progression. Furthermore, we discuss the potential for pharmacologically mimicking the beneficial effects of caloric restriction in HFpEF using clinically approved and emerging caloric restriction mimetics. We surmise that these compounds could offer novel therapeutic avenues for HFpEF and alleviate the challenges associated with the implementation of caloric restriction and other lifestyle modifications to reduce the burden of HFpEF at a population level.
Collapse
Affiliation(s)
- Alexander Fuerlinger
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
- BioTechMed-Graz, 8010, Graz, Austria
| | - Alina Stockner
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
- BioTechMed-Graz, 8010, Graz, Austria
- Faculty of Medicine, University of Maribor, 2000, Maribor, Slovenia
| | - Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria.
- BioTechMed-Graz, 8010, Graz, Austria.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805, Villejuif, France.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, 75006, France.
| |
Collapse
|
6
|
Yang S, Penna V, Lavine KJ. Functional diversity of cardiac macrophages in health and disease. Nat Rev Cardiol 2025:10.1038/s41569-024-01109-8. [PMID: 39743564 DOI: 10.1038/s41569-024-01109-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Macrophages make up a substantial portion of the stromal compartment of the heart in health and disease. In the past decade, the origins of these cardiac macrophages have been established as two broad populations derived from either embryonic or definitive haematopoiesis and that can be distinguished by the expression of CC-motif chemokine receptor 2 (CCR2). These cardiac macrophage populations are transcriptionally distinct and have differing cell surface markers and divergent roles in cardiac homeostasis and disease. Embryonic-derived CCR2- macrophages are a tissue-resident population that participates in tissue development, repair and maintenance, whereas CCR2+ macrophages are derived from definitive haematopoiesis and contribute to inflammation and tissue damage. Studies from the past 5 years have leveraged single-cell RNA sequencing technologies to expand our understanding of cardiac macrophage diversity, particularly of the monocyte-derived macrophage populations that reside in the injured and diseased heart. Emerging technologies in spatial transcriptomics have enabled the identification of distinct disease-associated cellular neighbourhoods consisting of macrophages, other immune cells and fibroblasts, highlighting the involvement of macrophages in cell-cell communication. Together, these discoveries lend new insights into the role of specific macrophage populations in the pathogenesis of cardiac disease, which can pave the way for the identification of new therapeutic targets and the development of diagnostic tools. In this Review, we discuss the developmental origin of cardiac macrophages and describe newly identified cell states and associated cellular neighbourhoods in the steady state and injury settings. We also discuss various contributions and effector functions of cardiac macrophages in homeostasis and disease.
Collapse
Affiliation(s)
- Steven Yang
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Vinay Penna
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
7
|
Thorp EB, Filipp M. Contributions of Inflammation to Cardiometabolic Heart Failure with Preserved Ejection Fraction. ANNUAL REVIEW OF PATHOLOGY 2025; 20:143-167. [PMID: 39357068 DOI: 10.1146/annurev-pathmechdis-111523-023405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
The most common form of heart failure is heart failure with preserved ejection fraction (HFpEF). While heterogeneous in origin, the most common form of HFpEF is the cardiometabolic manifestation. Obesity and aging promote systemic inflammation that appears integral to cardiometabolic HFpEF pathophysiology. Accumulation of immune cells within the heart, fueled by an altered metabolome, contribute to cardiac inflammation and fibrosis. In spite of this, broad anti-inflammatory therapy has not shown significant benefit in patient outcomes. Thus, understanding of the nuances to metabolic and age-related inflammation during HFpEF is paramount for more targeted interventions. Here, we review clinical evidence of inflammation in the context of HFpEF and summarize our mechanistic understanding of immunometabolic inflammation, highlighting pathways of therapeutic potential along the way.
Collapse
Affiliation(s)
- Edward B Thorp
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; ,
| | - Mallory Filipp
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; ,
| |
Collapse
|
8
|
Potere N, Bonaventura A, Abbate A. Novel Therapeutics and Upcoming Clinical Trials Targeting Inflammation in Cardiovascular Diseases. Arterioscler Thromb Vasc Biol 2024; 44:2371-2395. [PMID: 39387118 PMCID: PMC11602387 DOI: 10.1161/atvbaha.124.319980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Cardiovascular disease (CVD) remains a major health burden despite significant therapeutic advances accomplished over the last decades. It is widely and increasingly recognized that systemic inflammation not only represents a major cardiovascular risk and prognostic factor but also plays key pathogenic roles in CVD development and progression. Despite compelling preclinical evidence suggesting large potential of anti-inflammatory pharmacological interventions across numerous CVDs, clinical translation remains incomplete, mainly due to (1) yet undefined molecular signaling; (2) challenges of safety and efficacy profile of anti-inflammatory drugs; and (3) difficulties in identifying optimal patient candidates and responders to anti-inflammatory therapeutics, as well as optimal therapeutic windows. Randomized controlled trials demonstrated the safety/efficacy of canakinumab and colchicine in secondary cardiovascular prevention, providing confirmation for the involvement of a specific inflammatory pathway (NLRP3 [NACHT, LRR, and PYD domain-containing protein 3] inflammasome/IL [interleukin]-1β) in atherosclerotic CVD. Colchicine was recently approved by the US Food and Drug Administration for this indication. Diverse anti-inflammatory drugs targeting distinct inflammatory pathways are widely used for the management of other CVDs including myocarditis and pericarditis. Ongoing research efforts are directed to implementing anti-inflammatory therapeutic strategies across a growing number of CVDs, through repurposing of available anti-inflammatory drugs and development of novel anti-inflammatory compounds, which are herein concisely discussed. This review also summarizes the main characteristics and findings of completed and upcoming randomized controlled trials directly targeting inflammation in CVDs, and discusses major challenges and future perspectives in the exciting and constantly expanding landscape of cardioimmunology.
Collapse
Affiliation(s)
- Nicola Potere
- Department of Medicine and Ageing Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Aldo Bonaventura
- Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi Varese, Italy
| | - Antonio Abbate
- Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
9
|
Castillo RL, Farías J, Sandoval C, González-Candia A, Figueroa E, Quezada M, Cruz G, Llanos P, Jorquera G, Kostin S, Carrasco R. Role of NLRP3 Inflammasome in Heart Failure Patients Undergoing Cardiac Surgery as a Potential Determinant of Postoperative Atrial Fibrillation and Remodeling: Is SGLT2 Cotransporter Inhibition an Alternative for Cardioprotection? Antioxidants (Basel) 2024; 13:1388. [PMID: 39594530 PMCID: PMC11591087 DOI: 10.3390/antiox13111388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
In heart failure (HF) patients undergoing cardiac surgery, an increased activity of mechanisms related to cardiac remodeling may determine a higher risk of postoperative atrial fibrillation (POAF). Given that atrial fibrillation (AF) has a negative impact on the course and management of HF, including the need for anticoagulation therapy, identifying the factors associated with AF occurrence after cardiac surgery is crucial for the prognosis of these patients. POAF is thought to occur when various clinical and biochemical triggers act on susceptible cardiac tissue (first hit), with oxidative stress and inflammation during cardiopulmonary bypass (CPB) surgery being potential contributing factors (second hit). However, the molecular mechanisms involved in these processes remain poorly characterized. Recent research has shown that patients who later develop POAF often have pre-existing abnormalities in calcium handling and activation of NLRP3-inflammasome signaling in their atrial cardiomyocytes. These molecular changes may make cardiomyocytes more susceptible to spontaneous Ca2+-releases and subsequent arrhythmias, particularly when exposed to inflammatory mediators. Additionally, some clinical studies have linked POAF with elevated preoperative inflammatory markers, but there is a need for further research in order to better understand the impact of CPB surgery on local and systemic inflammation. This knowledge would make it possible to determine whether patients susceptible to POAF have pre-existing inflammatory conditions or cellular electrophysiological factors that make them more prone to developing AF and cardiac remodeling. In this context, the NLRP3 inflammasome, expressed in cardiomyocytes and cardiac fibroblasts, has been identified as playing a key role in the development of HF and AF, making patients with pre-existing HF with reduced ejection fraction (HFrEF) the focus of several clinical studies with interventions that act at this level. On the other hand, HFpEF has been linked to metabolic and non-ischemic risk factors, but more research is needed to better characterize the myocardial remodeling events associated with HFpEF. Therefore, since ventricular remodeling may differ between HFrEF and HFpEF, it is necessary to perform studies in both groups of patients due to their pathophysiological variations. Clinical evidence has shown that pharmacological therapies that are effective for HFrEF may not provide the same anti-remodeling benefits in HFpEF patients, particularly compared to traditional adrenergic and renin-angiotensin-aldosterone system inhibitors. On the other hand, there is growing interest in medications with pleiotropic or antioxidant/anti-inflammatory effects, such as sodium-glucose cotransporter 2 inhibitors (SGLT-2is). These drugs may offer anti-remodeling effects in both HFrEF and HFpEF by inhibiting pro-inflammatory, pro-oxidant, and NLRP3 signaling pathways and their mediators. The anti-inflammatory, antioxidant, and anti-remodeling effects of SGLT-2 i have progressively expanded from HFrEF and HFpEF to other forms of cardiac remodeling. However, these advances in research have not yet encompassed POAF despite its associations with inflammation, oxidative stress, and remodeling. Currently, the direct or indirect effects of NLRP3-dependent pathway inhibition on the occurrence of POAF have not been clinically assessed. However, given that NLRP3 pathway inhibition may also indirectly affect other pathways, such as inhibition of NF-kappaB or inhibition of matrix synthesis, which are strongly linked to POAF and cardiac remodeling, it is reasonable to hypothesize that this type of intervention could play a role in preventing these events.
Collapse
Affiliation(s)
- Rodrigo L. Castillo
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- Unidad de Paciente Crítico, Hospital del Salvador, Santiago 7500922, Chile
| | - Jorge Farías
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile;
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| | - Alejandro González-Candia
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2841959, Chile; (A.G.-C.); (E.F.)
| | - Esteban Figueroa
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2841959, Chile; (A.G.-C.); (E.F.)
| | - Mauricio Quezada
- Facultad de Medicina, Universidad Finis Terrae, Santiago 7501015, Chile;
| | - Gonzalo Cruz
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile;
| | - Paola Llanos
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago 8380544, Chile
| | - Gonzalo Jorquera
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile;
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago 8331051, Chile;
| | - Sawa Kostin
- Faculty of Health Sciences, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany;
| | - Rodrigo Carrasco
- Departamento de Cardiología, Clínica Alemana de Santiago, Santiago 7500922, Chile;
| |
Collapse
|
10
|
Wang H, Hu X, Zhang Y, Zhu A, Fan J, Wu Z, Wang X, Hu W, Ju D. Simultaneously blocking ANGPTL3 and IL-1β for the treatment of atherosclerosis through lipid-lowering and anti-inflammation. Inflamm Res 2024; 73:1931-1944. [PMID: 39254873 DOI: 10.1007/s00011-024-01941-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/10/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
OBJECTIVE Blood lipid levels play a critical role in the progression of atherosclerosis. However, even with adequate lipid reduction, significant residual cardiovascular risk remains. Therefore, it is necessary to seek novel therapeutic strategies for atherosclerosis that can not only lower lipid levels but also inhibit inflammation simultaneously. METHODS The fusion protein FD03-IL-1Ra was designed by linking the Angiopoietin-like 3 (ANGPTL3) nanobody and human interleukin-1 receptor antagonist (IL-1Ra) sequences to a mutated human immunoglobulin gamma 1 (IgG1) Fc. This construct was transfected into HEK293 cells for expression. The purity and thermal stability of the fusion protein were assessed using SDS-PAGE, SEC-HPLC, and differential scanning calorimetry. Binding affinities of the fusion protein to ANGPTL3 and IL-1 receptor were measured using Biacore T200. The biological activity of the fusion protein was validated through in vitro experiments. The therapeutic efficacy of the fusion protein was evaluated in an ApoE-/- mouse model of atherosclerosis, including serum lipid level determination, histological analysis of aorta and aortic sinus sections, and detection of inflammatory and oxidative stress markers. ImageJ software was utilized for quantitative image analysis. Statistical analysis was performed using one-way ANOVA followed by Bonferroni post hoc test. RESULTS The FD03-IL-1Ra fusion protein was successfully expressed, with no polymer formation detected, and it demonstrated good thermal and conformational stability. High affinity for both murine and human ANGPTL3 was exhibited by FD03-IL-1Ra, and it was able to antagonize hANGPTL3's inhibition of LPL activity. FD03-IL-1Ra also showed high affinity for both murine and human IL-1R, inhibiting IL-6 expression in A549 cells induced by IL-1β stimulation, as well as suppressing IL-1β-induced activity inhibition in A375.S2 cells. Our study revealed that the fusion protein effectively lowered serum lipid levels and alleviated inflammatory responses in mice. Furthermore, the fusion protein enhanced plaque stability by increasing collagen content within atherosclerotic plaques. CONCLUSIONS These findings highlighted the potential of bifunctional interleukin-1 receptor antagonist and ANGPTL3 antibody fusion proteins for ameliorating the progression of atherosclerosis, presenting a promising novel therapeutic approach targeting both inflammation and lipid levels.
Collapse
Affiliation(s)
- Hanqi Wang
- Department of Cardiology, Minhang Hospital, Fudan University, No. 170 Zisong Road, Minhang District, Shanghai, China
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, No. 826 Zhangheng Road, Pudong New District, Shanghai, China
| | - Xiaozhi Hu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, No. 826 Zhangheng Road, Pudong New District, Shanghai, China
| | - Yuting Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, No. 826 Zhangheng Road, Pudong New District, Shanghai, China
| | - An Zhu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, No. 826 Zhangheng Road, Pudong New District, Shanghai, China
| | - Jiajun Fan
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, No. 826 Zhangheng Road, Pudong New District, Shanghai, China
| | - Zhengyu Wu
- TAU Cambridge Ltd, The Bradfield Centre UNIT 184, Cambridge Science Park, Cambridge, CB4 0GA, UK
| | - Xuebin Wang
- Department of Pharmacy, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, 355 Luding Road, Putuo District, Shanghai, China.
| | - Wei Hu
- Department of Cardiology, Minhang Hospital, Fudan University, No. 170 Zisong Road, Minhang District, Shanghai, China.
| | - Dianwen Ju
- Department of Cardiology, Minhang Hospital, Fudan University, No. 170 Zisong Road, Minhang District, Shanghai, China.
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, No. 826 Zhangheng Road, Pudong New District, Shanghai, China.
| |
Collapse
|
11
|
Liu H, Magaye R, Kaye DM, Wang BH. Heart failure with preserved ejection fraction: The role of inflammation. Eur J Pharmacol 2024; 980:176858. [PMID: 39074526 DOI: 10.1016/j.ejphar.2024.176858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Heart failure (HF) is a debilitating clinical syndrome affecting 64.3 million patients worldwide. More than 50% of HF cases are attributed to HF with preserved ejection fraction (HFpEF), an entity growing in prevalence and mortality. Although recent breakthroughs reveal the prognostic benefits of sodium-glucose co-transporter 2 inhibitors (SGLT2i) in HFpEF, there is still a lack of effective pharmacological therapy available. This highlights a major gap in medical knowledge that must be addressed. Current evidence attributes HFpEF pathogenesis to an interplay between cardiometabolic comorbidities, inflammation, and renin-angiotensin-aldosterone-system (RAAS) activation, leading to cardiac remodelling and diastolic dysfunction. However, conventional RAAS blockade has demonstrated limited benefits in HFpEF, which emphasises that alternative therapeutic targets should be explored. Presently, there is limited literature examining the use of anti-inflammatory HFpEF therapies despite growing evidence supporting its importance in disease progression. Hence, this review aims to explore current perspectives on HFpEF pathogenesis, including the importance of inflammation-driven cardiac remodelling and the therapeutic potential of anti-inflammatory therapies.
Collapse
Affiliation(s)
- Hongyi Liu
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia; Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - Ruth Magaye
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - David M Kaye
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - Bing H Wang
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia; Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| |
Collapse
|
12
|
Cotter G, Davison B, Freund Y, Mebazaa A, Voors A, Edwards C, Novosadova M, Takagi K, Hayrapetyan H, Mshetsyan A, Mayranush D, Cohen‐Solal A, ter Maaten JM, Biegus J, Ponikowski P, Filippatos G, Chioncel O, Pagnesi M, Simon T, Metra M, Mann DL. Corticosteroid burst therapy in patients with acute heart failure: Design of the CORTAHF pilot study. ESC Heart Fail 2024; 11:2672-2680. [PMID: 38943232 PMCID: PMC11424380 DOI: 10.1002/ehf2.14930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024] Open
Abstract
AIMS Inflammation has emerged as a potential key pathophysiological mechanism in heart failure (HF) in general and acute HF (AHF) specifically, with inflammatory biomarkers shown to be highly predictive of adverse outcomes in these patients. The CORTAHF study builds on both these data and the fact that steroid burst therapy has been shown to be effective in the treatment of respiratory diseases and COVID-19. Our hypothesis is that in patients with AHF and elevated C-reactive protein (CRP) levels without symptoms or signs of infection, a 7-day course of steroid therapy will lead to reduced inflammation and short-term improvement in quality of life and a reduced risk of worsening HF (WHF) events. METHODS AND RESULTS The study, which is currently ongoing, will include 100 patients with AHF ages 18-85, regardless of ejection fraction, screened within 12 h of presentation. Patients will be included who have NT-proBNP > 1500 pg/mL and CRP > 20 mg/L at screening. Exclusion criteria include haemodynamic instability and symptoms and signs of infection. After signed consent, eligible patients will be randomized according to a central randomization scheme stratified by centre 1:1 to either treatment once daily for 7 days with 40 mg prednisone orally or to standard care. Patients will be assessed at study day 2, day 4 or at discharge if earlier, and at days 7 and 31 at the hospital; and at day 91 through a telephone follow-up. The primary endpoint is the change in CRP level from baseline to day 7, estimated from a mixed model for repeated measures (MMRM) including all measured timepoints, in patients without a major protocol violation. Secondary endpoints include the time to the first event of WHF adverse event, readmission for HF, or death through day 91; and changes to day 7 in EQ-5D visual analogue scale score and utility index. Additional clinical and laboratory measures will be assessed. CONCLUSIONS The results of the study will add to the knowledge of the role of inflammation in AHF and potentially inform the design of larger studies with possibly longer duration of anti-inflammatory therapies in AHF.
Collapse
Affiliation(s)
- Gad Cotter
- Université Paris Cité, INSERM UMR‐S 942 (MASCOT)ParisFrance
- Heart InitiativeDurhamNCUSA
- Momentum Research, Inc.DurhamNCUSA
| | - Beth Davison
- Université Paris Cité, INSERM UMR‐S 942 (MASCOT)ParisFrance
- Heart InitiativeDurhamNCUSA
- Momentum Research, Inc.DurhamNCUSA
| | - Yonathan Freund
- IMProving Emergency Care FHUSorbonne UniversitéParisFrance
- Emergency Department and Service Mobile d'Urgence et de Réanimation (SMUR)Hôpital Pitié‐Salpêtrière, Assistance Publique‐Hôpitaux de Paris (AP‐HP)ParisFrance
| | - Alexandre Mebazaa
- Université Paris Cité, INSERM UMR‐S 942 (MASCOT)ParisFrance
- Department of Anesthesiology and Critical Care and Burn Unit, Saint‐Louis and Lariboisière HospitalsFHU PROMICE, DMU Parabol, APHP NordParisFrance
| | - Adriaan Voors
- Department of CardiologyUniversity Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | | | | | | | | | | | | | - Alain Cohen‐Solal
- Université Paris Cité, INSERM UMR‐S 942 (MASCOT)ParisFrance
- Department of Cardiology, APHP NordLariboisière University HospitalParisFrance
| | - Jozine M. ter Maaten
- Department of CardiologyUniversity Medical Centre Groningen, University of GroningenGroningenThe Netherlands
| | - Jan Biegus
- Institute of Heart DiseasesWroclaw Medical UniversityWroclawaPoland
| | - Piotr Ponikowski
- Institute of Heart DiseasesWroclaw Medical UniversityWroclawaPoland
| | - Gerasimos Filippatos
- National and Kapodistrian University of Athens, School of MedicineAttikon University HospitalHaidariGreece
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases ‘Prof. C.C. Iliescu’University of Medicine ‘Carol Davila’BucharestRomania
| | - Matteo Pagnesi
- Department of Cardiology, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Tabassome Simon
- IMProving Emergency Care FHUSorbonne UniversitéParisFrance
- Department of Clinical Pharmacology and Clinical Research Platform Paris‐East (URCEST‐CRC‐CRB)St Antoine Hospital, APHPParisFrance
| | - Marco Metra
- Department of Cardiology, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Douglas L. Mann
- Department of Medicine, Division of Cardiovascular, Center for Cardiovascular ResearchWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
13
|
Buch MH, Mallat Z, Dweck MR, Tarkin JM, O'Regan DP, Ferreira V, Youngstein T, Plein S. Current understanding and management of cardiovascular involvement in rheumatic immune-mediated inflammatory diseases. Nat Rev Rheumatol 2024; 20:614-634. [PMID: 39232242 DOI: 10.1038/s41584-024-01149-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 09/06/2024]
Abstract
Immune-mediated inflammatory diseases (IMIDs) are a spectrum of disorders of overlapping immunopathogenesis, with a prevalence of up to 10% in Western populations and increasing incidence in developing countries. Although targeted treatments have revolutionized the management of rheumatic IMIDs, cardiovascular involvement confers an increased risk of mortality and remains clinically under-recognized. Cardiovascular pathology is diverse across rheumatic IMIDs, ranging from premature atherosclerotic cardiovascular disease (ASCVD) to inflammatory cardiomyopathy, which comprises myocardial microvascular dysfunction, vasculitis, myocarditis and pericarditis, and heart failure. Epidemiological and clinical data imply that rheumatic IMIDs and associated cardiovascular disease share common inflammatory mechanisms. This concept is strengthened by emergent trials that indicate improved cardiovascular outcomes with immune modulators in the general population with ASCVD. However, not all disease-modifying therapies that reduce inflammation in IMIDs such as rheumatoid arthritis demonstrate equally beneficial cardiovascular effects, and the evidence base for treatment of inflammatory cardiomyopathy in patients with rheumatic IMIDs is lacking. Specific diagnostic protocols for the early detection and monitoring of cardiovascular involvement in patients with IMIDs are emerging but are in need of ongoing development. This Review summarizes current concepts on the potentially targetable inflammatory mechanisms of cardiovascular pathology in rheumatic IMIDs and discusses how these concepts can be considered for the diagnosis and management of cardiovascular involvement across rheumatic IMIDs, with an emphasis on the potential of cardiovascular imaging for risk stratification, early detection and prognostication.
Collapse
Affiliation(s)
- Maya H Buch
- Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, UK.
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Ziad Mallat
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Marc R Dweck
- Centre for Cardiovascular Science, Chancellors Building, Little France Crescent, University of Edinburgh, Edinburgh, UK
| | - Jason M Tarkin
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Declan P O'Regan
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Vanessa Ferreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Taryn Youngstein
- National Heart & Lung Institute, Imperial College London, London, UK
- Department of Rheumatology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Sven Plein
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, UK
| |
Collapse
|
14
|
Bonaventura A, Moroni F, Golino M, Del Buono MG, Vecchié A, Potere N, Abbate A. IL-1 blockade in cardiovascular disease: an appraisal of the evidence across different inflammatory paradigms. Minerva Cardiol Angiol 2024; 72:477-488. [PMID: 37705370 DOI: 10.23736/s2724-5683.23.06390-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Pre-clinical and clinical studies suggest a role for inflammation in the pathophysiology of cardiovascular (CV) diseases. The NLRP3 (NACHT, leucine-rich repeat, and pyrin domain-containing protein 3) inflammasome is activated during tissue injury and releases interleukin-1β (IL-1β). We describe three paradigms in which the NLRP3 inflammasome and IL-1β contribute to CV diseases. During acute myocardial infarction (AMI), necrotic cell debris, including IL-1α, induce NLRP3 inflammasome activation and further damage the myocardium contributing to heart failure (HF) (acute injury paradigm). In chronic HF, IL-1β is induced by persistent myocardial overload and injury, neurohumoral activation and systemic comorbidities favoring infiltration and activation of immune cells into the myocardium, microvascular inflammation, and a pro-fibrotic response (chronic inflammation paradigm). In recurrent pericarditis, an autoinflammatory response triggered by cell injury and maintained by the NLRP3 inflammasome/IL-1β axis is present (autoinflammatory disease paradigm). Anakinra, recombinant IL-1 receptor antagonist, inhibits the acute inflammatory response in patients with ST elevation myocardial infarction (STEMI) and acute HF. Canakinumab, IL-1β antibody, blunts systemic inflammation and prevents complications of atherosclerosis in stable patients with prior AMI. In chronic HF, anakinra reduces systemic inflammation and improves cardiorespiratory fitness. In recurrent pericarditis, anakinra and rilonacept, a soluble IL-1 receptor chimeric fusion protein blocking IL-1α and IL-1β, treat and prevent acute flares. In conclusion, the NLRP3 inflammasome and IL-1 contribute to the pathophysiology of CV diseases, and IL-1 blockade is beneficial with different roles in the acute injury, chronic inflammation and autoinflammatory disease paradigms. Further research is needed to guide the optimal use of IL-1 blockers in clinical practice.
Collapse
Affiliation(s)
- Aldo Bonaventura
- Division of Internal Medicine, Medical Center, Ospedale di Circolo & Fondazione Macchi, ASST Sette Laghi, Varese, Italy -
| | - Francesco Moroni
- Heart and Vascular Center, Division of Cardiology, Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Michele Golino
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
- VCU Health Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Marco G Del Buono
- Department of Cardiovascular and Thoracic Sciences, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Alessandra Vecchié
- Division of Internal Medicine, Medical Center, Ospedale di Circolo & Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Nicola Potere
- Department of Medicine and Ageing Sciences, "G. D'Annunzio" University, Chieti, Italy
| | - Antonio Abbate
- Heart and Vascular Center, Division of Cardiology, Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
15
|
Martí-Carvajal AJ, Gemmato-Valecillos MA, Monge Martín D, Dayer M, Alegría-Barrero E, De Sanctis JB, Parise Vasco JM, Riera Lizardo RJ, Nicola S, Martí-Amarista CE, Correa-Pérez A. Interleukin-receptor antagonist and tumour necrosis factor inhibitors for the primary and secondary prevention of atherosclerotic cardiovascular diseases. Cochrane Database Syst Rev 2024; 9:CD014741. [PMID: 39297531 PMCID: PMC11411914 DOI: 10.1002/14651858.cd014741.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ACVD) is worsened by chronic inflammatory diseases. Interleukin receptor antagonists (IL-RAs) and tumour necrosis factor-alpha (TNF) inhibitors have been studied to see if they can prevent cardiovascular events. OBJECTIVES The purpose of this study was to assess the clinical benefits and harms of IL-RAs and TNF inhibitors in the primary and secondary prevention of ACVD. SEARCH METHODS The Cochrane Heart Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE (including In-Process & Other Non-Indexed Citations), Ovid Embase, EBSCO CINAHL plus, and clinical trial registries for ongoing and unpublished studies were searched in February 2024. The reference lists of relevant studies, reviews, meta-analyses and health technology reports were searched to identify additional studies. No limitations on language, date of publication or study type were set. SELECTION CRITERIA RCTs that recruited people with and without pre-existing ACVD, comparing IL-RAs or TNF inhibitors versus placebo or usual care, were selected. The primary outcomes considered were all-cause mortality, myocardial infarction, unstable angina, and adverse events. DATA COLLECTION AND ANALYSIS Two or more review authors, working independently at each step, selected studies, extracted data, assessed the risk of bias and used GRADE to judge the certainty of evidence. MAIN RESULTS We included 58 RCTs (22,053 participants; 21,308 analysed), comparing medication efficacy with placebo or usual care. Thirty-four trials focused on primary prevention and 24 on secondary prevention. The interventions included IL-1 RAs (anakinra, canakinumab), IL-6 RA (tocilizumab), TNF-inhibitors (etanercept, infliximab) compared with placebo or usual care. The certainty of evidence was low to very low due to biases and imprecision; all trials had a high risk of bias. Primary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality(RR 0.33, 95% CI 0.01 to 7.58, 1 trial), myocardial infarction (RR 0.71, 95% CI 0.04 to 12.48, I² = 39%, 2 trials), unstable angina (RR 0.24, 95% CI 0.03 to 2.11, I² = 0%, 2 trials), stroke (RR 2.42, 95% CI 0.12 to 50.15; 1 trial), adverse events (RR 0.85, 95% CI 0.59 to 1.22, I² = 54%, 3 trials), or infection (rate ratio 0.84, 95% 0.55 to 1.29, I² = 0%, 4 trials). Evidence is very uncertain about whether anakinra and cankinumab may reduce heart failure (RR 0.21, 95% CI 0.05 to 0.94, I² = 0%, 3 trials). Peripheral vascular disease (PVD) was not reported as an outcome. IL-6 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.68, 95% CI 0.12 to 3.74, I² = 30%, 3 trials), myocardial infarction (RR 0.27, 95% CI 0.04 to1.68, I² = 0%, 3 trials), heart failure (RR 1.02, 95% CI 0.11 to 9.63, I² = 0%, 2 trials), PVD (RR 2.94, 95% CI 0.12 to 71.47, 1 trial), stroke (RR 0.34, 95% CI 0.01 to 8.14, 1 trial), or any infection (rate ratio 1.10, 95% CI: 0.88 to 1.37, I2 = 18%, 5 trials). Adverse events may increase (RR 1.13, 95% CI 1.04 to 1.23, I² = 33%, 5 trials). No trial assessed unstable angina. TNF inhibitors The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.78, 95% CI 0.63 to 4.99, I² = 10%, 3 trials), myocardial infarction (RR 2.61, 95% CI 0.11 to 62.26, 1 trial), stroke (RR 0.46, 95% CI 0.08 to 2.80, I² = 0%; 3 trials), heart failure (RR 0.85, 95% CI 0.06 to 12.76, 1 trial). Adverse events may increase (RR 1.13, 95% CI 1.01 to 1.25, I² = 51%, 13 trials). No trial assessed unstable angina or PVD. Secondary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.94, 95% CI 0.84 to 1.06, I² = 0%, 8 trials), unstable angina (RR 0.88, 95% CI 0.65 to 1.19, I² = 0%, 3 trials), PVD (RR 0.85, 95% CI 0.19 to 3.73, I² = 38%, 3 trials), stroke (RR 0.94, 95% CI 0.74 to 1.2, I² = 0%; 7 trials), heart failure (RR 0.91, 95% 0.5 to 1.65, I² = 0%; 7 trials), or adverse events (RR 0.92, 95% CI 0.78 to 1.09, I² = 3%, 4 trials). There may be little to no difference between the groups in myocardial infarction (RR 0.88, 95% CI 0.0.75 to 1.04, I² = 0%, 6 trials). IL6-RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.09, 95% CI 0.61 to 1.96, I² = 0%, 2 trials), myocardial infarction (RR 0.46, 95% CI 0.07 to 3.04, I² = 45%, 3 trials), unstable angina (RR 0.33, 95% CI 0.01 to 8.02, 1 trial), stroke (RR 1.03, 95% CI 0.07 to 16.25, 1 trial), adverse events (RR 0.89, 95% CI 0.76 to 1.05, I² = 0%, 2 trials), or any infection (rate ratio 0.66, 95% CI 0.32 to 1.36, I² = 0%, 4 trials). No trial assessed PVD or heart failure. TNF inhibitors The evidence is very uncertain about the effect of the intervention on all-cause mortality (RR 1.16, 95% CI 0.69 to 1.95, I² = 47%, 5 trials), heart failure (RR 0.92, 95% 0.75 to 1.14, I² = 0%, 4 trials), or adverse events (RR 1.15, 95% CI 0.84 to 1.56, I² = 32%, 2 trials). No trial assessed myocardial infarction, unstable angina, PVD or stroke. Adverse events may be underestimated and benefits inflated due to inadequate reporting. AUTHORS' CONCLUSIONS This Cochrane review assessed the benefits and harms of using interleukin-receptor antagonists and tumour necrosis factor inhibitors for primary and secondary prevention of atherosclerotic diseases compared with placebo or usual care. However, the evidence for the predetermined outcomes was deemed low or very low certainty, so there is still a need to determine whether these interventions provide clinical benefits or cause harm from this perspective. In summary, the different biases and imprecision in the included studies limit their external validity and represent a limitation to determining the effectiveness of the intervention for both primary and secondary prevention of ACVD.
Collapse
Key Words
- humans
- angina, unstable
- angina, unstable/mortality
- angina, unstable/prevention & control
- antibodies, monoclonal, humanized
- antibodies, monoclonal, humanized/administration & dosage
- antibodies, monoclonal, humanized/adverse effects
- atherosclerosis
- atherosclerosis/mortality
- atherosclerosis/prevention & control
- bias
- cause of death
- myocardial infarction
- myocardial infarction/mortality
- myocardial infarction/prevention & control
- primary prevention
- primary prevention/methods
- randomized controlled trials as topic
- receptors, interleukin-1
- receptors, interleukin-1/antagonists & inhibitors
- secondary prevention
- secondary prevention/methods
- tumor necrosis factor-alpha
- tumor necrosis factor-alpha/antagonists & inhibitors
Collapse
Affiliation(s)
- Arturo J Martí-Carvajal
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
- Facultad de Medicina (Centro Cochrane Madrid), Universidad Francisco de Vitoria, Madrid, Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia , Venezuela
| | - Mario A Gemmato-Valecillos
- Icahn School of Medicine at Mount Sinai/ NYCHH Elmhurst Hospital Center, 79-01 Broadway, Elmhurst, New York 11373, USA
| | | | - Mark Dayer
- Cardiovascular Research Institute, Mater Private Network, Dublin, Ireland
- Faculty of Health, University of Plymouth, Plymouth, UK
| | | | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Palacky University, Faculty of Medicine and Dentistry, Olomouc, Czech Republic
| | - Juan Marcos Parise Vasco
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | - Ricardo J Riera Lizardo
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
| | - Susana Nicola
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | | | - Andrea Correa-Pérez
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Hospital Pharmacy and Medical Devices Department, Hospital Central de la Defensa "Gómez Ulla" CSVE, Madrid, Spain
| |
Collapse
|
16
|
Pascual-Figal D, Núñez Villota J, Pérez-Martínez MT, González-Juanatey JR, Taibo-Urquía M, Llàcer Iborra P, González-Martín J, Villar S, Soler M, Mirabet S, Aimo A, Riquelme-Pérez A, Anguita Sánchez M, Martínez-Sellés M, Sánchez PL, Ibáñez B, Bayés-Genís A. Colchicine in acute heart failure: Rationale and design of a randomized double-blind placebo-controlled trial (COLICA). Eur J Heart Fail 2024; 26:1999-2007. [PMID: 38837516 DOI: 10.1002/ejhf.3300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/13/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
AIMS Heart failure (HF) elicits a pro-inflammatory state, which is associated with impaired clinical outcomes, but no anti-inflammatory therapies have demonstrated a clinical benefit yet. Inflammatory pathways related with the interleukin-1 axis are overactivated during episodes of acute HF. Colchicine, an anti-inflammatory drug with proven benefits in acute pericarditis and ischaemic heart disease, may target this inflammatory response. This study aims to assess the efficacy of colchicine in acute HF patients. METHODS COLICA is a multicentre, randomized, double-blind, placebo-controlled trial enrolling 278 patients across 12 sites. Patients presenting with acute HF, clinical evidence of congestion requiring ≥40 mg of intravenous furosemide and N-terminal pro-B-type natriuretic peptide (NT-proBNP) >900 pg/ml, are eligible for participation. Patients are enrolled irrespective of left ventricular ejection fraction, HF type (new-onset or not) and setting (hospital or outpatient clinic). Patients are randomized 1:1 within the first 24 h of presentation to either placebo or colchicine, with an initial loading dose of 2 mg followed by 0.5 mg every 12 h for 8 weeks (reduced dose if <70 kg, >75 years old, or glomerular filtration rate <50 ml/min/1.73 m2). The primary efficacy endpoint is the time-averaged proportional change in NT-proBNP concentrations from baseline to week 8. Key secondary and exploratory outcomes include symptoms, diuretic use, worsening HF episodes, related biomarkers of cardiac stress and inflammation, total and cardiovascular readmissions, mortality and safety events. CONCLUSION COLICA will be the first randomized trial testing the efficacy and safety of colchicine for acute HF.
Collapse
Affiliation(s)
- Domingo Pascual-Figal
- Hospital Clínico Universitario Virgen de la Arrixaca, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER cardiovascular, Madrid, Spain
| | - Julio Núñez Villota
- CIBER cardiovascular, Madrid, Spain
- Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Maria Teresa Pérez-Martínez
- Hospital Clínico Universitario Virgen de la Arrixaca, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
| | | | - Mikel Taibo-Urquía
- CIBER cardiovascular, Madrid, Spain
- IIS-Hospital Fundación Jiménez Diaz, Madrid, Spain
| | | | | | - Sandra Villar
- CIBER cardiovascular, Madrid, Spain
- Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Meritxel Soler
- CIBER cardiovascular, Madrid, Spain
- Hospital Germans-Trial i Pujol, Barcelona, Spain
| | - Sonia Mirabet
- CIBER cardiovascular, Madrid, Spain
- Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Alberto Aimo
- Fondazione Toscana Gabriele Monasterio, Health Sciences Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Alejandro Riquelme-Pérez
- Hospital Clínico Universitario Virgen de la Arrixaca, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
| | | | - Manuel Martínez-Sellés
- CIBER cardiovascular, Madrid, Spain
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Europea, Universidad Complutense, Madrid, Spain
| | - Pedro L Sánchez
- CIBER cardiovascular, Madrid, Spain
- Hospital Universitario de Salamanca, IBSAL, Salamanca, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER cardiovascular, Madrid, Spain
- IIS-Hospital Fundación Jiménez Diaz, Madrid, Spain
| | - Antoni Bayés-Genís
- CIBER cardiovascular, Madrid, Spain
- Hospital Germans-Trial i Pujol, Barcelona, Spain
| |
Collapse
|
17
|
Tada A, Doi S, Harada T, Ibe T, Naser JA, Amdahl M, Reddy YNV, Borlaug BA. Autoimmune Disorders in Heart Failure With Preserved Ejection Fraction. JACC. HEART FAILURE 2024; 12:1257-1269. [PMID: 38819353 DOI: 10.1016/j.jchf.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Inflammation plays a fundamental role in the pathogenesis of heart failure with preserved ejection fraction (HFpEF). In most patients, inflammation develops secondary to cardiometabolic comorbidities, but in some, HFpEF develops in the setting of an underlying systemic inflammatory disease such as rheumatoid arthritis or systemic lupus erythematosus. OBJECTIVES This study aimed to investigate the prevalence, pathophysiology, and outcome of patients with HFpEF and autoimmune or primary inflammatory disorders. METHODS Of 982 consecutively evaluated patients with HFpEF diagnosed, 79 (8.0%) had autoimmune disorders. HFpEF was defined by invasive cardiopulmonary hemodynamic exercise testing. RESULTS Female sex, higher heart rate, lower hemoglobin, absence of atrial fibrillation, and absence of coronary artery disease were independently associated with autoimmune disorders. Hemodynamics at rest and exercise did not differ between the groups, but peripheral oxygen extraction was lower in those with autoimmune disorders, reflected by lower arterial-venous oxygen content difference at rest (4.2 ± 0.7 mL/dL vs 4.6 ± 1.0 mL/dL; P < 0.001) and during exercise (9.3 ± 2.2 mL/dL vs 10.4 ± 2.2 mL/dL; P < 0.001), suggesting a greater peripheral deficit, and ventilatory efficiency (VE/VCo2 slope, regression slope relating minute ventilation to carbon dioxide output) was also more impaired (38.0 ± 7.9 vs 36.2 ± 7.3; P = 0.043). Patients with autoimmune disorders had a higher risk of death or heart failure (HF) hospitalization compared with those without in adjusted analyses (HR: 1.95 [95% CI: 1.17-3.27]; P = 0.011) over a median follow-up of 3.0 years, which was primarily attributable to higher risk of HF hospitalization (HR: 2.87 [95% CI: 1.09-7.57]; P = 0.033). CONCLUSIONS Patients with HFpEF and autoimmune disorders have similar hemodynamic derangements but greater peripheral deficits in oxygen transport and higher risk for adverse outcome compared with those without.
Collapse
Affiliation(s)
- Atsushi Tada
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Shunichi Doi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Tomonari Harada
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Tatsuro Ibe
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jwan A Naser
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew Amdahl
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Yogesh N V Reddy
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
18
|
Carbone S, Abbate A. Inflammation and Cardiorespiratory Fitness in HFpEF: A Decade of Progress. JACC. HEART FAILURE 2024; 12:1270-1273. [PMID: 38960521 DOI: 10.1016/j.jchf.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 07/05/2024]
Affiliation(s)
- Salvatore Carbone
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia, USA; Virginia Commonwealth University Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA.
| | - Antonio Abbate
- Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
19
|
Jafree E, Del Buono MG, Canada JM, Carbone S, Kron J, Arena R, Van Tassell B, Abbate A, Trankle CR. Interleukin-1 Inhibition for the Prevention and Treatment of Heart Failure. J Cardiovasc Pharmacol 2024; 83:522-530. [PMID: 37815298 PMCID: PMC11004086 DOI: 10.1097/fjc.0000000000001497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023]
Abstract
ABSTRACT Heart failure (HF) is a complex syndrome that remains a leading cause of morbidity and mortality worldwide. Abundant evidence suggests inflammation plays a key role in the development and perpetuation of HF, but there are currently no anti-inflammatory treatments approved for use in HF. Interleukin-1, the prototypical proinflammatory cytokine, has been implicated in adverse cardiac remodeling and left ventricular dysfunction. Multiple early phase clinical trials using interleukin-1 blockade in patients at risk for or diagnosed with HF have suggested favorable safety and efficacy in reducing inflammatory biomarkers, as well as positive signals in surrogate and clinical end points. Additional large scale clinical trials are urgently needed to confirm the safety and efficacy of this therapeutic approach specifically in HF. In this narrative review, we discuss current evidence regarding interleukin-1 blockade in the prevention and treatment of HF.
Collapse
Affiliation(s)
- Ehsan Jafree
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Justin M. Canada
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| | - Salvatore Carbone
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA
| | - Jordana Kron
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| | - Ross Arena
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Benjamin Van Tassell
- Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- Division of Cardiology, University of Virginia, Charlottesville, VA
| | - Cory R. Trankle
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
20
|
Hegner P, Wester M, Tafelmeier M, Provaznik Z, Klatt S, Schmid C, Maier LS, Arzt M, Wagner S, Lebek S. Systemic inflammation predicts diastolic dysfunction in patients with sleep disordered breathing. Eur Respir J 2024; 63:2400579. [PMID: 38636989 PMCID: PMC11154749 DOI: 10.1183/13993003.00579-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) constitutes approximately half of all patients with heart failure and causes mortality similar to heart failure with reduced ejection fraction [1]. HFpEF is highly relevant as novel evidence-based therapies emerge but treatment options remain limited [1]. Diastolic dysfunction is a hallmark of HFpEF and is also very common in up to 80% of high-risk cardiovascular patients undergoing cardiac surgery [2]. Even without overt HFpEF, echocardiographic diastolic dysfunction is independently associated with increased mortality [3]. Another important characteristic of HFpEF is the frequent presence of comorbidities, with one of the most important being sleep disordered breathing (SDB). SDB affects over one billion patients in the general population and is highly prevalent in cardiovascular high-risk patients, which underscores its high socioeconomic relevance [4]. Interestingly, SDB patients frequently exhibit diastolic dysfunction [5]; however, the underlying mechanisms remain elusive thus far [6]. In this cross-sectional experimental study, we analysed the role of inflammation and fibrosis for diastolic dysfunction in cardiovascular high-risk patients stratified by the prevalence of SDB, which may provide a groundwork for future therapeutic strategies. Systemic and myocardial inflammation are associated with diastolic cardiac dysfunction in patients with sleep disordered breathing, which may have therapeutic implications https://bit.ly/3vxAECC
Collapse
Affiliation(s)
- Philipp Hegner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Wester
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Maria Tafelmeier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Zdenek Provaznik
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Susanne Klatt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Christof Schmid
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lars Siegfried Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
- S. Wagner and S. Lebek contributed equally to this article as senior authors
| | - Simon Lebek
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- S. Wagner and S. Lebek contributed equally to this article as senior authors
| |
Collapse
|
21
|
Ovchinnikov A, Potekhina A, Arefieva T, Filatova A, Ageev F, Belyavskiy E. Use of Statins in Heart Failure with Preserved Ejection Fraction: Current Evidence and Perspectives. Int J Mol Sci 2024; 25:4958. [PMID: 38732177 PMCID: PMC11084261 DOI: 10.3390/ijms25094958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Systemic inflammation and coronary microvascular endothelial dysfunction are essential pathophysiological factors in heart failure (HF) with preserved ejection fraction (HFpEF) that support the use of statins. The pleiotropic properties of statins, such as anti-inflammatory, antihypertrophic, antifibrotic, and antioxidant effects, are generally accepted and may be beneficial in HF, especially in HFpEF. Numerous observational clinical trials have consistently shown a beneficial prognostic effect of statins in patients with HFpEF, while the results of two larger trials in patients with HFrEF have been controversial. Such differences may be related to a more pronounced impact of the pleiotropic properties of statins on the pathophysiology of HFpEF and pro-inflammatory comorbidities (arterial hypertension, diabetes mellitus, obesity, chronic kidney disease) that are more common in HFpEF. This review discusses the potential mechanisms of statin action that may be beneficial for patients with HFpEF, as well as clinical trials that have evaluated the statin effects on left ventricular diastolic function and clinical outcomes in patients with HFpEF.
Collapse
Affiliation(s)
- Artem Ovchinnikov
- Laboratory of Myocardial Fibrosis and Heart Failure with Preserved Ejection Fraction, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (A.P.); (A.F.)
- Department of Clinical Functional Diagnostics, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, 127473 Moscow, Russia
| | - Alexandra Potekhina
- Laboratory of Myocardial Fibrosis and Heart Failure with Preserved Ejection Fraction, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (A.P.); (A.F.)
| | - Tatiana Arefieva
- Laboratory of Cell Immunology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia;
- Faculty of Basic Medicine, Lomonosov Moscow State University, Leninskie Gory, 1, 119991 Moscow, Russia
| | - Anastasiia Filatova
- Laboratory of Myocardial Fibrosis and Heart Failure with Preserved Ejection Fraction, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (A.P.); (A.F.)
- Laboratory of Cell Immunology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia;
| | - Fail Ageev
- Out-Patient Department, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia;
| | - Evgeny Belyavskiy
- Medizinisches Versorgungszentrum des Deutsches Herzzentrum der Charite, Augustenburger Platz 1, 13353 Berlin, Germany;
| |
Collapse
|
22
|
Boulet J, Sridhar VS, Bouabdallaoui N, Tardif JC, White M. Inflammation in heart failure: pathophysiology and therapeutic strategies. Inflamm Res 2024; 73:709-723. [PMID: 38546848 PMCID: PMC11058911 DOI: 10.1007/s00011-023-01845-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/09/2023] [Accepted: 12/19/2023] [Indexed: 04/30/2024] Open
Abstract
A role for inflammation in the development and progression of heart failure (HF) has been proposed for decades. Multiple studies have demonstrated the potential involvement of several groups of cytokines and chemokines in acute and chronic HF, though targeting these pathways in early therapeutic trials have produced mixed results. These studies served to highlight the complexity and nuances of how pro-inflammatory pathways contribute to the pathogenesis of HF. More recent investigations have highlighted how inflammation may play distinct roles based on HF syndrome phenotypes, findings that may guide the development of novel therapies. In this review, we propose a contemporary update on the role of inflammation mediated by the innate and adaptive immune systems with HF, highlighting differences that exist across the ejection fraction spectrum. This will specifically be looked at through the lens of established and novel biomarkers of inflammation. Subsequently, we review how improvements in inflammatory pathways may mediate clinical benefits of existing guideline-directed medical therapies for HF, as well as future therapies in the pipeline targeting HF and inflammation.
Collapse
Affiliation(s)
- Jacinthe Boulet
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Vikas S Sridhar
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada
| | - Nadia Bouabdallaoui
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Jean-Claude Tardif
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Michel White
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada.
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, QC, H1C 1C8, Montreal, Canada.
| |
Collapse
|
23
|
Toldo S, Abbate A. The role of the NLRP3 inflammasome and pyroptosis in cardiovascular diseases. Nat Rev Cardiol 2024; 21:219-237. [PMID: 37923829 PMCID: PMC11550901 DOI: 10.1038/s41569-023-00946-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2023] [Indexed: 11/06/2023]
Abstract
An intense, stereotyped inflammatory response occurs in response to ischaemic and non-ischaemic injury to the myocardium. The NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome is a finely regulated macromolecular protein complex that senses the injury and triggers and amplifies the inflammatory response by activation of caspase 1; cleavage of pro-inflammatory cytokines, such as pro-IL-1β and pro-IL-18, to their mature forms; and induction of inflammatory cell death (pyroptosis). Inhibitors of the NLRP3 inflammasome and blockers of IL-1β and IL-18 activity have been shown to reduce injury to the myocardium and pericardium, favour resolution of the inflammation and preserve cardiac function. In this Review, we discuss the components of the NLRP3 inflammasome and how it is formed and activated in various ischaemic and non-ischaemic cardiac pathologies (acute myocardial infarction, cardiac dysfunction and remodelling, atherothrombosis, myocarditis and pericarditis, cardiotoxicity and cardiac sarcoidosis). We also summarize current preclinical and clinical evidence from studies of agents that target the NLRP3 inflammasome and related cytokines.
Collapse
Affiliation(s)
- Stefano Toldo
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
24
|
Chisavu L, Ivan VM, Mihaescu A, Chisavu F, Schiller O, Marc L, Bob F, Schiller A. Novel Biomarkers in Evaluating Cardiac Function in Patients on Hemodialysis-A Pilot Prospective Observational Cohort Study. Diagnostics (Basel) 2024; 14:664. [PMID: 38535084 PMCID: PMC10969652 DOI: 10.3390/diagnostics14060664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 01/03/2025] Open
Abstract
Chronic kidney disease patients treated by hemodialysis present a high cardiovascular morbidity and mortality. There is an imperative need for novel biomarkers for identifying these patients and to offer possible therapeutically interventions. We performed a prospective observational cohort study on 77 patients in the period of October 2021-October 2023. We measured serum plasma levels of interleukin 1-beta, galectin 3, human suppression of tumorigenicity factor 2, bone morphogenetic protein 2 and fibroblastic growth factor 23 at the inclusion site. We evaluated the correlations of these biomarkers with cardiac function and structure evaluated by echocardiography. The mean age was 61.02 (±11.81) years, with 45 (56.2%) males and with a dialysis vintage of 4.95 (2.4-7.8) years. Median ejection fraction was 51 (43-54%), and more than two-thirds of the patients presented valvular calcifications. Overall mortality was 22%. Interleukin 1-beta was correlated positively with ejection fraction and global longitudinal strain and negatively with left atrium diameter and left ventricle telesystolic diameter. Galectin 3 values were negatively correlated with aortic valve fibrosis and mitral valve calcifications, and human suppression tumorigenicity factor 2 was negatively correlated with mitral valve calcifications. Some of these novel biomarkers could be used to better assess cardiovascular disease in patients on maintenance hemodialysis.
Collapse
Affiliation(s)
- Lazar Chisavu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Nephrology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Viviana Mihaela Ivan
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Cardiology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Adelina Mihaescu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Nephrology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Flavia Chisavu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Louis Turcanu’ Emergency County Hospital for Children, 300011 Timisoara, Romania
| | | | - Luciana Marc
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Nephrology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Flaviu Bob
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Nephrology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Adalbert Schiller
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Nephrology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
25
|
Verma R, Dhingra NK, Connelly KA. Obesity/cardiometabolic phenotype of heart failure with preserved ejection fraction: mechanisms to recent trials. Curr Opin Cardiol 2024; 39:92-97. [PMID: 38294186 DOI: 10.1097/hco.0000000000001113] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
PURPOSE OF REVIEW Heart failure with preserved ejection fraction (HFpEF) is a leading and growing cause of morbidity and mortality globally. Of the various phenotypes identified, the obesity (or cardiometabolic) phenotype appears to be most common. The purpose of this review is to provide the clinician with an abridged understanding of recent developments that have elucidated obesity/visceral adiposity as a central mechanism linking inflammation/immune dysregulation to the development of the HFpEF syndrome. Recent clinical trials examining the efficacy of pharmacological treatments that target obesity in HFpEF will also be discussed. RECENT FINDINGS Recent data indicate that visceral adiposity and insulin resistance in HFpEF serve as key mechanisms driving inflammation and immune dysregulation, which play a critical role in the development of cardiac stiffness, diastolic dysfunction and fibrosis in HFpEF. In obesity, alterations in macrophage polarization, changes in innate and adaptive immune systems and altered myocardial energetics promote metabolic inflammation in HFpEF. Finally, emerging data suggest that inflammatory biomarkers, specifically, IL-6, may provide useful information about HFpEF severity and symptom burden in obesity. SUMMARY The obesity phenotype of HFpEF is seen in upward of 80% with HFpEF. Obesity is not just a bystander, but plays an essential role in the pathobiology and clinical course of HFpEF. Targeting overweight/obesity in HFpEF with GLP-1 receptor agonists holds promise in these patients.
Collapse
Affiliation(s)
- Raj Verma
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Kim A Connelly
- Division of Cardiology, St Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Kong Y, Wang X, Qie R. Immunotherapy-associated cardiovascular toxicities: insights from preclinical and clinical studies. Front Oncol 2024; 14:1347140. [PMID: 38482205 PMCID: PMC10932998 DOI: 10.3389/fonc.2024.1347140] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/12/2024] [Indexed: 11/02/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have become a widely accepted and effective treatment for various types of solid tumors. Recent studies suggest that cardiovascular immune-related adverse events (irAEs) specifically have an incidence rate ranging from 1.14% to more than 5%. Myocarditis is the most common observed cardiovascular irAE. Others include arrhythmias, pericardial diseases, vasculitis, and a condition resembling takotsubo cardiomyopathy. Programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) pathway, cytotoxic T-lymphocyte antigen-4 (CTLA-4) pathway, and the recently discovered lymphocyte-activation gene 3 (LAG-3) pathway, play a critical role in boosting the body's natural immune response against cancer cells. While ICIs offer significant benefits in terms of augmenting immune function, they can also give rise to unwanted inflammatory side effects known as irAEs. The occurrence of irAEs can vary in severity, ranging from mild to severe, and can impact the overall clinical efficacy of these agents. This review aims to summarize the underlying mechanisms of cardiovascular irAE from both preclinical and clinical studies for a better understanding of cardiovascular irAE in clinical application.
Collapse
Affiliation(s)
- Youqian Kong
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoyu Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Rui Qie
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
27
|
Lazzerini PE, Cupelli M, Cartocci A, Bertolozzi I, Salvini V, Accioli R, Salvadori F, Marzotti T, Verrengia D, Cevenini G, Bisogno S, Bicchi M, Donati G, Bernardini S, Laghi‐Pasini F, Acampa M, Capecchi PL, El‐Sherif N, Boutjdir M. Elevated Interleukin-6 Levels Are Associated With an Increased Risk of QTc Interval Prolongation in a Large Cohort of US Veterans. J Am Heart Assoc 2024; 13:e032071. [PMID: 38348789 PMCID: PMC11010073 DOI: 10.1161/jaha.123.032071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/13/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND Although accumulating data indicate that IL-6 (interleukin-6) can promote heart rate-corrected QT interval (QTc) prolongation via direct and indirect effects on cardiac electrophysiology, current evidence comes from basic investigations and small clinical studies only. Therefore, IL-6 is still largely ignored in the clinical management of long-QT syndrome and related arrhythmias. The aim of this study was to estimate the risk of QTc prolongation associated with elevated IL-6 levels in a large population of unselected subjects. METHODS AND RESULTS An observational study using the Veterans Affairs Informatics and Computing Infrastructure was performed. Participants were US veterans who had an ECG and were tested for IL-6. Descriptive statistics and univariate and multivariate regression analyses were performed to study the relationship between IL-6 and QTc prolongation risk. Study population comprised 1085 individuals, 306 showing normal (<5 pg/mL), 376 moderately high (5-25 pg/mL), and 403 high (>25 pg/mL) IL-6 levels. Subjects with elevated IL-6 showed a concentration-dependent increase in the prevalence of QTc prolongation, and those presenting with QTc prolongation exhibited higher circulating IL-6 levels. Stepwise multivariate regression analyses demonstrated that increased IL-6 level was significantly associated with a risk of QTc prolongation up to 2 times the odds of the reference category of QTc (e.g. QTc >470 ms men/480 ms women ms: odds ratio, 2.28 [95% CI, 1.12-4.50] for IL-6 >25 pg/mL) regardless of the underlying cause. Specifically, the mean QTc increase observed in the presence of elevated IL-6 was quantitatively comparable (IL-6 >25 pg/mL:+6.7 ms) to that of major recognized QT-prolonging risk factors, such as hypokalemia and history of myocardial infarction. CONCLUSIONS Our data provide evidence that a high circulating IL-6 level is a robust risk factor for QTc prolongation in a large cohort of US veterans, supporting a potentially important arrhythmogenic role for this cytokine in the general population.
Collapse
Affiliation(s)
| | - Michael Cupelli
- VA New York Harbor Healthcare SystemNew YorkNYUSA
- SUNY Downstate Health Sciences UniversityNew YorkNYUSA
| | | | - Iacopo Bertolozzi
- Cardiology Intensive Therapy Unit, Department of Internal MedicineNuovo Ospedale San Giovanni di Dio (former Cardiology Intensive Therapy Unit, Department of Internal Medicine, Hospital of Carrara, Carrara, Italy)FlorenceItaly
| | - Viola Salvini
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | - Riccardo Accioli
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | - Fabio Salvadori
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | - Tommaso Marzotti
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | - Decoroso Verrengia
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | | | - Stefania Bisogno
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | - Maurizio Bicchi
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | - Giovanni Donati
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | - Sciaila Bernardini
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | - Franco Laghi‐Pasini
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | - Maurizio Acampa
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | | | - Nabil El‐Sherif
- VA New York Harbor Healthcare SystemNew YorkNYUSA
- SUNY Downstate Health Sciences UniversityNew YorkNYUSA
| | - Mohamed Boutjdir
- VA New York Harbor Healthcare SystemNew YorkNYUSA
- SUNY Downstate Health Sciences UniversityNew YorkNYUSA
- NYU Grossman School of MedicineNew YorkNYUSA
| |
Collapse
|
28
|
Del Buono MG, Bonaventura A, Vecchié A, Moroni F, Golino M, Bressi E, De Ponti R, Dentali F, Montone RA, Kron J, Lazzerini PE, Crea F, Abbate A. Pathogenic pathways and therapeutic targets of inflammation in heart diseases: A focus on Interleukin-1. Eur J Clin Invest 2024; 54:e14110. [PMID: 37837616 DOI: 10.1111/eci.14110] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/26/2023] [Accepted: 10/06/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND An exuberant and dysregulated inflammatory response contributes to the development and progression of cardiovascular diseases (CVDs). METHODS This narrative review includes original articles and reviews published over the past 20 years and found through PubMed. The following search terms (or combination of terms) were considered: "acute pericarditis," "recurrent pericarditis," "myocarditis," "cardiac sarcoidosis," "atherosclerosis," "acute myocardial infarction," "inflammation," "NLRP3 inflammasome," "Interleukin-1" and "treatment." RESULTS Recent evidence supports the role of inflammation across a wide spectrum of CVDs including myocarditis, pericarditis, inflammatory cardiomyopathies (i.e. cardiac sarcoidosis) as well as atherosclerotic CVD and heart failure. Interleukins (ILs) are the signalling mediators of the inflammatory response. The NACHT, leucine-rich repeat and pyrin-domain containing protein 3 (NLRP3) inflammasome play a key role in producing IL-1β, the prototypical pro-inflammatory cytokine involved in CVDs. Other pro-inflammatory cytokines (e.g. tumour necrosis factor) have been implicated in cardiac sarcoidosis. As a proof of this, IL-1 blockade has been proven efficacious in pericarditis and chronic coronary syndrome. CONCLUSION Tailored strategies aiming at quenching the inflammatory response have emerged as promising to treat CVDs. In this review article, we summarize recent evidence regarding the role of inflammation across a broad spectrum of CVDs. We also review novel evidence regarding targeted therapeutic strategies.
Collapse
Affiliation(s)
- Marco Giuseppe Del Buono
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Aldo Bonaventura
- Department of Internal Medicine, Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Alessandra Vecchié
- Department of Internal Medicine, Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Francesco Moroni
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Michele Golino
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Edoardo Bressi
- Department of Cardiology, Policlinico Casilino, Rome, Italy
| | - Roberto De Ponti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Francesco Dentali
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Rocco Antonio Montone
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Jordana Kron
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
29
|
Markousis-Mavrogenis G, Baumhove L, Al-Mubarak AA, Aboumsallem JP, Bomer N, Voors AA, van der Meer P. Immunomodulation and immunopharmacology in heart failure. Nat Rev Cardiol 2024; 21:119-149. [PMID: 37709934 DOI: 10.1038/s41569-023-00919-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 09/16/2023]
Abstract
The immune system is intimately involved in the pathophysiology of heart failure. However, it is currently underused as a therapeutic target in the clinical setting. Moreover, the development of novel immunomodulatory therapies and their investigation for the treatment of patients with heart failure are hampered by the fact that currently used, evidence-based treatments for heart failure exert multiple immunomodulatory effects. In this Review, we discuss current knowledge on how evidence-based treatments for heart failure affect the immune system in addition to their primary mechanism of action, both to inform practising physicians about these pleiotropic actions and to create a framework for the development and application of future immunomodulatory therapies. We also delineate which subpopulations of patients with heart failure might benefit from immunomodulatory treatments. Furthermore, we summarize completed and ongoing clinical trials that assess immunomodulatory treatments in heart failure and present several therapeutic targets that could be investigated in the future. Lastly, we provide future directions to leverage the immunomodulatory potential of existing treatments and to foster the investigation of novel immunomodulatory therapeutics.
Collapse
Affiliation(s)
- George Markousis-Mavrogenis
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lukas Baumhove
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ali A Al-Mubarak
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Cardiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
30
|
Zhang H, Dhalla NS. The Role of Pro-Inflammatory Cytokines in the Pathogenesis of Cardiovascular Disease. Int J Mol Sci 2024; 25:1082. [PMID: 38256155 PMCID: PMC10817020 DOI: 10.3390/ijms25021082] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
With cardiovascular disease (CVD) being a primary source of global morbidity and mortality, it is crucial that we understand the molecular pathophysiological mechanisms at play. Recently, numerous pro-inflammatory cytokines have been linked to several different CVDs, which are now often considered an adversely pro-inflammatory state. These cytokines most notably include interleukin-6 (IL-6),tumor necrosis factor (TNF)α, and the interleukin-1 (IL-1) family, amongst others. Not only does inflammation have intricate and complex interactions with pathophysiological processes such as oxidative stress and calcium mishandling, but it also plays a role in the balance between tissue repair and destruction. In this regard, pre-clinical and clinical evidence has clearly demonstrated the involvement and dynamic nature of pro-inflammatory cytokines in many heart conditions; however, the clinical utility of the findings so far remains unclear. Whether these cytokines can serve as markers or risk predictors of disease states or act as potential therapeutic targets, further extensive research is needed to fully understand the complex network of interactions that these molecules encompass in the context of heart disease. This review will highlight the significant advances in our understanding of the contributions of pro-inflammatory cytokines in CVDs, including ischemic heart disease (atherosclerosis, thrombosis, acute myocardial infarction, and ischemia-reperfusion injury), cardiac remodeling (hypertension, cardiac hypertrophy, cardiac fibrosis, cardiac apoptosis, and heart failure), different cardiomyopathies as well as ventricular arrhythmias and atrial fibrillation. In addition, this article is focused on discussing the shortcomings in both pathological and therapeutic aspects of pro-inflammatory cytokines in CVD that still need to be addressed by future studies.
Collapse
Affiliation(s)
- Hannah Zhang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
31
|
Gremese E, Tolusso B, Bruno D, Paglionico AM, Perniola S, Ferraccioli G, Alivernini S. COVID-19 illness: Different comorbidities may require different immunological therapeutic targets. Eur J Clin Invest 2023; 53:e14096. [PMID: 37724937 DOI: 10.1111/eci.14096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/02/2023] [Accepted: 07/26/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND The SARS-CoV-2 pandemic has led to more than 6,870.000 deaths worldwide. Despite recent therapeutic advances, deaths in Intensive Care Units still range between 34 and 72%, comprising substantial unmet need as we move to an endemic phase. The general agreement is that in the first few days of infection, antiviral drugs and neutralizing monoclonal antibodies should be adopted. When the patient is hospitalized and develops severe pneumonia, progressing to a systemic disease, immune modifying therapy with corticosteroids is indicated. Such interventions, however, are less effective in the context of comorbidities (e.g., diabetes, hypertension, heart failure, atrial fibrillation, obesity and central nervous system-CNS diseases) which are by themselves associated with poor outcomes. Such comorbidities comprise common and some distinct underlying inflammatory pathobiology regulated by differential cytokine taxonomy. METHODS Searching in the PubMed database, literature pertaining to the biology underlying the different comorbidities, and the data from the studies related to various immunological treatments for the Covid-19 disease were carefully analyzed. RESULTS Several experimental and clinical data have demonstrated that hypertension and atrial fibrillation present an IL-6 dependent signature, whereas diabetes, obesity, heart failure and CNS diseases may exhibit an IL-1a/b predominant signature. Distinct selective cytokine targeting may offer advantage in treating severe COVID-19 illness based on single or multiple associated comorbidities. When the patient does not immediately respond, a broader target range through JAKs pathway inhibitors may be indicated. CONCLUSIONS Herein, we discuss the biological background associated with distinct comorbidities which might impact the SARS-CoV-2 infection course and how these should to be addressed to improve the current therapeutic outcome.
Collapse
Affiliation(s)
- Elisa Gremese
- Clinical Immunology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
- Immunology Core Facility, GSTEP, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Barbara Tolusso
- Immunology Core Facility, GSTEP, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Dario Bruno
- Clinical Immunology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Anna Maria Paglionico
- Clinical Immunology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Simone Perniola
- Clinical Immunology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | | | - Stefano Alivernini
- Catholic University of the Sacred Heart, Rome, Italy
- Immunology Core Facility, GSTEP, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Rheumatology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| |
Collapse
|
32
|
Zhao M, Zheng Z, Yin Z, Zhang J, Peng S, Liu J, Pan W, Wei C, Xu Y, Qin JJ, Wan J, Wang M. DEL-1 deficiency aggravates pressure overload-induced heart failure by promoting neutrophil infiltration and neutrophil extracellular traps formation. Biochem Pharmacol 2023; 218:115912. [PMID: 37956894 DOI: 10.1016/j.bcp.2023.115912] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023]
Abstract
Recent studies have shown that neutrophils play an important role in the development and progression of heart failure. Developmental endothelial locus-1 (DEL-1) is an anti-inflammatory glycoprotein that has been found to have protective effects in various cardiovascular diseases. However, the role of DEL-1 in chronic heart failure is not well understood. In a mouse model of pressure overload-induced non-ischemic cardiac failure, we found that neutrophil infiltration in the heart increased and DEL-1 levels decreased in the early stages of heart failure. DEL-1 deficiency worsened pressure overload-induced cardiac dysfunction and remodeling in mice. Mechanistically, DEL-1 deficiency promotes neutrophil infiltration and the formation of neutrophil extracellular traps (NETs) through the regulation of P38 signaling. In vitro experiments showed that DEL-1 can inhibit P38 signaling and NETs formation in mouse neutrophils in a MAC-1-dependent manner. Depleting neutrophils, inhibiting NETs formation, and inhibiting P38 signaling all reduced the exacerbation of heart failure caused by DEL-1 deletion. Overall, our findings suggest that DEL-1 deficiency worsens pressure overload-induced heart failure by promoting neutrophil infiltration and NETs formation.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| |
Collapse
|
33
|
Cheng X, Zhao H, Wen X, Li G, Guo S, Zhang D. NLRP3-inflammasome inhibition by MCC950 attenuates cardiac and pulmonary artery remodelling in heart failure with preserved ejection fraction. Life Sci 2023; 333:122185. [PMID: 37858713 DOI: 10.1016/j.lfs.2023.122185] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/03/2023] [Accepted: 10/15/2023] [Indexed: 10/21/2023]
Abstract
AIMS The purpose of this study was to evaluate the role of the NLRP3-inflammasome in heart failure with preserved ejection fraction (HFpEF). MAIN METHODS Serum inflammatory cytokines were detected in patients with heart failure. Correlation analysis was performed to investigate the relationship between serum inflammatory cytokines and left ventricular diastolic function. A 'two-hit' (metabolic stress and mechanical stress) mouse model of HFpEF was established. Furthermore, MCC950 was used to determine the role of NLRP3-inflammasome inhibition in cardiac and pulmonary artery remodelling in HFpEF mice. KEY FINDINGS Compared with heart failure patients with reduced ejection fraction, patients with HFpEF have significantly elevated serum inflammatory cytokine levels. Serum NLRP3 and interleukin-1β levels were positively correlated with the diastolic function of HFpEF. In the HFpEF mouse model, the inhibition of the NLRP3-inflammasome by MCC950 improved exercise intolerance, glucose intolerance, and left ventricular diastolic function, but had no significant effect on systolic function. Meanwhile, MCC950 attenuated the release of inflammatory cytokines, cardiomyocyte hypertrophy and cardiac fibrosis. Mechanistically, the potential protective effects of MCC950 are achieved by inhibiting activation of the NLRP3-IL-1β pathway and cascade expansion of downstream inflammatory cytokines. Additionally, the inhibition of NLRP3-inflammasome by MCC950 reduced pulmonary artery pressure and improved pulmonary artery remodelling in HFpEF. SIGNIFICANCE The NLRP3-inflammasome plays a considerable role in inflammation and cardiac and pulmonary artery remodelling in HFpEF by activating the cascade reaction of inflammatory cytokines. This study is the first to comprehensively elucidate the role of the NLRP3-inflammasome in HFpEF, and will provide reference for future study.
Collapse
Affiliation(s)
- Xiaocheng Cheng
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Hanru Zhao
- Department of Cardiology, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, China
| | - Xuesong Wen
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Guoxing Li
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shuliang Guo
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Dongying Zhang
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
34
|
Boulmpou A, Boutou AK, Pella E, Sarafidis P, Papadopoulos CE, Vassilikos V. Cardiopulmonary Exercise Testing in Heart Failure With Preserved Ejection Fraction: Technique Principles, Current Evidence, and Future Perspectives. Cardiol Rev 2023; 31:299-317. [PMID: 36723460 DOI: 10.1097/crd.0000000000000454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a multifactorial clinical syndrome involving a rather complex pathophysiologic substrate and quite a challenging diagnosis. Exercise intolerance is a major feature of HFpEF, and in many cases, diagnosis is suspected in subjects presenting with exertional dyspnea. Cardiopulmonary exercise testing (CPET) is a noninvasive, dynamic technique that provides an integrative evaluation of cardiovascular, pulmonary, hematopoietic, neuropsychological, and metabolic functions during maximal or submaximal exercise. The assessment is based on the principle that system failure typically occurs when the system is under stress, and thus, CPET is currently considered to be the gold standard for identifying exercise intolerance, allowing the differential diagnosis of underlying causes. CPET is used in observational studies and clinical trials in HFpEF; however, in most cases, only a few from a wide variety of CPET parameters are examined, while the technique is largely underused in everyday cardiology practice. This article discusses the basic principles and methodology of CPET and studies that utilized CPET in patients with HFpEF, in an effort to increase awareness of CPET capabilities among practicing cardiologists.
Collapse
Affiliation(s)
- Aristi Boulmpou
- From the Third Department of Cardiology, Ippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Afroditi K Boutou
- Department of Respiratory Medicine, G. Papanikolaou Hospital, Thessaloniki, Greece
| | - Eva Pella
- Department of Nephrology, Ippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Pantelis Sarafidis
- Department of Nephrology, Ippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christodoulos E Papadopoulos
- From the Third Department of Cardiology, Ippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vassilios Vassilikos
- From the Third Department of Cardiology, Ippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
35
|
Alogna A, Koepp KE, Sabbah M, Espindola Netto JM, Jensen MD, Kirkland JL, Lam CSP, Obokata M, Petrie MC, Ridker PM, Sorimachi H, Tchkonia T, Voors A, Redfield MM, Borlaug BA. Interleukin-6 in Patients With Heart Failure and Preserved Ejection Fraction. JACC. HEART FAILURE 2023; 11:1549-1561. [PMID: 37565977 PMCID: PMC10895473 DOI: 10.1016/j.jchf.2023.06.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Interleukin (IL)-6 is a central inflammatory mediator and potential therapeutic target in heart failure (HF). Prior studies have shown that IL-6 concentrations are elevated in patients with HF, but much fewer data are available in heart failure with preserved ejection fraction (HFpEF). OBJECTIVES This study aims to determine how IL-6 relates to changes in cardiac function, congestion, body composition, and exercise tolerance in HFpEF. METHODS Clinical, laboratory, body composition, exercise capacity, physiologic and health status data across 4 National Heart, Lung, and Blood Institute-sponsored trials were analyzed according to the tertiles of IL-6. RESULTS IL-6 was measured in 374 patients with HFpEF. Patients with highest IL-6 levels had greater body mass index; higher N-terminal pro-B-type natriuretic peptide, C-reactive protein, and tumor necrosis factor-α levels; worse renal function; and lower hemoglobin levels, and were more likely to have diabetes. Although cardiac structure and function measured at rest were similar, patients with HFpEF and highest IL-6 concentrations had more severely impaired peak oxygen consumption (12.3 ± 3.3 mL/kg/min 13.1 ± 3.1 mL/kg/min 14.4 ± 3.9 mL/kg/min, P < 0.0001) as well as 6-minute walk distance (276 ± 107 m vs 332 ± 106 m vs 352 ± 116 m, P < 0.0001), even after accounting for increases in IL-6 related to excess body mass. IL-6 concentrations were associated with increases in total body fat and trunk fat, more severe symptoms during submaximal exercise, and poorer patient-reported health status. CONCLUSIONS IL-6 levels are commonly elevated in HFpEF, and are associated with greater symptom severity, poorer exercise capacity, and more upper body fat accumulation. These findings support testing the hypothesis that therapies that inhibit IL-6 in patients with HFpEF may improve clinical status. (Clinical Trial Registrations: Phosphodiesterase-5 Inhibition to Improve Clinical Status and Exercise Capacity in Diastolic Heart Failure [RELAX], NCT00763867; Nitrate's Effect on Activity Tolerance in Heart Failure With Preserved Ejection Fraction, NCT02053493; Inorganic Nitrite Delivery to Improve Exercise Capacity in HFpEF, NCT02742129; Inorganic Nitrite to Enhance Benefits From Exercise Training in Heart Failure With Preserved Ejection Fraction [HFpEF], NCT02713126).
Collapse
Affiliation(s)
- Alessio Alogna
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA; Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Campus Virchow-Klinikum, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Katlyn E Koepp
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael Sabbah
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jair M Espindola Netto
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | | | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA; Division of General Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Carolyn S P Lam
- National Heart Centre Singapore and Duke-National University of Singapore, Singapore
| | - Masaru Obokata
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark C Petrie
- BHF Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hidemi Sorimachi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Adriaan Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Margaret M Redfield
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
36
|
Golino M, Moroni F, Abbate A. Connecting the Dots: Inflammatory Burden and Outcomes in Heart Failure. J Am Heart Assoc 2023; 12:e031786. [PMID: 37776202 PMCID: PMC10727251 DOI: 10.1161/jaha.123.031786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 10/02/2023]
Affiliation(s)
- Michele Golino
- Robert M. Berne Cardiovascular Research Center, and Division of CardiologyUniversity of VirginiaCharlottesvilleVAUSA
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVAUSA
| | - Francesco Moroni
- Robert M. Berne Cardiovascular Research Center, and Division of CardiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, and Division of CardiologyUniversity of VirginiaCharlottesvilleVAUSA
| |
Collapse
|
37
|
Moroni F, Golino M, Carbone S, Trankle C, Del Buono MG, Talasaz A, Arena R, Canada JM, Biondi‐Zoccai G, Van Tassel B, Abbate A. Interleukin-1 blockade in heart failure: an on-treatment and off-treatment cardiorespiratory fitness analysis. ESC Heart Fail 2023; 10:3199-3202. [PMID: 37403287 PMCID: PMC10567671 DOI: 10.1002/ehf2.14460] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/30/2023] [Accepted: 06/21/2023] [Indexed: 07/06/2023] Open
Abstract
AIMS Interleukin-1 (IL-1) blockade may improve exercise capacity in patients with heart failure (HF) patients. The extent of the improvement and its persistence beyond discontinuation of IL-1 blockade is unknown. METHODS AND RESULTS The primary objective was to determine changes in cardiorespiratory fitness and cardiac function on-treatment with IL-1 blocker, anakinra, and off-treatment, after treatment cessation. We performed cardiopulmonary exercise testing, Doppler echocardiography, and biomarkers in 73 patients with HF, 37 (51%) females, 52 (71%) Black-African-American, before and after treatment with anakinra 100 mg daily. In a subset of 46 patients, testing was also repeated after treatment cessation. Quality of life was assessed in each patient using standardized questionnaires. Data are presented as median and interquartile range. Treatment with anakinra for 4 [2-12] weeks was associated with a significant improvement in high-sensitivity C-reactive protein (from 6.2 [3.3-15.4] to 1.4 [0.8-3.4] mg/L, P < 0.001), peak oxygen consumption (VO2peak , from 13.9 [11.6-16.6] to 15.2 [12.9-17.4] mL/kg/min, P < 0.001). Ventilatory efficiency, exercise time, Doppler-derived signs and biomarkers of elevated intracardiac pressures, and quality-of-life measures also improved with anakinra. In the 46 patients in whom off-treatment data were available 12 [4-12] weeks later, many of the favourable changes seen with anakinra were largely reversed (from 1.5 [1.0-3.4] to 5.9 [1.8-13.1], P = 0.001 for C-reactive protein, and from 16.2 [14.0-18.4] to 14.9 [11.5-17.8] mL/kg/min, P = 0.017, for VO2peak ). CONCLUSIONS These data validate IL-1 as an active and dynamic modulator of cardiac function and cardiorespiratory fitness in HF.
Collapse
Affiliation(s)
- Francesco Moroni
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVAUSA
- Department of Internal MedicineUniversity of VirginiaCharlottesvilleVAUSA
- Department of Medicine, Division of Cardiovascular MedicineUniversità Milano‐BicoccaMilanItaly
| | - Michele Golino
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVAUSA
- Department of Medicine and SurgeryUniversity of InsubriaVareseItaly
| | - Salvatore Carbone
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVAUSA
- Department of Kinesiology & Health Sciences, College of Humanities & SciencesVirginia Commonwealth UniversityRichmondVAUSA
| | - Cory Trankle
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVAUSA
| | - Marco Giuseppe Del Buono
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVAUSA
- Department of Cardiovascular MedicineFondazione Policlinico Universitario A. Gemelli IRCCSRomeItaly
| | - Azita Talasaz
- Department of Pharmacotherapy & Outcomes SciencesVirginia Commonwealth UniversityRichmondVAUSA
| | - Ross Arena
- Department of Physical Therapy, College of Applied Health SciencesUniversity of Illinois ChicagoChicagoILUSA
| | - Justin M. Canada
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVAUSA
| | - Giuseppe Biondi‐Zoccai
- Department of Medical‐Surgical Sciences and BiotechnologiesSapienza University of RomeLatinaItaly
- Mediterranea CardiocentroNaplesItaly
| | - Benjamin Van Tassel
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVAUSA
- Department of Pharmacotherapy & Outcomes SciencesVirginia Commonwealth UniversityRichmondVAUSA
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, and Division of CardiologyUniversity of VirginiaCharlottesvilleVAUSA
| |
Collapse
|
38
|
Golino M, Moroni F, Carbone S, Corna G, Trankle C, Billingsley HE, Del Buono MG, Talasaz AH, Thomas GK, De Ponti R, Turlington J, Markley R, Arena R, Canada JM, Van Tassell B, Abbate A. Differential Response to Interleukin-1 Blockade With Anakinra on Cardiorespiratory Fitness in Patients With Heart Failure With Preserved Ejection Fraction Stratified According to Left Ventricular Ejection Fraction. J Am Heart Assoc 2023; 12:e031251. [PMID: 37624043 PMCID: PMC10547307 DOI: 10.1161/jaha.123.031251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/18/2023] [Indexed: 08/26/2023]
Affiliation(s)
- Michele Golino
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVA
- Department of Medicine and SurgeryUniversity of InsubriaVareseItaly
| | - Francesco Moroni
- Department of Internal MedicineUniversity of VirginiaCharlottesvilleVA
- Department of MedicineUniversità Milano‐BicoccaMilanItaly
| | - Salvatore Carbone
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVA
- Department of Kinesiology and Health Sciences, College of Humanities and SciencesVirginia Commonwealth UniversityRichmondVA
| | - Giuliana Corna
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVA
| | - Cory Trankle
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVA
| | - Hayley E. Billingsley
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVA
- Department of Kinesiology and Health Sciences, College of Humanities and SciencesVirginia Commonwealth UniversityRichmondVA
| | - Marco G. Del Buono
- Department of Cardiovascular MedicineFondazione Policlinico Universitario A. Gemelli IRCCSRomeItaly
| | - Azita H. Talasaz
- Department of Pharmacotherapy and Outcomes SciencesVirginia Commonwealth UniversityRichmondVA
| | | | - Roberto De Ponti
- Department of Medicine and SurgeryUniversity of InsubriaVareseItaly
| | | | | | - Ross Arena
- Department of Physical Therapy, College of Applied Health SciencesUniversity of Illinois at ChicagoChicagoIL
| | | | - Benjamin Van Tassell
- Pauley Heart CenterVirginia Commonwealth UniversityRichmondVA
- Department of Pharmacotherapy and Outcomes SciencesVirginia Commonwealth UniversityRichmondVA
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, and Division of CardiologyUniversity of VirginiaCharlottesvilleVA
| |
Collapse
|
39
|
Daou D, Gillette TG, Hill JA. Inflammatory Mechanisms in Heart Failure with Preserved Ejection Fraction. Physiology (Bethesda) 2023; 38:0. [PMID: 37013947 PMCID: PMC10396273 DOI: 10.1152/physiol.00004.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/27/2023] [Accepted: 04/02/2023] [Indexed: 04/05/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is now the most common form of heart failure and a significant public health concern for which limited effective therapies exist. Inflammation triggered by comorbidity burden is a critical element of HFpEF pathophysiology. Here, we discuss evidence for comorbidity-driven systemic and myocardial inflammation and the mechanistic role of inflammation in pathological myocardial remodeling in HFpEF.
Collapse
Affiliation(s)
- Daniel Daou
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Thomas G Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
40
|
Lund LH, Lam CS, Pizzato PE, Gabrielsen A, Michaëlsson E, Nelander K, Ericsson H, Holden J, Folkvaljon F, Mattsson A, Collén A, Aurell M, Whatling C, Baldus S, Drelich G, Goudev A, Merkely B, Bergh N, Shah SJ. Rationale and design of ENDEAVOR: A sequential phase 2b-3 randomized clinical trial to evaluate the effect of myeloperoxidase inhibition on symptoms and exercise capacity in heart failure with preserved or mildly reduced ejection fraction. Eur J Heart Fail 2023; 25:1696-1707. [PMID: 37470101 PMCID: PMC10592288 DOI: 10.1002/ejhf.2977] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/15/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023] Open
Abstract
AIMS Mitiperstat (formerly AZD4831) is a novel selective myeloperoxidase inhibitor. Currently, no effective therapies target comorbidity-induced systemic inflammation, which may be a key mechanism underlying heart failure with preserved or mildly reduced ejection fraction (HFpEF/HFmrEF). Circulating neutrophils secrete myeloperoxidase, causing oxidative stress, microvascular endothelial dysfunction, interstitial fibrosis, cardiomyocyte remodelling and diastolic dysfunction. Mitiperstat may therefore improve function of the heart and other organs, and ameliorate heart failure symptoms and exercise intolerance. ENDEAVOR is a combined, seamless phase 2b-3 study of the efficacy and safety of mitiperstat in patients with HFpEF/HFmrEF. METHODS In phase 2b, approximately 660 patients with heart failure and ejection fraction >40% are being randomized 1:1:1 to mitiperstat 2.5 mg, 5 mg or placebo for 48 weeks. Eligible patients have baseline 6-min walk distance (6MWD) of 30-400 m with a <50 m difference between screening and randomization and Kansas City Cardiomyopathy Questionnaire total symptom score (KCCQ-TSS) ≤90 points at screening and randomization. The dual primary endpoints are change from baseline to week 16 in 6MWD and KCCQ-TSS. The sample size provides 85% power to detect placebo-adjusted improvements of 21 m in 6MWD and 6.0 points in KCCQ-TSS at overall two-sided alpha of 0.05. Safety is monitored throughout treatment, with a focus on maculopapular rash. In phase 3 of ENDEAVOR, approximately 820 patients will be randomized 1:1 to mitiperstat or placebo. CONCLUSION ENDEAVOR is the first phase 2b-3 study to evaluate whether myeloperoxidase inhibition can improve symptoms and exercise capacity in patients with HFpEF/HFmrEF.
Collapse
Affiliation(s)
- Lars H. Lund
- Department of Medicine, Karolinska Institute, and Heart, Vascular and Neuro Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Carolyn S.P. Lam
- National Heart Centre Singapore and Duke National University of Singapore, Singapore
| | - Patricia E. Pizzato
- Early Clinical Development, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Gabrielsen
- Early Clinical Development, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Erik Michaëlsson
- Early Clinical Development, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Karin Nelander
- Early Biometrics and Statistical Innovation, Data Science and AI, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Hans Ericsson
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Julie Holden
- Patient Safety, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Folke Folkvaljon
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Andrea Mattsson
- Late-Stage Development, Cardiovascular, Renal and Metabolism – Biometrics, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Collén
- Projects, Research and Early Development, Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Malin Aurell
- Early Clinical Development, Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Carl Whatling
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stephan Baldus
- Department of Internal Medicine and Cardiology, University Hospital Cologne, Cologne, Germany
| | | | - Assen Goudev
- Clinic of Cardiology, Tsaritsa Joanna University Hospital – ISUL, Sofia, Bulgaria
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Niklas Bergh
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sanjiv J. Shah
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
41
|
Li Z, Lin C, Cai X, Hu S, Lv F, Yang W, Zhu X, Ji L. Anti-inflammatory therapies were associated with reduced risk of myocardial infarction in patients with established cardiovascular disease or high cardiovascular risks: A systematic review and meta-analysis of randomized controlled trials. Atherosclerosis 2023; 379:117181. [PMID: 37527612 DOI: 10.1016/j.atherosclerosis.2023.06.972] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND AND AIMS We aimed to evaluate the association between anti-inflammatory therapies and the incidence of cardiovascular events in patients with established cardiovascular disease (CVD) or high cardiovascular risks. METHODS Literature retrieval was conducted in PubMed, Medline, Embase, the Cochrane Central Register of Controlled Trials and Clinicaltrial.gov website from the inception to December 2022. Randomized controlled trials comparing anti-inflammatory therapies with placebo in patients with established CVD or high cardiovascular risks were included. The results of the meta-analysis were computed as the risk ratio (RR) with 95% confidence interval (CI). RESULTS Compared with placebo, anti-inflammatory therapies were associated with decreased incidence of myocardial infarction (MI) (RR = 0.93, 95% CI, 0.88 to 0.98), which was mainly driven by therapies targeting central IL-6 signaling pathway (RR = 0.83, 95% CI, 0.74 to 0.93). IL-1 inhibitors treatment was associated with reduced risks of heart failure (RR = 0.38, 95% CI, 0.18 to 0.80) while lower incidence of stroke was observed in patients with colchicine treatment (RR = 0.47, 95% CI, 0.28 to 0.77). MI events were less frequent in patients over 65 years of age (RR = 0.90, 95% CI, 0.83 to 0.98) or with follow-up duration over 1 year (RR = 0.90, 95% CI, 0.85 to 0.96) when comparing anti-inflammatory therapies with placebo. CONCLUSIONS Anti-inflammatory therapies, especially those targeting the central IL-6 signaling pathway, may serve as promising treating strategies to ameliorate the risk of MI. IL-1 inhibitor and colchicine were associated with decreased risks of heart failure and stroke, respectively. MI risk reduction by anti-inflammatory therapies seemed to be more prominent in older patients with long follow-up duration.
Collapse
Affiliation(s)
- Zonglin Li
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Chu Lin
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China.
| | - Suiyuan Hu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Fang Lv
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Wenjia Yang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Xingyun Zhu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
42
|
Vaz-Salvador P, Adão R, Vasconcelos I, Leite-Moreira AF, Brás-Silva C. Heart Failure with Preserved Ejection Fraction: a Pharmacotherapeutic Update. Cardiovasc Drugs Ther 2023; 37:815-832. [PMID: 35098432 PMCID: PMC8801287 DOI: 10.1007/s10557-021-07306-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 02/06/2023]
Abstract
While guidelines for management of heart failure with reduced ejection fraction (HFrEF) are consensual and have led to improved survival, treatment options for heart failure with preserved ejection fraction (HFpEF) remain limited and aim primarily for symptom relief and improvement of quality of life. Due to the shortage of therapeutic options, several drugs have been investigated in multiple clinical trials. The majority of these trials have reported disappointing results and have suggested that HFpEF might not be as simply described by ejection fraction as previously though. In fact, HFpEF is a complex clinical syndrome with various comorbidities and overlapping distinct phenotypes that could benefit from personalized therapeutic approaches. This review summarizes the results from the most recent phase III clinical trials for HFpEF and the most promising drugs arising from phase II trials as well as the various challenges that are currently holding back the development of new pharmacotherapeutic options for these patients.
Collapse
Affiliation(s)
- Pedro Vaz-Salvador
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rui Adão
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Inês Vasconcelos
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Adelino F. Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research and Development Center - UnIC, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Rua Do Campo Alegre, 823 4150-180 Porto, Portugal
| |
Collapse
|
43
|
BoYe, Bradshaw AD, Abrahante JE, Dragon JA, Häußler TN, Bell SP, Hirashima F, LeWinter M, Zile MR, Meyer M. Left Ventricular Gene Expression in Heart Failure With Preserved Ejection Fraction-Profibrotic and Proinflammatory Pathways and Genes. Circ Heart Fail 2023; 16:e010395. [PMID: 37582166 PMCID: PMC10430768 DOI: 10.1161/circheartfailure.123.010395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/16/2023] [Indexed: 08/17/2023]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is increasingly prevalent and has few treatments. The molecular mechanisms and resultant signaling pathways that underlie the development of HFpEF are poorly defined. It has been proposed that activation of proinflammatory pathways plays a role in the development of cardiac fibrosis. The signature of gene expression (transcriptome) of previously validated left ventricular biopsies obtained from patients with HFpEF and matched referent controls allows for an unbiased assessment of proinflammatory and profibrotic signaling pathways and genes. METHODS Epicardial left ventricular biopsies from stringently selected HFpEF patients (HFpEF, n=16) and referent control patients (CTR, n=14) were obtained during aortocoronary bypass surgery. The subepicardial myocardium was flash-frozen to build a repository that was parallel-processed for RNA sequencing to allow for an unsupervised in-depth comparison of the left ventricular transcriptome. RESULTS The average patient age was 67±10 years. When compared with controls, patients with HFpEF were hypertensive with a higher body mass index (kg/m2: 30±5 versus 37±6; P<0.01) and elevated NT-proBNP levels (pg/mL: 155 [89-328] versus 1554 [888-2178]; P<0.001). The transcriptome analysis revealed differential expression of 477 genes many of which were involved in profibrotic pathways including extracellular matrix production and posttranslational modification but no proinflammatory signature. CONCLUSIONS The transcriptome analysis of left ventricular myocardial samples from patients with HFpEF confirms an overabundant extracellular matrix gene expression, the basis of myocardial fibrosis, without a signature of activated proinflammatory pathways or genes.
Collapse
Affiliation(s)
- BoYe
- Lillehei Heart Institute and Genomics Center of the University of Minnesota, Minneapolis, MN, USA
| | - Amy D Bradshaw
- Medical University of South Carolina, RHJ Department of Veterans Affairs Medical Center, Charleston, SC, USA
| | - Juan E. Abrahante
- Lillehei Heart Institute and Genomics Center of the University of Minnesota, Minneapolis, MN, USA
| | - Julie A. Dragon
- University of Vermont Medical Center, Cardiology, Cardiothoracic Surgery and Vermont Integrative Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Tim N. Häußler
- University of Vermont Medical Center, Cardiology, Cardiothoracic Surgery and Vermont Integrative Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Stephen P. Bell
- University of Vermont Medical Center, Cardiology, Cardiothoracic Surgery and Vermont Integrative Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Fuyuki Hirashima
- University of Vermont Medical Center, Cardiology, Cardiothoracic Surgery and Vermont Integrative Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Martin LeWinter
- University of Vermont Medical Center, Cardiology, Cardiothoracic Surgery and Vermont Integrative Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Michael R. Zile
- Medical University of South Carolina, RHJ Department of Veterans Affairs Medical Center, Charleston, SC, USA
| | - Markus Meyer
- Lillehei Heart Institute and Genomics Center of the University of Minnesota, Minneapolis, MN, USA
- University of Vermont Medical Center, Cardiology, Cardiothoracic Surgery and Vermont Integrative Genomics Resource, University of Vermont, Burlington, VT, USA
| |
Collapse
|
44
|
Sikking MA, Stroeks SLVM, Waring OJ, Henkens MTHM, Riksen NP, Hoischen A, Heymans SRB, Verdonschot JAJ. Clonal Hematopoiesis of Indeterminate Potential From a Heart Failure Specialist's Point of View. J Am Heart Assoc 2023; 12:e030603. [PMID: 37489738 PMCID: PMC10492961 DOI: 10.1161/jaha.123.030603] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/06/2023] [Indexed: 07/26/2023]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is a common bone marrow abnormality induced by age-related DNA mutations, which give rise to proinflammatory immune cells. These immune cells exacerbate atherosclerotic cardiovascular disease and may induce or accelerate heart failure. The mechanisms involved are complex but point toward a central role for proinflammatory macrophages and an inflammasome-dependent immune response (IL-1 [interleukin-1] and IL-6 [interleukin-6]) in the atherosclerotic plaque or directly in the myocardium. Intracardiac inflammation may decrease cardiac function and induce cardiac fibrosis, even in the absence of atherosclerotic cardiovascular disease. The pathophysiology and consequences of CHIP may differ among implicated genes as well as subgroups of patients with heart failure, based on cause (ischemic versus nonischemic) and ejection fraction (reduced ejection fraction versus preserved ejection fraction). Evidence is accumulating that CHIP is associated with cardiovascular mortality in ischemic and nonischemic heart failure with reduced ejection fraction and involved in the development of heart failure with preserved ejection fraction. CHIP and corresponding inflammatory pathways provide a highly potent therapeutic target. Randomized controlled trials in patients with well-phenotyped heart failure, where readily available anti-inflammatory therapies are used to intervene with clonal hematopoiesis, may pave the way for a new area of heart failure treatment. The first clinical trials that target CHIP are already registered.
Collapse
Affiliation(s)
- Maurits A. Sikking
- Department of CardiologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
| | - Sophie L. V. M. Stroeks
- Department of CardiologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
| | - Olivia J. Waring
- Department of PathologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
| | - Michiel T. H. M. Henkens
- Department of PathologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
- Netherlands Heart Institute (NLHI)Utrechtthe Netherlands
| | - Niels P. Riksen
- Department of Internal MedicineRadboud University Medical CenterNijmegenthe Netherlands
| | - Alexander Hoischen
- Department of Human GeneticsRadboud University Medical CenterNijmegenthe Netherlands
| | - Stephane R. B. Heymans
- Department of CardiologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
- Department of Cardiovascular ResearchUniversity of LeuvenBelgium
| | - Job A. J. Verdonschot
- Department of CardiologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
- Department of Clinical GeneticsMaastricht University Medical Center (MUMC)Maastrichtthe Netherlands
| |
Collapse
|
45
|
Affiliation(s)
- Roua Hassoun
- Department of Cellular and Translational Physiology, Institute of Physiology (R.H., N.H.), Ruhr University Bochum, Germany
- Institut für Forschung und Lehre, Molecular and Experimental Cardiology (R.H., N.H.), Ruhr University Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB (R.H., N.H.), Ruhr University Bochum, Germany
| | - Nazha Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology (R.H., N.H.), Ruhr University Bochum, Germany
- Institut für Forschung und Lehre, Molecular and Experimental Cardiology (R.H., N.H.), Ruhr University Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB (R.H., N.H.), Ruhr University Bochum, Germany
| |
Collapse
|
46
|
Shchendrygina A, Rachina S, Cherkasova N, Suvorov A, Komarova I, Mukhina N, Ananicheva N, Gasanova D, Sitnikova V, Koposova A, Smirnova J, Moiseewa E, Drogashevskaya D. 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:e002360. [PMID: 37586845 PMCID: PMC10432645 DOI: 10.1136/openhrt-2023-002360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/03/2023] [Indexed: 08/18/2023] Open
Abstract
INTRODUCTION Systemic low-grade inflammation is a fundamental pathophysiological mechanism of heart failure with preserved left ventricular ejection fraction (HFpEF). The efficacy of anti-inflammatory therapy in HFpEF is largely understudied. The aim of the study is to assess the anti-inflammatory effect of colchicine in HFpEF by looking at inflammatory biomarkers: high-sensitivity C reactive protein (hsCRP) and soluble suppression of tumorigenicity 2 (sST2). METHODS AND ANALYSIS This is a single-centre, prospective, randomised controlled, open-label, blinded-endpoint crossover clinical trial of stable but symptomatic patients with HFpEF. Patients will be randomised to either colchicine treatment 0.5 mg two times per day or usual care for 12 weeks followed by a 2-week washout period and crossover to 12 weeks of treatment with the alternate therapy. The primary objective is to investigate if administration of colchicine compared with usual care reduces inflammation in patients with HFpEF measured by primary endpoint sST2 and co-primary endpoint hsCRP at baseline and 12-week follow-up. Secondary objective is to determine if treatment with colchicine influences N-terminal pro-B-type natriuretic peptide levels, left ventricular diastolic function and remodelling, right ventricular systolic function and left atrial volumetric characteristics. We are aiming to enrol a total of 40 participants. This trial will answer the question if colchicine treatment reduces systemic low-grade inflammation and influences left ventricular diastolic function and remodelling with patients with HFpEF. ETHICS AND DISSEMINATION Ethical approval was obtained from the Ethics Committee of Sechenov University (reference: 03-22). TRIAL REGISTRATION NUMBER NCT05637398.
Collapse
Affiliation(s)
| | - Svetlana Rachina
- I M Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Natalia Cherkasova
- I M Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Aleksandr Suvorov
- Institute of Biodesign and Complex Systems Modeling, I M Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Irina Komarova
- I M Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Nadezhda Mukhina
- I M Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Natalia Ananicheva
- City Clinical Hospital named after S S Yudin, Moscow, Russian Federation
| | - Diana Gasanova
- I M Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Violetta Sitnikova
- I M Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Aleksandra Koposova
- I M Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Julia Smirnova
- I M Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Elizaveta Moiseewa
- I M Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | | |
Collapse
|
47
|
Peh ZH, Dihoum A, Hutton D, Arthur JSC, Rena G, Khan F, Lang CC, Mordi IR. Inflammation as a therapeutic target in heart failure with preserved ejection fraction. Front Cardiovasc Med 2023; 10:1125687. [PMID: 37456816 PMCID: PMC10339321 DOI: 10.3389/fcvm.2023.1125687] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for around half of all cases of heart failure and may become the dominant type of heart failure in the near future. Unlike HF with reduced ejection fraction there are few evidence-based treatment strategies available. There is a significant unmet need for new strategies to improve clinical outcomes in HFpEF patients. Inflammation is widely thought to play a key role in HFpEF pathophysiology and may represent a viable treatment target. In this review focusing predominantly on clinical studies, we will summarise the role of inflammation in HFpEF and discuss potential therapeutic strategies targeting inflammation.
Collapse
Affiliation(s)
- Zhen Hui Peh
- School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - Adel Dihoum
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Dana Hutton
- School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - J. Simon C. Arthur
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Graham Rena
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Faisel Khan
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Ify R. Mordi
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
48
|
Shi YJ, Dong GJ, Guo M. Targeting epicardial adipose tissue: A potential therapeutic strategy for heart failure with preserved ejection fraction with type 2 diabetes mellitus. World J Diabetes 2023; 14:724-740. [PMID: 37383601 PMCID: PMC10294070 DOI: 10.4239/wjd.v14.i6.724] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/10/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome with various comorbidities, multiple cardiac and extracardiac pathophysiologic abnormalities, and diverse phenotypic presentations. Since HFpEF is a heterogeneous disease with different phenotypes, individualized treatment is required. HFpEF with type 2 diabetes mellitus (T2DM) represents a specific phenotype of HFpEF, with about 45%-50% of HFpEF patients suffering from T2DM. Systemic inflammation associated with dysregulated glucose metabolism is a critical pathological mechanism of HFpEF with T2DM, which is intimately related to the expansion and dysfunction (inflammation and hypermetabolic activity) of epicardial adipose tissue (EAT). EAT is well established as a very active endocrine organ that can regulate the pathophysiological processes of HFpEF with T2DM through the paracrine and endocrine mechanisms. Therefore, suppressing abnormal EAT expansion may be a promising therapeutic strategy for HFpEF with T2DM. Although there is no treatment specifically for EAT, lifestyle management, bariatric surgery, and some pharmaceutical interventions (anti-cytokine drugs, statins, proprotein convertase subtilisin/kexin type 9 inhibitors, metformin, glucagon-like peptide-1 receptor agonists, and especially sodium-glucose cotransporter-2 inhibitors) have been shown to attenuate the inflammatory response or expansion of EAT. Importantly, these treatments may be beneficial in improving the clinical symptoms or prognosis of patients with HFpEF. Accordingly, well-designed randomized controlled trials are needed to validate the efficacy of current therapies. In addition, more novel and effective therapies targeting EAT are needed in the future.
Collapse
Affiliation(s)
- Yu-Jiao Shi
- Department of Cardiovascular Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Guo-Ju Dong
- Department of Cardiovascular Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Ming Guo
- Department of Cardiovascular Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing 100091, China
| |
Collapse
|
49
|
Stencel J, Alai HR, Dhore-patil A, Urina-Jassir D, Le Jemtel TH. Obesity, Preserved Ejection Fraction Heart Failure, and Left Ventricular Remodeling. J Clin Med 2023; 12:3341. [PMID: 37176781 PMCID: PMC10179420 DOI: 10.3390/jcm12093341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/30/2023] [Accepted: 05/06/2023] [Indexed: 05/15/2023] Open
Abstract
Owing to the overwhelming obesity epidemic, preserved ejection fraction heart failure commonly ensues in patients with severe obesity and the obese phenotype of preserved ejection fraction heart failure is now commonplace in clinical practice. Severe obesity and preserved ejection fraction heart failure share congruent cardiovascular, immune, and renal derangements that make it difficult to ascertain whether the obese phenotype of preserved ejection fraction heart failure is the convergence of two highly prevalent conditions or severe obesity enables the development and progression of the syndrome of preserved ejection fraction heart failure. Nevertheless, the obese phenotype of preserved ejection fraction heart failure provides a unique opportunity to assess whether sustained and sizeable loss of excess body weight via metabolic bariatric surgery reverses the concentric left ventricular remodeling that patients with preserved ejection fraction heart failure commonly display.
Collapse
Affiliation(s)
- Jason Stencel
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, Tulane University Heart and Vascular Institute, New Orleans, LA 70112, USA; (J.S.); (H.R.A.); (A.D.-p.); (D.U.-J.)
| | - Hamid R. Alai
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, Tulane University Heart and Vascular Institute, New Orleans, LA 70112, USA; (J.S.); (H.R.A.); (A.D.-p.); (D.U.-J.)
- Southeast Louisiana VA Healthcare System (SLVHCS), New Orleans, LA 70119, USA
| | - Aneesh Dhore-patil
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, Tulane University Heart and Vascular Institute, New Orleans, LA 70112, USA; (J.S.); (H.R.A.); (A.D.-p.); (D.U.-J.)
| | - Daniela Urina-Jassir
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, Tulane University Heart and Vascular Institute, New Orleans, LA 70112, USA; (J.S.); (H.R.A.); (A.D.-p.); (D.U.-J.)
| | - Thierry H. Le Jemtel
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, Tulane University Heart and Vascular Institute, New Orleans, LA 70112, USA; (J.S.); (H.R.A.); (A.D.-p.); (D.U.-J.)
| |
Collapse
|
50
|
Golino M, Moroni F, Del Buono MG, Canada JM, Talasaz AH, Piñel S, Mbualungu J, Vecchiè A, Ho AC(J, Thomas GK, Carbone S, Billingsley HE, Turlington J, Markley R, Trankle C, De Ponti R, Van Tassell B, Abbate A. Change in Eosinophil Count in Patients with Heart Failure Treated with Anakinra. Cells 2023; 12:1129. [PMID: 37190038 PMCID: PMC10137267 DOI: 10.3390/cells12081129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Interleukin-1 blockade with anakinra leads to a transient increase in eosinophil blood count (eosinophils) in patients with acute myocardial infarction. We aimed to investigate the effect of anakinra on changes in eosinophils in patients with heart failure (HF) and their correlation with cardiorespiratory fitness (CRF). METHODS We measured eosinophils in 64 patients with HF (50% females), 55 (51-63) years of age, before and after treatment, and, in a subset of 41 patients, also after treatment cessation. We also evaluated CRF, measuring peak oxygen consumption (VO2) with a treadmill test. RESULTS Treatment with anakinra significantly and transiently increased eosinophils, from 0.2 [0.1-0.3] to 0.3 [0.1-0.4] × 103 cells/µL (p < 0.001) and from 0.3 [0.2-0.5] to 0.2 [0.1-0.3] × 103 cells/µL, with suspension (p < 0.001). Changes in eosinophils correlated with the changes in peak VO2 (Spearman's Rho = +0.228, p = 0.020). Eosinophils were higher in patients with injection site reactions (ISR) (n = 8, 13%; 0.5 [0.4-0.6] vs. 0.2 [0.1-0.4] × 103 cells/µL, p = 0.023), who also showed a greater increase in peak VO2 (3.0 [0.9-4.3] vs. 0.3 [-0.6-1.8] mLO2·kg-1·min-1, p = 0.015). CONCLUSION Patients with HF treated with anakinra experience a transient increase in eosinophils, which is associated with ISR and a greater improvement in peak VO2.
Collapse
Affiliation(s)
- Michele Golino
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Francesco Moroni
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
- Department of Internal Medicine, University of Virginia, Charlottesville, VA 22904, USA
- Department of Medicine, Università Milano-Bicocca, 20126 Milan, Italy
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Justin M. Canada
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
| | - Azita H. Talasaz
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
- Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Sebastian Piñel
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
| | - James Mbualungu
- Robert M. Berne Cardiovascular Research Center, Division of Cardiology—Heart and Vascular Center, University of Virginia, Charlottesville, VA 22904, USA
| | - Alessandra Vecchiè
- Medicina Generale 1, Medical Center, Department of Internal Medicine, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, 21100 Varese, Italy
| | - Ai-Chen (Jane) Ho
- Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
- Department of Clinical & Administrative Sciences, School of Pharmacy, Notre Dame of Maryland University, Baltimore, MD 21210, USA
| | - Georgia K. Thomas
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
| | - Salvatore Carbone
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Hayley E. Billingsley
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Jeremy Turlington
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
| | - Roshanak Markley
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
| | - Cory Trankle
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
| | - Roberto De Ponti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Benjamin Van Tassell
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
- Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Antonio Abbate
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23284, USA; (M.G.); (F.M.)
- Robert M. Berne Cardiovascular Research Center, Division of Cardiology—Heart and Vascular Center, University of Virginia, Charlottesville, VA 22904, USA
| |
Collapse
|