1
|
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
|
2
|
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
|
3
|
Cecrdlova E, Krupickova L, Fialova M, Novotny M, Tichanek F, Svachova V, Mezerova K, Viklicky O, Striz I. Insights into IL-1 family cytokines in kidney allograft transplantation: IL-18BP and free IL-18 as emerging biomarkers. Cytokine 2024; 180:156660. [PMID: 38801805 DOI: 10.1016/j.cyto.2024.156660] [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: 11/03/2023] [Revised: 04/15/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
Proinflammatory cytokines and their inhibitors are involved in the regulation of multiple immune reactions including response to transplanted organs. In this prospective study, we evaluated changes in serum concentrations of six IL-1 family cytokines (IL-1 alpha, IL-1 beta, IL-1RA, IL-18, IL-18BP, and IL-36 beta) in 138 kidney allograft recipients and 48 healthy donors. Samples were collected before transplantation and then after one week, three months and one year, additional sera were obtained at the day of biopsy positive for acute rejection. We have shown, that concentrations of proinflammatory members of the IL-1 family (IL-1β, IL-18, IL-36 β) and anti-inflammatory IL-18BP decreased immediately after the transplantation. The decline of serum IL-1RA and IL-1α was not observed in subjects with acute rejection. IL-18, including specifically its free form, is the only cytokine which increase serum concentrations in the period between one week and three months in both groups of patients without upregulation of its inhibitor, IL-18BP. Serum concentrations of calculated free IL-18 were upregulated in the acute rejection group at the time of acute rejection. We conclude that IL-1 family cytokines are involved mainly in early phases of the response to kidney allograft. Serum concentrations of free IL-18 and IL-18BP represent possible biomarkers of acute rejection, and targeting IL-18 might be of therapeutic value.
Collapse
Affiliation(s)
- E Cecrdlova
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic
| | - L Krupickova
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic
| | - M Fialova
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic
| | - M Novotny
- Institute for Clinical and Experimental Medicine, Transplant Center, Department of Nephrology, Prague, Czech Republic
| | - F Tichanek
- Institute for Clinical and Experimental Medicine, Department of Data Science, Prague, Czech Republic
| | - V Svachova
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic
| | - K Mezerova
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic
| | - O Viklicky
- Institute for Clinical and Experimental Medicine, Transplant Center, Department of Nephrology, Prague, Czech Republic
| | - I Striz
- Institute for Clinical and Experimental Medicine, Department of Clinical and Transplant Immunology, Prague, Czech Republic.
| |
Collapse
|
4
|
Moroni F, Corna G, Del Buono MG, Golino M, Talasaz AH, Decotto S, Markley R, Trankle C, Biondi-Zoccai G, Carbone S, Agatiello CR, Van Tassell B, Abbate A. Impact of C-reactive protein levels and role of anakinra in patients with ST-elevation myocardial infarction. Int J Cardiol 2024; 398:131610. [PMID: 38016623 PMCID: PMC10896664 DOI: 10.1016/j.ijcard.2023.131610] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/30/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Interleukin-1 blockade with anakinra reduces C-reactive protein (CRP) levels and prevents heart failure (HF) events after ST-segment myocardial infarction (STEMI). The effectiveness of anakinra according to the degree of systemic inflammation in STEMI has not been addressed. METHODS We analyzed 139 patients from three Virginia Commonwealth University Anakinra Response Trial randomized clinical trials to assess whether CRP levels predicted HF hospitalization or death in patients with STEMI, and if CRP levels influenced the effects of treatment with anakinra. RESULTS CRP cut-off levels for prediction of the composite of death or HF hospitalization for CRP at admission, 3 and 14 days were, respectively 6.45 mg/L (100% of sensitivity and 66.1% specificity), 26 mg/L (100% of sensitivity and 78% specificity) and 9.56 mg/L (100% of sensitivity and 80% specificity). More patients with elevated CRP levels died or had a HF hospitalization (5/47 [11%] vs 0/82 [0%], p = 0.004 for CRP at admission; 5/32 [15.6%] vs 0/92 [0%], p < 0.001 for day 3 and 5/26 [19%] vs 0/89 [0%], p < 0.001 for day 14). A greater number of patients treated with anakinra had low CRP levels at 3 and 14 days compared to placebo (Odds Ratio 0.11 [95% IC 0.04-0.28], p < 0.0001 and OR 0.35 [95% CI 0.14-0.86], p = 0.02, respectively). Anakinra significantly prevented death or HF hospitalization in patients with high inflammatory burden (p = 0.04 for admission, p = 0.24 for day 3, and p = 0.05 for day 14). CONCLUSION Patients with elevated CRP had higher incidence of HF hospitalization or death. Anakinra reduced the number of patients with elevated CRP levels and prevented death or HF hospitalization in patients with elevated CRP levels.
Collapse
Affiliation(s)
- Francesco Moroni
- Robert M. Berne Cardiovascular Research Center, and Division of Cardiology, University of Virginia, Charlottesville, VA, United States; Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States; Cardiovascular Division, Medicine Department, Università Milano-Bicocca, Milan, Italy
| | - Giuliana Corna
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States; Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Marco Giuseppe Del Buono
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States; Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Michele Golino
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States; Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Azita H Talasaz
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Santiago Decotto
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States; Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Roshanak Markley
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Cory Trankle
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Roma, Italy; Mediterranea Cardiocentro, Via Orazio, 2, 80122 Napoli, NA, Italy
| | - Salvatore Carbone
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Carla R Agatiello
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Benjamin Van Tassell
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States.
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, and Division of Cardiology, University of Virginia, Charlottesville, VA, United States.
| |
Collapse
|
5
|
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
|
6
|
Secondary Cardiovascular Prevention after Acute Coronary Syndrome: Emerging Risk Factors and Novel Therapeutic Targets. J Clin Med 2023; 12:jcm12062161. [PMID: 36983163 PMCID: PMC10056379 DOI: 10.3390/jcm12062161] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
The control of cardiovascular risk factors, the promotion of a healthy lifestyle, and antithrombotic therapy are the cornerstones of secondary prevention after acute coronary syndrome (ACS). However, many patients have recurrent ischemic events despite the optimal control of traditional modifiable risk factors and the use of tailored pharmacological therapy, including new-generation antiplatelet and lipid-lowering agents. This evidence emphasizes the importance of identifying novel risk factors and targets to optimize secondary preventive strategies. Lipoprotein(a) (Lp(a)) has emerged as an independent predictor of adverse events after ACS. New molecules such as anti-PCSK9 monoclonal antibodies, small interfering RNAs, and antisense oligonucleotides can reduce plasma Lp(a) levels and are associated with a long-term outcome benefit after the index event. The inflammatory stimulus and the inflammasome, pivotal elements in the development and progression of atherosclerosis, have been widely investigated in patients with coronary artery disease. More recently, randomized clinical trials including post-ACS patients treated with colchicine and monoclonal antibodies targeting cytokines yielded promising results in the reduction in major cardiovascular events after an ACS. Gut dysbiosis has also raised great interest for its potential pathophysiological role in cardiovascular disease. This evidence, albeit preliminary and needing confirmation by larger population-based studies, suggests the possibility of targeting the gut microbiome in particularly high-risk populations. The risk of recurrent ischemic events after ACS is related to the complex interaction between intrinsic predisposing factors and environmental triggers. The identification of novel risk factors and targets is fundamental to customizing patient clinical management with a precision medicine perspective.
Collapse
|
7
|
Mauro AG, Bonaventura A, Vecchié A, Mezzaroma E, Carbone S, Narayan P, Potere N, Cannatà A, Paolini JF, Bussani R, Montecucco F, Sinagra G, Van Tassel BW, Abbate A, Toldo S. The Role of NLRP3 Inflammasome in Pericarditis: Potential for Therapeutic Approaches. JACC Basic Transl Sci 2021; 6:137-150. [PMID: 33665514 PMCID: PMC7907621 DOI: 10.1016/j.jacbts.2020.11.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 01/08/2023]
Abstract
Human samples of patients with chronic pericarditis and appropriate control subjects were stained for the inflammasome components. A mouse model of pericarditis was developed through the intrapericardial injection of zymosan A. Different inflammasome blockers were tested in the mouse model. Patients with pericarditis presented an intensification of the inflammasome activation compared with control subjects. The experimental model showed the pathological features of pericarditis. Among inflammasome blockers, NLRP3 inflammasome inhibitor, anakinra, and interleukin-1 trap were found to significantly improve pericardial alterations. Colchicine partially improved the pericardial inflammation. An intense activation of the inflammasome in pericarditis was demonstrated both in humans and in mice.
Collapse
Affiliation(s)
- Adolfo G. Mauro
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Aldo Bonaventura
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Alessandra Vecchié
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Eleonora Mezzaroma
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Salvatore Carbone
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Pratyush Narayan
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Nicola Potere
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Antonio Cannatà
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste, University of Trieste, Trieste, Italy
- Molecular Cardiology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
- Department of Cardiovascular Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | | | - Rossana Bussani
- Institute of Pathological Anatomy, Riuniti Hospital, University of Trieste, Trieste, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genova, Italian Cardiovascular Network, Genoa, Italy
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste, University of Trieste, Trieste, Italy
| | - Benjamin W. Van Tassel
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Antonio Abbate
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Stefano Toldo
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Cardiothoracic Surgery, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
8
|
Imazio M, Andreis A, De Ferrari GM, Cremer PC, Mardigyan V, Maestroni S, Luis SA, Lopalco G, Emmi G, Lotan D, Marcolongo R, Lazaros G, De Biasio M, Cantarini L, Dagna L, Cercek AC, Pivetta E, Varma B, Berkson L, Tombetti E, Iannone F, Prisco D, Caforio ALP, Vassilopoulos D, Tousoulis D, De Luca G, Giustetto C, Rinaldi M, Oh JK, Klein AL, Brucato A, Adler Y. Anakinra for corticosteroid-dependent and colchicine-resistant pericarditis: The IRAP (International Registry of Anakinra for Pericarditis) study. Eur J Prev Cardiol 2020; 27:956-964. [PMID: 31610707 DOI: 10.1177/2047487319879534] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
AIMS Novel therapies are needed for recurrent pericarditis, particularly when corticosteroid dependent and colchicine resistant. Based on limited data, interleukin-1 blockade with anakinra may be beneficial. The aim of this multicentre registry was to evaluate the broader effectiveness and safety of anakinra in a 'real world' population. METHODS AND RESULTS This registry enrolled consecutive patients with recurrent pericarditis who were corticosteroid dependent and colchicine resistant and treated with anakinra. The primary outcome was the pericarditis recurrence rate after treatment. Secondary outcomes included emergency department visits, hospitalisations, corticosteroid use and adverse events. Among 224 patients (46 ± 14 years old, 63% women, 75% idiopathic), the median duration of disease was 17 months (interquartile range 9-33). Most patients had elevated C-reactive protein (91%) and pericardial effusion (88%). After a median treatment of 6 months (3-12), pericarditis recurrences were reduced six-fold (2.33-0.39 per patient per year), emergency department admissions were reduced 11-fold (1.08-0.10 per patient per year), hospitalisations were reduced seven-fold (0.99-0.13 per patient per year). Corticosteroid use was decreased by anakinra (respectively from 80% to 27%; P < 0.001). No serious adverse events occurred; adverse events consisted mostly of transient skin reactions (38%) at the injection site. Adverse events led to discontinuation in 3%. A full-dose treatment duration of over 3 months followed by a tapering period of over 3 months were the therapeutic schemes associated with a lower risk of recurrence. CONCLUSION In patients with recurrent pericarditis, anakinra appears efficacious and safe in reducing recurrences, emergency department admissions and hospitalisations.
Collapse
Affiliation(s)
- Massimo Imazio
- Coordinating Center: University Cardiology, A.O.U. Città della Salute e della Scienza di Torino, Italy
| | - Alessandro Andreis
- Coordinating Center: University Cardiology, A.O.U. Città della Salute e della Scienza di Torino, Italy
| | - Gaetano Maria De Ferrari
- Coordinating Center: University Cardiology, A.O.U. Città della Salute e della Scienza di Torino, Italy
| | | | | | | | | | - Giuseppe Lopalco
- Department of Emergency and Organ Transplantation (DETO), University of Bari, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Firenze, Italy
| | - Dor Lotan
- Leviev Heart Center, Chaim Sheba Medical Center (affiliated to Tel Aviv University), Israel
| | - Renzo Marcolongo
- Department of Cardiac Thoracic Vascular Sciences and Public Health, Policlinico Universitario, Italy
| | - George Lazaros
- 1st Cardiology Clinic, Hippokration General Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | | | - Luca Cantarini
- Research Center of Systemic Autoinflammatory Diseases, University of Siena, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, Italy
| | | | - Emanuele Pivetta
- Emergency Medicine Division and High Dependency Unit and CPO Piemonte, A.O.U. Città della Salute e della Scienza di Torino, Italy
| | - Beni Varma
- Heart and Vascular Institute, Cleveland Clinic, USA
| | | | - Enrico Tombetti
- Dipartimento Scienze Cliniche e biomediche Luigi Sacco, ASST Fatebenefratelli-Sacco, Università degli Studi di Milano, Italy
| | - Florenzo Iannone
- Department of Emergency and Organ Transplantation (DETO), University of Bari, Italy
| | - Domenico Prisco
- Department of Experimental and Clinical Medicine, University of Firenze, Italy
| | - Alida Linda P Caforio
- Department of Cardiac Thoracic Vascular Sciences and Public Health, Policlinico Universitario, Italy
| | - Dimitrios Vassilopoulos
- 1st Cardiology Clinic, Hippokration General Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Dimitrios Tousoulis
- 1st Cardiology Clinic, Hippokration General Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, Italy
| | - Carla Giustetto
- Coordinating Center: University Cardiology, A.O.U. Città della Salute e della Scienza di Torino, Italy
| | - Mauro Rinaldi
- Coordinating Center: University Cardiology, A.O.U. Città della Salute e della Scienza di Torino, Italy
| | - Jae K Oh
- Department of Cardiovascular Medicine, Mayo Clinic, USA
| | | | - Antonio Brucato
- Dipartimento Scienze Cliniche e biomediche Luigi Sacco, ASST Fatebenefratelli-Sacco, Università degli Studi di Milano, Italy
| | - Yehuda Adler
- Leviev Heart Center, Chaim Sheba Medical Center (affiliated to Tel Aviv University), Israel
| |
Collapse
|
9
|
Hung AM, Tsuchida Y, Nowak KL, Sarkar S, Chonchol M, Whitfield V, Salas N, Dikalova A, Yancey PG, Huang J, Linton MF, Ikizler TA, Kon V. IL-1 Inhibition and Function of the HDL-Containing Fraction of Plasma in Patients with Stages 3 to 5 CKD. Clin J Am Soc Nephrol 2019; 14:702-711. [PMID: 31015261 PMCID: PMC6500942 DOI: 10.2215/cjn.04360418] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 03/15/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND OBJECTIVES Systemic inflammation modulates cardiovascular disease risk and functionality of HDL in the setting of CKD. Whether interventions that modify systemic inflammation can improve HDL function in CKD is unknown. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We conducted a post hoc analysis of two randomized, clinical trials, IL-1 trap in participants with GFR 15-59 ml/min per 1.73 m2 (study A) and IL-1 receptor antagonist in participants on maintenance hemodialysis (study B), to evaluate if IL-1 blockade had improved the anti-inflammatory activity (IL-6, TNF-α, and Nod-like receptor protein 3), antioxidant function (superoxide production), and net cholesterol efflux capacity of HDL. HDL function was measured using LPS-stimulated THP-1 macrophages or peritoneal macrophages of apoE-deficient mice exposed to the apoB-depleted, HDL-containing fraction obtained from the plasma of the study participants, collected before and after the interventions to block IL-1 effects. Analysis of covariance was used for between group comparisons. RESULTS The mean age of the participants was 60±13 years, 72% (n=33) were men, and 39% (n=18) were black. There were 32 CKD (16 IL-1 trap and 16 placebo) and 14 maintenance hemodialysis (7 IL-1 receptor antagonist and 7 placebo) participants. Compared with placebo, IL-1 inhibition, in study A and B reduced cellular expression of TNF-α by 15% (P=0.05) and 64% (P=0.02), IL-6 by 38% (P=0.004) and 56% (P=0.08), and Nod-like receptor protein 3 by 16% (P=0.01) and 25% (P=0.02), respectively. The intervention blunted superoxide production in the treated arm compared with placebo, with the values being higher by 17% in the placebo arm in study A (P<0.001) and 12% in the placebo arm in study B (P=0.004). Net cholesterol efflux capacity was not affected by either intervention. CONCLUSIONS IL-1 blockade improves the anti-inflammatory and antioxidative properties of the HDL-containing fraction of plasma in patients with stages 3-5 CKD, including those on maintenance hemodialysis.
Collapse
Affiliation(s)
- Adriana M Hung
- Division of Nephrology, Tennessee Valley Healthcare System, Nashville, Tennessee; .,Department of Medicine and
| | - Yohei Tsuchida
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Kristen L Nowak
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | | | - Michel Chonchol
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | | | | | | | | | | | | | - T Alp Ikizler
- Division of Nephrology, Tennessee Valley Healthcare System, Nashville, Tennessee.,Department of Medicine and
| | - Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee; and
| |
Collapse
|
10
|
Mantovani A, Dinarello CA, Molgora M, Garlanda C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity 2019; 50:778-795. [PMID: 30995499 PMCID: PMC7174020 DOI: 10.1016/j.immuni.2019.03.012] [Citation(s) in RCA: 690] [Impact Index Per Article: 115.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 02/06/2023]
Abstract
Forty years after its naming, interleukin-1 (IL-1) is experiencing a renaissance brought on by the growing understanding of its context-dependent roles and advances in the clinic. Recent studies have identified important roles for members of the IL-1 family-IL-18, IL-33, IL-36, IL-37, and IL-38-in inflammation and immunity. Here, we review the complex functions of IL-1 family members in the orchestration of innate and adaptive immune responses and their diversity and plasticity. We discuss the varied roles of IL-1 family members in immune homeostasis and their contribution to pathologies, including autoimmunity and auto-inflammation, dysmetabolism, cardiovascular disorders, and cancer. The trans-disease therapeutic activity of anti-IL-1 strategies argues for immunity and inflammation as a metanarrative of modern medicine.
Collapse
Affiliation(s)
- Alberto Mantovani
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy; Humanitas University, via Rita Levi Montalcini, 20090 Pieve Emanuele Milan, Italy; William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; Department of Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Martina Molgora
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy; Humanitas University, via Rita Levi Montalcini, 20090 Pieve Emanuele Milan, Italy.
| |
Collapse
|
11
|
Abstract
Interleukin-1 (IL-1) is the prototypical pro-inflammatory cytokine that occupies an apical place in the inflammatory cascade and also modulates cardiac function, functioning as a soluble cardiodepressant factor. Preclinical research over the past 4 decades has shown that blocking IL-1 processing or activity favorably affects cardiomyocyte survival and cardiac function in experimental animal models, paving the way for clinical studies in patients with heart disease. The promising results of phase II clinical trials of IL-1 blockade in patients with acute myocardial infarction and heart failure have been followed by a successful phase III trial in patients with prior acute myocardial infarction. Three IL-1 blockers with different mechanism of action are currently available for clinical use, although currently none have an indication for heart disease. We herein review the bench-to-bedside clinical translation of IL-1 targeting strategies and discuss the potential use of IL-1 blockade in patients with heart disease.
Collapse
|
12
|
Abstract
The heart is extremely sensitive to ischaemic injury. During an acute myocardial infarction (AMI) event, the injury is initially caused by reduced blood supply to the tissues, which is then further exacerbated by an intense and highly specific inflammatory response that occurs during reperfusion. Numerous studies have highlighted the central role of the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in this process. The inflammasome, an integral part of the innate immune system, is a macromolecular protein complex that finely regulates the activation of caspase 1 and the production and secretion of powerful pro-inflammatory cytokines such as IL-1β and IL-18. In this Review, we summarize evidence supporting the therapeutic value of NLRP3 inflammasome-targeted strategies in experimental models, and the data supporting the role of the NLRP3 inflammasome in AMI and its consequences on adverse cardiac remodelling, cytokine-mediated systolic dysfunction, and heart failure.
Collapse
Affiliation(s)
- Stefano Toldo
- Pauley Heart Center, Sanger Hall, 1201 East Marshall Street, Richmond, Virginia 23298, USA.,VCU Johnson Center for Critical Care and Pulmonary Research, Molecular Medicine Research Building, 1220 East Broad Street, Richmond, Virginia 23298, USA.,Division of Cardiothoracic Surgery, Sanger Hall, 1201 East Marshall Street, Richmond, Virginia 23298, USA
| | - Antonio Abbate
- Pauley Heart Center, Sanger Hall, 1201 East Marshall Street, Richmond, Virginia 23298, USA.,VCU Johnson Center for Critical Care and Pulmonary Research, Molecular Medicine Research Building, 1220 East Broad Street, Richmond, Virginia 23298, USA
| |
Collapse
|
13
|
Libby P. Interleukin-1 Beta as a Target for Atherosclerosis Therapy: Biological Basis of CANTOS and Beyond. J Am Coll Cardiol 2017; 70:2278-2289. [PMID: 29073957 DOI: 10.1016/j.jacc.2017.09.028] [Citation(s) in RCA: 457] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/07/2017] [Accepted: 09/10/2017] [Indexed: 02/07/2023]
Abstract
Inflammatory pathways drive atherogenesis and link conventional risk factors to atherosclerosis and its complications. One inflammatory mediator has come to the fore as a therapeutic target in cardiovascular disease. The experimental and clinical evidence reviewed here support interleukin-1 beta (IL-1β) as both a local vascular and systemic contributor in this regard. Intrinsic vascular wall cells and lesional leukocytes alike can produce this cytokine. Local stimuli in the plaque favor the generation of active IL-1β through the action of a molecular assembly known as the inflammasome. Clinically applicable interventions that interfere with IL-1 action can improve cardiovascular outcomes, ushering in a new era of anti-inflammatory therapies for atherosclerosis. The translational path described here illustrates how advances in basic vascular biology may transform therapy. Biomarker-directed application of anti-inflammatory interventions promises to help us achieve a more precise and personalized allocation of therapy for our cardiovascular patients.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
14
|
The IL-1RI Co-Receptor TILRR ( FREM1 Isoform 2) Controls Aberrant Inflammatory Responses and Development of Vascular Disease. JACC Basic Transl Sci 2017; 2:398-414. [PMID: 28920098 PMCID: PMC5582195 DOI: 10.1016/j.jacbts.2017.03.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/21/2017] [Accepted: 03/28/2017] [Indexed: 02/05/2023]
Abstract
The IL-1RI co-receptor, TILRR, is a potent amplifier of IL-1–induced responses. Blocking TILRR inhibits IL-1 receptor function and activation of inflammatory genes. TILRR expression is high in atherosclerotic lesions but low in healthy tissue, allowing distinct inhibition at sites of inflammation. Genetic deletion of TILRR and antibody blocking of TILRR function reduce plaque development and progression of atherosclerosis. Lesions exhibit low levels of macrophages and increased levels of smooth muscle cells and collagen, characteristics of stable plaques.
Expression of the interleukin-1 receptor type I (IL-1RI) co-receptor Toll-like and interleukin-1 receptor regulator (TILRR) is significantly increased in blood monocytes following myocardial infarction and in the atherosclerotic plaque, whereas levels in healthy tissue are low. TILRR association with IL-1RI at these sites causes aberrant activation of inflammatory genes, which underlie progression of cardiovascular disease. The authors show that genetic deletion of TILRR or antibody blocking of TILRR function reduces development of atherosclerotic plaques. Lesions exhibit decreased levels of monocytes, with increases in collagen and smooth muscle cells, characteristic features of stable plaques. The results suggest that TILRR may constitute a rational target for site- and signal-specific inhibition of vascular disease.
Collapse
Key Words
- ApoE, apolipoprotein E
- DK, double knockout
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- IL, interleukin
- IL-1RI
- IL-1RI, interleukin-1 receptor type I
- IgG, immunoglobulin G
- IκBα, inhibitor kappa B alpha
- KO, knockout
- LDLR–/–, low-density lipoprotein receptor–/–
- LPS, lipopolysaccharide
- NF-κB
- NF-κB, nuclear factor-kappa B
- NSTEMI, non–ST-segment elevation myocardial infarction
- PBS, phosphate-buffered saline
- PCR, polymerase chain reaction
- SDS, sodium dodecyl sulfate
- STEMI, ST-segment elevation myocardial infarction
- TILRR
- TILRR, toll-like and interleukin-1 receptor regulator
- heparan sulfate proteoglycan
- iBALT, inducible bronchus-associated lymphoid tissue
- interleukin-1 receptor
- qPCR, quantitative polymerase chain reaction
Collapse
|
15
|
Ridker PM. From C-Reactive Protein to Interleukin-6 to Interleukin-1: Moving Upstream To Identify Novel Targets for Atheroprotection. Circ Res 2016; 118:145-56. [PMID: 26837745 PMCID: PMC4793711 DOI: 10.1161/circresaha.115.306656] [Citation(s) in RCA: 649] [Impact Index Per Article: 72.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/01/2015] [Indexed: 12/23/2022]
Abstract
Plasma levels of the inflammatory biomarker high-sensitivity C-reactive protein (hsCRP) predict vascular risk with an effect estimate as large as that of total or high-density lipoprotein cholesterol. Further, randomized trial data addressing hsCRP have been central to understanding the anti-inflammatory effects of statin therapy and have consistently demonstrated on-treatment hsCRP levels to be as powerful a predictor of residual cardiovascular risk as on-treatment levels of low-density lipoprotein cholesterol. Yet, although hsCRP is clinically useful as a biomarker for risk prediction, most mechanistic studies suggest that CRP itself is unlikely to be a target for intervention. Moving upstream in the inflammatory cascade from CRP to interleukin (IL)-6 to IL-1 provides novel therapeutic opportunities for atheroprotection that focus on the central IL-6 signaling system and ultimately on inhibition of the IL-1β-producing NOD-like receptor family pyrin domain containing 3 inflammasome. Cholesterol crystals, neutrophil extracellular traps, atheroprone flow, and local tissue hypoxia activate the NOD-like receptor family pyrin domain containing 3 inflammasome. As such, a unifying concept of hsCRP as a downstream surrogate biomarker for upstream IL-1β activity has emerged. From a therapeutic perspective, small ischemia studies show reductions in acute-phase hsCRP production with the IL-1 receptor antagonist anakinra and the IL-6 receptor blocker tocilizumab. A phase IIb study conducted among diabetic patients at high vascular risk indicates that canakinumab, a human monoclonal antibody that targets IL-1β, markedly reduces plasma levels of IL-6, hsCRP, and fibrinogen with little change in atherogenic lipids. Canakinumab in now being tested as a method to prevent recurrent cardiovascular events in a randomized trial of 10 065 post-myocardial infarction patients with elevated hsCRP that is fully enrolled and due to complete in 2017. Clinical trials using alternative anti-inflammatory agents active against the CRP/IL-6/IL-1 axis, including low-dose methotrexate and colchicine, are being explored. If successful, these trials will close the loop on the inflammatory hypothesis of atherosclerosis and serve as examples of how fundamental biologic principles can be translated into personalized medical practice.
Collapse
Affiliation(s)
- Paul M Ridker
- From the Center for Cardiovascular Disease Prevention, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
16
|
Affiliation(s)
- Antonio Abbate
- Virginia Commonwealth University, Richmond, VA 23298, USA.
| | | | | |
Collapse
|
17
|
Mezzaroma E, Mikkelsen RB, Toldo S, Mauro AG, Sharma K, Marchetti C, Alam A, Van Tassell BW, Gewirtz DA, Abbate A. Role of Interleukin-1 in Radiation-Induced Cardiomyopathy. Mol Med 2015; 21:210-8. [PMID: 25822795 DOI: 10.2119/molmed.2014.00243] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/25/2015] [Indexed: 01/14/2023] Open
Abstract
Thoracic X-ray therapy (XRT), used in cancer treatment, is associated with increased risk of heart failure. XRT-mediated injury to the heart induces an inflammatory response leading to cardiomyopathy. The aim of this study was to determine the role of interleukin (IL)-1 in response to XRT injury to the heart and on the cardiomyopathy development in the mouse. Female mice with genetic deletion of the IL-1 receptor type I (IL-1R1 knockout mice [IL-1R1 KO]) and treatment with recombinant human IL-1 receptor antagonist anakinra, 10 mg/kg twice daily for 7 d, were used as independent approaches to determine the role of IL-1. Wild-type (wt) or IL-1R1 KO mice were treated with a single session of XRT (20 or 14 gray [Gy]). Echocardiography (before and after isoproterenol challenge) and left ventricular (LV) catheterization were performed to evaluate changes in LV dimensions and function. Masson's trichrome was used to assess myocardial fibrosis and pericardial thickening. After 20 Gy, the contractile reserve was impaired in wt mice at d 3, and the LV ejection fraction (EF) was reduced after 4 months when compared with sham-XRT. IL-1R1 KO mice had preserved contractile reserve at 3 d and 4 months and LVEF at 4 months after XRT. Anakinra treatment for 1 d before and 7 d after XRT prevented the impairment in contractile reserve. A significant increase in LV end-diastolic pressure, associated with increased myocardial interstitial fibrosis and pericardial thickening, was observed in wt mice, as well as in IL-1R1 KO-or anakinra-treated mice. In conclusion, induction of IL-1 by XRT mediates the development of some, such as the contractile impairment, but not all aspects of the XRT-induced cardiomyopathy, such as myocardial fibrosis or pericardial thickening.
Collapse
Affiliation(s)
- Eleonora Mezzaroma
- Virginia Commonwealth University (VCU) Pauley Heart Center, Richmond, Virginia, United States of America.,VCU Victoria Johnson Center, Richmond, Virginia, United States of America.,School of Pharmacy, VCU, Richmond, Virginia, United States of America
| | - Ross B Mikkelsen
- Radiation Oncology, Massey Cancer Center, VCU, Richmond, Virginia, United States of America
| | - Stefano Toldo
- Virginia Commonwealth University (VCU) Pauley Heart Center, Richmond, Virginia, United States of America.,VCU Victoria Johnson Center, Richmond, Virginia, United States of America
| | - Adolfo G Mauro
- Virginia Commonwealth University (VCU) Pauley Heart Center, Richmond, Virginia, United States of America.,VCU Victoria Johnson Center, Richmond, Virginia, United States of America
| | - Khushboo Sharma
- Pharmacology and Toxicology, Massey Cancer Center, VCU, Richmond, Virginia, United States of America
| | - Carlo Marchetti
- Virginia Commonwealth University (VCU) Pauley Heart Center, Richmond, Virginia, United States of America.,VCU Victoria Johnson Center, Richmond, Virginia, United States of America
| | - Asim Alam
- Radiation Oncology, Massey Cancer Center, VCU, Richmond, Virginia, United States of America
| | - Benjamin W Van Tassell
- Virginia Commonwealth University (VCU) Pauley Heart Center, Richmond, Virginia, United States of America.,VCU Victoria Johnson Center, Richmond, Virginia, United States of America.,School of Pharmacy, VCU, Richmond, Virginia, United States of America
| | - David A Gewirtz
- Pharmacology and Toxicology, Massey Cancer Center, VCU, Richmond, Virginia, United States of America
| | - Antonio Abbate
- Virginia Commonwealth University (VCU) Pauley Heart Center, Richmond, Virginia, United States of America.,VCU Victoria Johnson Center, Richmond, Virginia, United States of America
| |
Collapse
|
18
|
Luscher TF. Acute coronary syndromes and coronary intervention. Eur Heart J 2015; 36:323-4. [DOI: 10.1093/eurheartj/ehu508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
19
|
|