1
|
Meier DT, de Paula Souza J, Donath MY. Targeting the NLRP3 inflammasome-IL-1β pathway in type 2 diabetes and obesity. Diabetologia 2024:10.1007/s00125-024-06306-1. [PMID: 39496966 DOI: 10.1007/s00125-024-06306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/30/2024] [Indexed: 11/06/2024]
Abstract
Increased activity of the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome-IL-1β pathway is observed in obesity and contributes to the development of type 2 diabetes and its complications. In this review, we describe the pathological activation of IL-1β by metabolic stress, ageing and the microbiome and present data on the role of IL-1β in metabolism. We explore the physiological role of the IL-1β pathway in insulin secretion and the relationship between circulating levels of IL-1β and the development of diabetes and associated diseases. We highlight the paradoxical nature of IL-1β as both a friend and a foe in glucose regulation and provide details on clinical translation, including the glucose-lowering effects of IL-1 antagonism and its impact on disease modification. We also discuss the potential role of IL-1β in obesity, Alzheimer's disease, fatigue, gonadal dysfunction and related disorders such as rheumatoid arthritis and gout. Finally, we address the safety of NLRP3 inhibition and IL-1 antagonists and the prospect of using this therapeutic approach for the treatment of type 2 diabetes and its comorbidities.
Collapse
Affiliation(s)
- Daniel T Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Joyce de Paula Souza
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
2
|
Zubirán R, Neufeld EB, Dasseux A, Remaley AT, Sorokin AV. Recent Advances in Targeted Management of Inflammation In Atherosclerosis: A Narrative Review. Cardiol Ther 2024; 13:465-491. [PMID: 39031302 PMCID: PMC11333429 DOI: 10.1007/s40119-024-00376-3] [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: 02/29/2024] [Accepted: 06/26/2024] [Indexed: 07/22/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of morbidity and mortality despite effective low-density lipoprotein cholesterol-targeted therapies. This review explores the crucial role of inflammation in the residual risk of ASCVD, emphasizing its impact on atherosclerosis progression and plaque stability. Evidence suggests that high-sensitivity C-reactive protein (hsCRP), and potentially other inflammatory biomarkers, can be used to identify the inflammatory residual ASCVD risk phenotype and may serve as future targets for the development of more efficacious therapeutic approaches. We review the biological basis for the association of inflammation with ASCVD, propose new therapeutic strategies for the use of inflammation-targeted treatments, and discuss current challenges in the implementation of this new treatment paradigm for ASCVD.
Collapse
Affiliation(s)
- Rafael Zubirán
- Lipoprotein Metabolism Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Edward B Neufeld
- Lipoprotein Metabolism Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amaury Dasseux
- Lipoprotein Metabolism Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander V Sorokin
- Lipoprotein Metabolism Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
- Section of Inflammation and Cardiometabolic Diseases, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
- Section of Lipoprotein Metabolism, Clinical Research Center, National Heart, Lung and Blood Institute, 9000 Rockville Pike, Bldg 10, Room 5-5150, Bethesda, MD, 20892, USA.
| |
Collapse
|
3
|
Zhang D, Li Y, Pan J, Zheng Y, Xu X. Copper homeostasis and cuproptosis in radiation-induced injury. Biomed Pharmacother 2024; 178:117150. [PMID: 39047417 DOI: 10.1016/j.biopha.2024.117150] [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: 05/01/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Radiation therapy for cancer treatment brings about a series of radiation injuries to normal tissues. In recent years, the discovery of copper-regulated cell death, cuproptosis, a novel form of programmed cell death, has attracted widespread attention and exploration in various biological functions and pathological mechanisms of copper metabolism and cuproptosis. Understanding its role in the process of radiation injury may open up new avenues and directions for exploration in radiation biology and radiation oncology, thereby improving tumor response and mitigating adverse reactions to radiotherapy. This review provides an overview of copper metabolism, the characteristics of cuproptosis, and their potential regulatory mechanisms in radiation injury.
Collapse
Affiliation(s)
- Daoming Zhang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuan Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jinghui Pan
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yongfa Zheng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Ximing Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
4
|
Welt FGP, Batchelor W, Spears JR, Penna C, Pagliaro P, Ibanez B, Drakos SG, Dangas G, Kapur NK. Reperfusion Injury in Patients With Acute Myocardial Infarction: JACC Scientific Statement. J Am Coll Cardiol 2024; 83:2196-2213. [PMID: 38811097 DOI: 10.1016/j.jacc.2024.02.056] [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: 12/07/2023] [Revised: 02/15/2024] [Accepted: 02/26/2024] [Indexed: 05/31/2024]
Abstract
Despite impressive improvements in the care of patients with ST-segment elevation myocardial infarction, mortality remains high. Reperfusion is necessary for myocardial salvage, but the abrupt return of flow sets off a cascade of injurious processes that can lead to further necrosis. This has been termed myocardial ischemia-reperfusion injury and is the subject of this review. The pathologic and molecular bases for myocardial ischemia-reperfusion injury are increasingly understood and include injury from reactive oxygen species, inflammation, calcium overload, endothelial dysfunction, and impaired microvascular flow. A variety of pharmacologic strategies have been developed that have worked well in preclinical models and some have shown promise in the clinical setting. In addition, there are newer mechanical approaches including mechanical unloading of the heart prior to reperfusion that are in current clinical trials.
Collapse
Affiliation(s)
- Frederick G P Welt
- Department of Medicine, Division of Cardiovascular Medicine, University of Utah Hospital, Salt Lake City, Utah, USA.
| | | | - J Richard Spears
- Department of Cardiovascular Medicine, Beaumont Systems, Royal Oak, Michigan, USA
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Madrid, Spain; Department of Cardiology, Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Stavros G Drakos
- Department of Medicine, Division of Cardiovascular Medicine, University of Utah Hospital, Salt Lake City, Utah, USA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | - George Dangas
- Division of Cardiology, Mount Sinai Health System, New York, New York, USA
| | - Navin K Kapur
- The CardioVascular Center and Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Obare LM, Temu T, Mallal SA, Wanjalla CN. Inflammation in HIV and Its Impact on Atherosclerotic Cardiovascular Disease. Circ Res 2024; 134:1515-1545. [PMID: 38781301 PMCID: PMC11122788 DOI: 10.1161/circresaha.124.323891] [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] [Indexed: 05/25/2024]
Abstract
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8+ and CD4+ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| | - Tecla Temu
- Department of Pathology, Harvard Medical School, Boston, MA (T.T.)
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia (S.A.M.)
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Corna G, Golino M, Talasaz AH, Moroni F, Del Buono MG, Damonte JI, Chiabrando JG, Mbualungu J, Trankle CR, Thomas GK, Markley R, Canada JM, Turlington J, Agatiello CR, VAN Tassell B, Abbate A. Response to interleukin-1 blockade with anakinra in women and men with ST-segment elevation myocardial infarction. Minerva Cardiol Angiol 2024; 72:67-75. [PMID: 37987681 DOI: 10.23736/s2724-5683.23.06439-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
BACKGROUND Interleukin-1 blockade with anakinra reduces high-sensitivity C-reactive protein (hsCRP) levels and prevents heart failure (HF) events after ST-segment myocardial infarction (STEMI). Sex-based differences in STEMI patients have been reported, but no data are available regarding response to anakinra. METHODS We analyzed the systemic inflammation and composite end-point of new-onset HF or death in women and men with STEMI treated with anakinra from three different Virginia Commonwealth University Anakinra Response Trial (VCUART) randomized clinical trials. RESULTS We analyzed 139 patients, 29 (21%) were women while 110 (79%) were men. Baseline hsCRP was higher in women compared to men (8.9 [5.2-13.5] vs. 4.2 [2.1-7.7] mg/L, P<0.001). Eighty-four patients were treated with anakinra (22 [75%] women and 62 [56%] men). The area under the curve of hsCRP (hsCRP-AUC) after 14 days was numerically lower in patients receiving anakinra versus placebo both in men (86 [37-130] vs. 223 [119-374] mg day/L) and in women (73 [46-313] vs. 242 [102-988] mg day/L) (P<0.001 for multiple groups, P for interaction 0.22). The incidence of the composite endpoint was also numerically lower in the anakinra group compared to placebo, both in men (4 [6.4%] vs. 14 [29.1%]) and in women (3 [13.6%] vs. 2 [28.5%]) (P=0.019 for multiple groups, P for interaction 0.44). There were no statistically significant differences between women and men in hsCRP-AUC and death or HF events when comparing separately the anakinra and placebo groups (all P>0.05). CONCLUSIONS Women were underrepresented in the VCUART trials, they appeared to have higher hsCRP levels at time of presentation, yet to benefit similar to men by treatment with anakinra in STEMI.
Collapse
Affiliation(s)
- Giuliana Corna
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Interventional Cardiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Michele Golino
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Azita H Talasaz
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Francesco Moroni
- Department of Internal Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Medicine, University of Milano-Bicocca, Milan, Italy
| | - Marco G Del Buono
- Department of Cardiovascular Medicine, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| | - Juan I Damonte
- Department of Interventional Cardiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Juan G Chiabrando
- Department of Interventional Cardiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - James Mbualungu
- Division of Cardiology, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Cory R Trankle
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Georgia K Thomas
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Roshanak Markley
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Justin M Canada
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Jeremy Turlington
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Carla R Agatiello
- Department of Interventional Cardiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Benjamin VAN Tassell
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- Division of Cardiology, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA -
| |
Collapse
|
9
|
Ter Mors B, Spieler V, Merino Asumendi E, Gantert B, Lühmann T, Meinel L. Bioresponsive Cytokine Delivery Responding to Matrix Metalloproteinases. ACS Biomater Sci Eng 2024; 10:29-37. [PMID: 37102329 DOI: 10.1021/acsbiomaterials.2c01320] [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] [Indexed: 04/28/2023]
Abstract
Cytokines are regulated in acute and chronic inflammation, including rheumatoid arthritis (RA) and myocardial infarction (MI). However, the dynamic windows within which cytokine activity/inhibition is desirable in RA and MI change timely and locally during the disease. Therefore, traditional, static delivery regimens are unlikely to meet the idiosyncrasy of these highly dynamic pathophysiological and individual processes. Responsive delivery systems and biomaterials, sensing surrogate markers of inflammation (i.e., matrix metalloproteinases - MMPs) and answering with drug release, may present drug activity at the right time, manner, and place. This article discusses MMPs as surrogate markers for disease activity in RA and MI to clock drug discharge to MMP concentration profiles from MMP-responsive drug delivery systems and biomaterials.
Collapse
Affiliation(s)
- Björn Ter Mors
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Valerie Spieler
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Eduardo Merino Asumendi
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Benedikt Gantert
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), 97080 Würzburg, Germany
| |
Collapse
|
10
|
Sazonova SI, Ilyushenkova JN, Syrkina AG, Trusov AA, Mochula OV, Mishkina AI, Ryabov VV. Potential utility of SPECT/CT with 99mTc-Tektrotyd for imaging of post-myocardial infarction inflammation. J Nucl Cardiol 2023; 30:2544-2555. [PMID: 37316747 DOI: 10.1007/s12350-023-03312-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/19/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND There is a need to develop methods for post-myocardial infarction (MI) inflammation monitoring. Scintigraphy with somatostatin receptor targeted radiotracers has potential in this field. The purpose was to study the association of 99mTc-Tektrotyd uptake intensity in MI area with heart contractility indices over 6-month follow-up. METHODS Fourteen patients with acute ST-segment elevation anterior MI (STEMI) were examined with 99mTc-Tektrotyd SPECT/CT, myocardial perfusion scintigraphy (MPS) at rest, cardiac magnetic resonance imaging (cMRI) and transthoracic echocardiography (TTE). Scintigraphic results were compared with 6-month TTE indices. RESULTS On the 7th day after a MI onset, cardiac 99mTc-Tektrotyd uptake was found in 7 of 14 patients. Median of 99mTc-Tektrotyd SUVmax was 1.59 (1.38; 2.83), the summed rest score (SRS) was 11 (5; 18), infarct size (by cMRI)-13.15 (3.3; 32.2) %. 99mTc-Tektrotyd SUVmax strongly correlated with 6-month heart contractility indices (r = 0.81, P < 0.05 for the end diastolic volume; r = 0.61 P < 0.05 for Δ end diastolic volume), with SRS (r = 0.85, P < 0.05) and infarct size (by cMRI) (r = 0.79, P < 0.05). CONCLUSION The intensity (SUVmax) of 99mTc-Tektrotyd uptake in the area of recent MI directly depends on the size of ischemic myocardial injury and correlates with changes of heart contractility indexes over the 6 month follow-up.
Collapse
Affiliation(s)
- Svetlana I Sazonova
- Nuclear Medicine Department, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Kievskaya 111a, 634012, Russian Federation, Tomsk.
| | - Julia N Ilyushenkova
- Nuclear Medicine Department, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Kievskaya 111a, 634012, Russian Federation, Tomsk
| | - Anna G Syrkina
- Cardiac Emergency Department, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russian Federation
| | - Andrey A Trusov
- Cardiac Emergency Department, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russian Federation
| | - Olga V Mochula
- Magnetic-Resonance Tomography Department, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russian Federation
| | - Anna I Mishkina
- Nuclear Medicine Department, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Kievskaya 111a, 634012, Russian Federation, Tomsk
| | - Vyacheslav V Ryabov
- Cardiac Emergency Department, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russian Federation
| |
Collapse
|
11
|
Francisco J, Del Re DP. Inflammation in Myocardial Ischemia/Reperfusion Injury: Underlying Mechanisms and Therapeutic Potential. Antioxidants (Basel) 2023; 12:1944. [PMID: 38001797 PMCID: PMC10669026 DOI: 10.3390/antiox12111944] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Acute myocardial infarction (MI) occurs when blood flow to the myocardium is restricted, leading to cardiac damage and massive loss of viable cardiomyocytes. Timely restoration of coronary flow is considered the gold standard treatment for MI patients and limits infarct size; however, this intervention, known as reperfusion, initiates a complex pathological process that somewhat paradoxically also contributes to cardiac injury. Despite being a sterile environment, ischemia/reperfusion (I/R) injury triggers inflammation, which contributes to infarct expansion and subsequent cardiac remodeling and wound healing. The immune response is comprised of subsets of both myeloid and lymphoid-derived cells that act in concert to modulate the pathogenesis and resolution of I/R injury. Multiple mechanisms, including altered metabolic status, regulate immune cell activation and function in the setting of acute MI, yet our understanding remains incomplete. While numerous studies demonstrated cardiac benefit following strategies that target inflammation in preclinical models, therapeutic attempts to mitigate I/R injury in patients were less successful. Therefore, further investigation leveraging emerging technologies is needed to better characterize this intricate inflammatory response and elucidate its influence on cardiac injury and the progression to heart failure.
Collapse
Affiliation(s)
| | - Dominic P. Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
12
|
Talasaz AH, Sculthorpe R, Pak M, Lipinski M, Roberts C, Markley R, Trankle CR, Canada JM, Wohlford GF, Golino M, Dixon D, Van Tassell BW, Abbate A. Comparison of Safety and Biological Efficacy of Anakinra (Kineret) Dispensed in Polycarbonate Plastic versus Borosilicate Glass Syringes: A Patient-Level Analysis of VCUART2 and VCUART3 Clinical Trials. J Pharmacol Exp Ther 2023; 386:138-142. [PMID: 36868827 PMCID: PMC10353132 DOI: 10.1124/jpet.122.001404] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/09/2023] [Accepted: 02/16/2023] [Indexed: 03/05/2023] Open
Abstract
Anakinra is a recombinant human interleukin-1 receptor antagonist approved for the treatment of inflammatory diseases. Kineret is available as a solution prepared in a borosilicate glass syringe. For implementing a placebo-controlled double-blind randomized clinical trial, anakinra is commonly transferred into plastic syringes. However, there is limited data on anakinra's stability in polycarbonate syringes. We described the results of our previous studies on the use of anakinra in glass (VCUART3) versus plastic syringes (VCUART2) compared with placebo. These studies were conducted in patients with ST-segment elevation myocardial infarction (STEMI), and we assessed the anti-inflammatory effects of anakinra versus placebo by comparing the area under the curve for high-sensitivity cardiac reactive protein (AUC-CRP) levels during the first 14 days of STEMI, its clinical effects on heart failure (HF) hospitalization, cardiovascular death, or new diagnosis of HF as well as adverse events profile between groups. The levels of AUC-CRP were 75 (50-255 mg·day/l) for anakinra in plastic syringes versus 255 (116-592 mg·day/l) in placebo and 60 (24-139 mg·day/l) and 86 (43-123 mg·day/l) for anakinra once and twice daily in glass syringes, respectively, compared with placebo 214 (131-394 mg·day/l). The rate of adverse events was also comparable between groups. There were no differences in the rate of HF hospitalization or cardiovascular death in patients who received anakinra in plastic or glass syringes. Fewer cases of new-onset heart failure occurred in patients receiving anakinra in plastic or glass syringes compared with placebo. Anakinra stored in plastic (polycarbonate) syringes provides comparable biologic and clinical effect to glass (borosilicate) syringes. SIGNIFICANCE STATEMENT: Anakinra (Kineret) 100 mg administered subcutaneously in patients with ST-segment elevation myocardial infarction (STEMI) for a duration of up to 14 days appears to have comparable safety and biological efficacy signals when delivered in prefilled glass or transferred into plastic polycarbonate syringes. This may have important implications for the feasibility of designing clinical trials in STEMI and other clinical conditions.
Collapse
Affiliation(s)
- Azita H Talasaz
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Robin Sculthorpe
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Mary Pak
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Michael Lipinski
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Charlotte Roberts
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Roshanak Markley
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Cory R Trankle
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Justin M Canada
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - George F Wohlford
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Michele Golino
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Dave Dixon
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Benjamin W Van Tassell
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Antonio Abbate
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| |
Collapse
|
13
|
Dimitroglou Y, Aggeli C, Theofilis P, Tsioufis P, Oikonomou E, Chasikidis C, Tsioufis K, Tousoulis D. Novel Anti-Inflammatory Therapies in Coronary Artery Disease and Acute Coronary Syndromes. Life (Basel) 2023; 13:1669. [PMID: 37629526 PMCID: PMC10455741 DOI: 10.3390/life13081669] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
Evidence suggests that inflammation plays an important role in atherosclerosis and the consequent clinical presentation, including stable coronary artery disease (CAD) and acute coronary syndromes (ACS). The most essential elements are cytokines, proteins with hormone-like properties that are produced by the immune cells, endothelial cells, platelets, fibroblasts, and some stromal cells. Interleukins (IL-1β and IL-6), chemokines, interferon-γ (IFN-γ), and tumor necrosis factor-alpha (TNF-α) are the cytokines commonly associated with endothelial dysfunction, vascular inflammation, and atherosclerosis. These molecules can be targeted by commonly used therapeutic substances or selective molecules that exert targeted anti-inflammatory actions. The most significant anti-inflammatory therapies are aspirin, statins, colchicine, IL-1β inhibitors, and IL-6 inhibitors, along with novel therapies such as TNF-α inhibitors and IL-1 receptor antagonists. Aspirin and statins are well-established therapies for atherosclerosis and CAD and their pleiotropic and anti-inflammatory actions contribute to their efficacy and favorable profile. Colchicine may also be considered in high-risk patients if recurrent ACS episodes occur when on optimal medical therapy according to the most recent guidelines. Recent randomized studies have also shown that therapies specifically targeting inflammatory interleukins and inflammation can reduce the risk for cardiovascular events, but these therapies are yet to be fully implemented in clinical practice. Preclinical research is also intense, targeting various inflammatory mediators that are believed to be implicated in CAD, namely repeated transfers of the soluble mutant of IFN-γ receptors, NLRP3 inflammasome inhibitors, IL-10 delivery by nanocarriers, chemokine modulatory treatments, and reacting oxygen species (ROS) targeting nanoparticles. Such approaches, although intriguing and promising, ought to be tested in clinical settings before safe conclusions can be drawn. Although the link between inflammation and atherosclerosis is significant, further studies are needed in order to elucidate this association and improve outcomes in patients with CAD.
Collapse
Affiliation(s)
- Yannis Dimitroglou
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Constantina Aggeli
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Panagiotis Theofilis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Panagiotis Tsioufis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Evangelos Oikonomou
- Third Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece;
| | - Christos Chasikidis
- Department of Cardiology, General Hospital of Corinth, 20100 Corinth, Greece;
| | - Konstantinos Tsioufis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Dimitris Tousoulis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| |
Collapse
|
14
|
Alkhalil M, De Maria GL, Akbar N, Ruparelia N, Choudhury RP. Prospects for Precision Medicine in Acute Myocardial Infarction: Patient-Level Insights into Myocardial Injury and Repair. J Clin Med 2023; 12:4668. [PMID: 37510783 PMCID: PMC10380764 DOI: 10.3390/jcm12144668] [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: 06/07/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
The past decade has seen a marked expansion in the understanding of the pathobiology of acute myocardial infarction and the systemic inflammatory response that it elicits. At the same time, a portfolio of tools has emerged to characterise some of these processes in vivo. However, in clinical practice, key decision making still largely relies on assessment built around the timing of the onset of chest pain, features on electrocardiograms and measurements of plasma troponin. Better understanding the heterogeneity of myocardial injury and patient-level responses should provide new opportunities for diagnostic stratification to enable the delivery of more rational therapies. Characterisation of the myocardium using emerging imaging techniques such as the T1, T2 and T2* mapping techniques can provide enhanced assessments of myocardial statuses. Physiological measures, which include microcirculatory resistance and coronary flow reserve, have been shown to predict outcomes in AMI and can be used to inform treatment selection. Functionally informative blood biomarkers, including cellular transcriptomics; microRNAs; extracellular vesicle analyses and soluble markers, all give insights into the nature and timing of the innate immune response and its regulation in acute MI. The integration of these and other emerging tools will be key to developing a fuller understanding of the patient-level processes of myocardial injury and repair and should fuel new possibilities for rational therapeutic intervention.
Collapse
Affiliation(s)
- Mohammad Alkhalil
- Cardiothoracic Centre, Freeman Hospital, Newcastle-upon-Tyne NE7 7DN, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne NE1 7RU, UK
| | | | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Neil Ruparelia
- Cardiology Department, Hammersmith Hospital, Imperial College London, London W12 0HS, UK
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
15
|
Pagliaro P, Penna C. Inhibitors of NLRP3 Inflammasome in Ischemic Heart Disease: Focus on Functional and Redox Aspects. Antioxidants (Basel) 2023; 12:1396. [PMID: 37507935 PMCID: PMC10376505 DOI: 10.3390/antiox12071396] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is caused by several mechanisms, including the production of reactive oxygen species (ROS), altered cellular osmolarity, and inflammatory response. Calcium overload, altered oxygen levels, and mitochondrial ROS are also involved in these MIRI processes, resulting in the irreversible opening of the mitochondrial permeability transition pore (mPTP). These mechanisms and processes are associated with NLRP3 inflammasome priming and activation, which can also induce cell death by pyroptosis through the up-regulation of the caspase-1 pathway and IL-18 release. In addition, endothelial dysfunction, both in the presence and absence of MIRI, is also accompanied by altered oxygen levels, decreased nitric oxide production, and ROS overproduction, resulting in the expression of adhesion molecules and leukocyte infiltration in which the NLRP3 inflammasome plays a central role, thus contributing, through endothelial dysfunction, to the alteration of coronary flow, typical of ischemic heart disease. Given the intricate interrelationship between ROS and NLRP3, ROS inhibitors can reduce NLRP3 inflammasome activation, while NLRP3 inhibitors can reduce oxidative stress and inflammation. NLRP3 inhibitors have been intensively studied as anti-inflammatory agents in basic cardiovascular sciences. In this review, we analyze the interrelation between ROS and NLRP3 in ischemic heart disease and the effects of some NLRP3 inhibitors as possible therapeutic agents in this disease condition. All compounds considered in this review need larger studies to confirm their appropriate use in clinical scenarios as anti-ischemic drugs.
Collapse
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, Turin University, Orbassano, 10043 Turin, Italy
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, Turin University, Orbassano, 10043 Turin, Italy
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
16
|
Weber BN, Giles JT, Liao KP. Shared inflammatory pathways of rheumatoid arthritis and atherosclerotic cardiovascular disease. Nat Rev Rheumatol 2023; 19:417-428. [PMID: 37231248 PMCID: PMC10330911 DOI: 10.1038/s41584-023-00969-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2023] [Indexed: 05/27/2023]
Abstract
The association between chronic inflammation and increased risk of cardiovascular disease in rheumatoid arthritis (RA) is well established. In the general population, inflammation is an established independent risk factor for cardiovascular disease, and much interest is placed on controlling inflammation to reduce cardiovascular events. As inflammation encompasses numerous pathways, the development of targeted therapies in RA provides an opportunity to understand the downstream effect of inhibiting specific pathways on cardiovascular risk. Data from these studies can inform cardiovascular risk management in patients with RA, and in the general population. This Review focuses on pro-inflammatory pathways targeted by existing therapies in RA and with mechanistic data from the general population on cardiovascular risk. Specifically, the discussions include the IL-1, IL-6 and TNF pathways, as well as the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signalling pathway, and the role of these pathways in RA pathogenesis in the joint alongside the development of atherosclerotic cardiovascular disease. Overall, some robust data support inhibition of IL-1 and IL-6 in decreasing the risk of cardiovascular disease, with growing data supporting IL-6 inhibition in both patients with RA and the general population to reduce the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Brittany N Weber
- Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jon T Giles
- Columbia University, Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Katherine P Liao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA.
- Rheumatology Section, VA Boston Medical Center, Boston, MA, USA.
| |
Collapse
|
17
|
Alshoubaki YK, Lu YZ, Legrand JMD, Karami R, Fossat M, Salimova E, Julier Z, Martino MM. A superior extracellular matrix binding motif to enhance the regenerative activity and safety of therapeutic proteins. NPJ Regen Med 2023; 8:25. [PMID: 37217533 DOI: 10.1038/s41536-023-00297-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
Among therapeutic proteins, cytokines and growth factors have great potential for regenerative medicine applications. However, these molecules have encountered limited clinical success due to low effectiveness and major safety concerns, highlighting the need to develop better approaches that increase efficacy and safety. Promising approaches leverage how the extracellular matrix (ECM) controls the activity of these molecules during tissue healing. Using a protein motif screening strategy, we discovered that amphiregulin possesses an exceptionally strong binding motif for ECM components. We used this motif to confer the pro-regenerative therapeutics platelet-derived growth factor-BB (PDGF-BB) and interleukin-1 receptor antagonist (IL-1Ra) a very high affinity to the ECM. In mouse models, the approach considerably extended tissue retention of the engineered therapeutics and reduced leakage in the circulation. Prolonged retention and minimal systemic diffusion of engineered PDGF-BB abolished the tumour growth-promoting adverse effect that was observed with wild-type PDGF-BB. Moreover, engineered PDGF-BB was substantially more effective at promoting diabetic wound healing and regeneration after volumetric muscle loss, compared to wild-type PDGF-BB. Finally, while local or systemic delivery of wild-type IL-1Ra showed minor effects, intramyocardial delivery of engineered IL-1Ra enhanced cardiac repair after myocardial infarction by limiting cardiomyocyte death and fibrosis. This engineering strategy highlights the key importance of exploiting interactions between ECM and therapeutic proteins for developing effective and safer regenerative therapies.
Collapse
Affiliation(s)
- Yasmin K Alshoubaki
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Yen-Zhen Lu
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Julien M D Legrand
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Rezvan Karami
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Mathilde Fossat
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Ekaterina Salimova
- Monash Biomedical Imaging, Monash University, Clayton, VIC, 3800, Australia
| | - Ziad Julier
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia.
- Victorian Heart Institute, Monash University, Clayton, VIC, 3800, Australia.
- Laboratory of Host Defense, World Premier Institute Immunology Frontier Research Center, Osaka University, Osaka, 565-0871, Japan.
| |
Collapse
|
18
|
Jones D, Spirito A, Sartori S, Smith KF, Pivato CA, Chiarito M, Cao D, Nicolas J, Beerkens F, Edens M, Pileggi B, Sen A, Zhang Z, Vogel B, Sweeny J, Baber U, Dangas G, Sharma SK, Kini A, Mehran R. PROGNOSTIC VALUE OF HIGH-SENSITIVITY C-REATIVE PROTEIN AMONG CHRONIC KIDNEY DISEASE PATIENTS UNDERGOING PERCUTANEOUS CORONARY INTERVENTION. J Cardiol 2023:S0914-5087(23)00108-9. [PMID: 37187289 DOI: 10.1016/j.jjcc.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND Data on the prognostic value of high-sensitivity C-reactive protein (hs-CRP) levels in patients with chronic kidney disease (CKD) undergoing percutaneous coronary intervention (PCI) are limited. METHODS Patients undergoing PCI at a tertiary center from January 2012 to December 2019 were included. CKD was defined as a glomerular filtration rate (GFR) <60 mL/min/1.73m2 and elevated hs-CRP was defined as >3 mg/L. Acute myocardial infarction (MI), acute heart failure, neoplastic disease, patients undergoing hemodialysis, or hs-CRP >10 mg/L were exclusion criteria. The primary outcome was major adverse cardiac events (MACE), a composite of all-cause death, MI, and target vessel revascularization at 1-year after PCI. RESULTS Out of 12,410 patients, 3,029 (24.4%) had CKD. Elevated hs-CRP levels were found in 31.8% of CKD and 25.8% of non-CKD patients. At 1 year, MACE occurred in 87 (11.0%) CKD patients with elevated hs-CRP and 163 (9.5%) with low hs-CRP (adj. HR 1.26, 95% CI 0.94-1.68); among non-CKD patients, in 200 (10%) and 470 (8.1%), respectively (adj. HR 1.21, 95% CI 1.00-1.45). Hs-CRP was associated with an increased risk of all-cause death in both CKD (Adj. HR 1.92, 95% CI 1.07-3.44) and no-CKD patients (adj. HR 3.02, 95% CI 1.74-5.22). There was no interaction between hs-CRP and CKD. CONCLUSIONS Among patients undergoing PCI without acute MI, elevated hs-CRP values were not associated with a higher risk of MACE at 1 year, but with increased mortality hazards irrespective of the CKD status. The prognostic value of hs-CRP was consistent in patients with or without CKD.
Collapse
Affiliation(s)
- Davis Jones
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alessandro Spirito
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samantha Sartori
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenneth F Smith
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carlo Andrea Pivato
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele-Milan, Milan, Italy
| | - Mauro Chiarito
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Humanitas Research Hospital IRCCS, Rozzano-, Milan, Milan, Italy
| | - Davide Cao
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Johny Nicolas
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Frans Beerkens
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madison Edens
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brunna Pileggi
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cardiopneumonology, Heart Institute of the University of Sao Paulo, Sao Paulo, Brazil
| | - Ananya Sen
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhongjie Zhang
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Birgit Vogel
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph Sweeny
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Usman Baber
- Department of Cardiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - George Dangas
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samin K Sharma
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Annapoorna Kini
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roxana Mehran
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
19
|
Di Fusco SA, Maggioni AP, Scicchitano P, Zuin M, D’Elia E, Colivicchi F. Lipoprotein (a), Inflammation, and Atherosclerosis. J Clin Med 2023; 12:jcm12072529. [PMID: 37048611 PMCID: PMC10095203 DOI: 10.3390/jcm12072529] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Growing evidence has shown that high levels of lipoprotein (a) (Lp(a)) and chronic inflammation may be responsible for the residual risk of cardiovascular events in patients managed with an optimal evidence-based approach. Clinical studies have demonstrated a correlation between higher Lp(a) levels and several atherosclerotic diseases including ischemic heart disease, stroke, and degenerative calcific aortic stenosis. The threshold value of Lp(a) serum concentrations associated with a significantly increased cardiovascular risk is >125 nmol/L (50 mg/dL). Current available lipid-lowering drugs have modest-to-no impact on Lp(a) levels. Chronic inflammation is a further condition potentially implicated in residual cardiovascular risk. Consistent evidence has shown an increased risk of cardiovascular events in patients with high sensitivity C reactive protein (>2 mg/dL), an inflammation biomarker. A number of anti-inflammatory drugs have been investigated in patients with or at risk of cardiovascular disease. Of these, canakinumab and colchicine have been found to be associated with cardiovascular risk reduction. Ongoing research aimed at improving risk stratification on the basis of Lp(a) and vessel inflammation assessment may help refine patient management. Furthermore, the identification of these conditions as cardiovascular risk factors has led to increased investigation into diagnostic and therapeutic strategies targeting them in order to reduce atherosclerotic cardiovascular disease burden.
Collapse
|
20
|
Lugrin J, Parapanov R, Milano G, Cavin S, Debonneville A, Krueger T, Liaudet L. The systemic deletion of interleukin-1α reduces myocardial inflammation and attenuates ventricular remodeling in murine myocardial infarction. Sci Rep 2023; 13:4006. [PMID: 36899010 PMCID: PMC10006084 DOI: 10.1038/s41598-023-30662-4] [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] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Myocardial inflammation following myocardial infarction (MI) is crucial for proper myocardial healing, yet, dysregulated inflammation may promote adverse ventricular remodeling and heart failure. IL-1 signaling contributes to these processes, as shown by dampened inflammation by inhibition of IL-1β or the IL-1 receptor. In contrast, the potential role of IL-1α in these mechanisms has received much less attention. Previously described as a myocardial-derived alarmin, IL-1α may also act as a systemically released inflammatory cytokine. We therefore investigated the effect of IL-1α deficiency on post-MI inflammation and ventricular remodeling in a murine model of permanent coronary occlusion. In the first week post-MI, global IL-1α deficiency (IL-1α KO mice) led to decreased myocardial expression of IL-6, MCP-1, VCAM-1, hypertrophic and pro-fibrotic genes, and reduced infiltration with inflammatory monocytes. These early changes were associated with an attenuation of delayed left ventricle (LV) remodeling and systolic dysfunction after extensive MI. In contrast to systemic Il1a-KO, conditional cardiomyocyte deletion of Il1a (CmIl1a-KO) did not reduce delayed LV remodeling and systolic dysfunction. In conclusion, systemic Il1a-KO, but not Cml1a-KO, protects against adverse cardiac remodeling after MI due to permanent coronary occlusion. Hence, anti-IL-1α therapies could be useful to attenuate the detrimental consequences of post-MI myocardial inflammation.
Collapse
Affiliation(s)
- J Lugrin
- Service of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- Laboratoire de Chirurgie Thoracique, Centre des Laboratoires d'Epalinges, Chemin des Boveresses 155, 1066, Epalinges, Switzerland.
| | - R Parapanov
- Service of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - G Milano
- Department Coeur-Vaisseaux, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - S Cavin
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - A Debonneville
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - T Krueger
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - L Liaudet
- Service of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
21
|
Onódi Z, Koch S, Rubinstein J, Ferdinandy P, Varga ZV. Drug repurposing for cardiovascular diseases: New targets and indications for probenecid. Br J Pharmacol 2023; 180:685-700. [PMID: 36484549 DOI: 10.1111/bph.16001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/12/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
The available pharmacological options in the management of cardiovascular diseases such as ischaemic heart disease and subsequent heart failure are effective in slowing the progression of this condition. However, the long-term prognosis is still poor, raising the demand for new therapeutic strategies. Drug repurposing is a time- and cost-effective drug development strategy that offers approved and abandoned drugs a new chance for new indications. Recently, drugs used for the management of gout-related inflammation such as canakinumab or colchicine have been considered for drug repurposing in cardiovascular indications. The old uricosuric drug, probenecid, has been identified as a novel therapeutic option in the management of specific cardiac diseases as well. Probenecid can modulate myocardial contractility and vascular tone and exerts anti-inflammatory properties. The mechanisms behind these beneficial effects might be related inhibition of inflammasomes, and to modulation purinergic-pannexin-1 signalling and TRPV2 channels, which are recently identified molecular targets of probenecid. In this review, we provide an overview on repurposing probenecid for ischaemic heart disease and subsequent heart failure by summarizing the related experimental and clinical data and propose its potential repurposing to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Sheryl Koch
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| |
Collapse
|
22
|
González L, Rivera K, Andia ME, Martínez Rodriguez G. The IL-1 Family and Its Role in Atherosclerosis. Int J Mol Sci 2022; 24:17. [PMID: 36613465 PMCID: PMC9820551 DOI: 10.3390/ijms24010017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
The IL-1 superfamily of cytokines is a central regulator of immunity and inflammation. The family is composed of 11 cytokines (with agonist, antagonist, and anti-inflammatory properties) and 10 receptors, all tightly regulated through decoy receptor, receptor antagonists, and signaling inhibitors. Inflammation not only is an important physiological response against infection and injury but also plays a central role in atherosclerosis development. Several clinical association studies along with experimental studies have implicated the IL-1 superfamily of cytokines and its receptors in the pathogenesis of cardiovascular disease. Here, we summarize the key features of the IL-1 family, its role in immunity and disease, and how it helps shape the development of atherosclerosis.
Collapse
Affiliation(s)
- Leticia González
- Centro de Imágenes Biomédicas—Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
- Instituto Milenio de Ingeniería e Inteligencia Artificial Para la Salud, iHEALTH, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Katherine Rivera
- Centro de Imágenes Biomédicas—Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
- Programa de Doctorado en Ciencias Médicas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
| | - Marcelo E. Andia
- Centro de Imágenes Biomédicas—Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
- Instituto Milenio de Ingeniería e Inteligencia Artificial Para la Salud, iHEALTH, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Gonzalo Martínez Rodriguez
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
| |
Collapse
|
23
|
Tsioufis P, Theofilis P, Tsioufis K, Tousoulis D. The Impact of Cytokines in Coronary Atherosclerotic Plaque: Current Therapeutic Approaches. Int J Mol Sci 2022; 23:ijms232415937. [PMID: 36555579 PMCID: PMC9788180 DOI: 10.3390/ijms232415937] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Coronary atherosclerosis is a chronic pathological process that involves inflammation together with endothelial dysfunction and lipoprotein dysregulation. Experimental studies during the past decades have established the role of inflammatory cytokines in coronary artery disease, namely interleukins (ILs), tumor necrosis factor (TNF)-α, interferon-γ, and chemokines. Moreover, their value as biomarkers in disease development and progression further enhance the validity of this interaction. Recently, cytokine-targeted treatment approaches have emerged as potential tools in the management of atherosclerotic disease. IL-1β, based on the results of the CANTOS trial, remains the most validated option in reducing the residual cardiovascular risk. Along the same line, colchicine was also proven efficacious in preventing major adverse cardiovascular events in large clinical trials of patients with acute and chronic coronary syndrome. Other commercially available agents targeting IL-6 (tocilizumab), TNF-α (etanercept, adalimumab, infliximab), or IL-1 receptor antagonist (anakinra) have mostly been assessed in the setting of other inflammatory diseases and further testing in atherosclerosis is required. In the future, potential targeting of the NLRP3 inflammasome, anti-inflammatory IL-10, or atherogenic chemokines could represent appealing options, provided that patient safety is proven to be of no concern.
Collapse
|
24
|
Hepprich M, Mudry JM, Gregoriano C, Jornayvaz FR, Carballo S, Wojtusciszyn A, Bart PA, Chiche JD, Fischli S, Baumgartner T, Cavelti-Weder C, Braun DL, Günthard HF, Beuschlein F, Conen A, West E, Isenring E, Zechmann S, Bucklar G, Aubry Y, Dey L, Müller B, Hunziker P, Schütz P, Cattaneo M, Donath MY. Canakinumab in patients with COVID-19 and type 2 diabetes - A multicentre, randomised, double-blind, placebo-controlled trial. EClinicalMedicine 2022; 53:101649. [PMID: 36128334 PMCID: PMC9481336 DOI: 10.1016/j.eclinm.2022.101649] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 08/12/2022] [Accepted: 08/12/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Patients with type 2 diabetes and obesity have chronic activation of the innate immune system possibly contributing to the higher risk of hyperinflammatory response to SARS-CoV2 and severe COVID-19 observed in this population. We tested whether interleukin-1β (IL-1β) blockade using canakinumab improves clinical outcome. METHODS CanCovDia was a multicenter, randomised, double-blind, placebo-controlled trial to assess the efficacy of canakinumab plus standard-of-care compared with placebo plus standard-of-care in patients with type 2 diabetes and a BMI > 25 kg/m2 hospitalised with SARS-CoV2 infection in seven tertiary-hospitals in Switzerland. Patients were randomly assigned 1:1 to a single intravenous dose of canakinumab (body weight adapted dose of 450-750 mg) or placebo. Canakinumab and placebo were compared based on an unmatched win-ratio approach based on length of survival, ventilation, ICU stay and hospitalization at day 29. This study is registered with ClinicalTrials.gov, NCT04510493. FINDINGS Between October 17, 2020, and May 12, 2021, 116 patients were randomly assigned with 58 in each group. One participant dropped out in each group for the primary analysis. At the time of randomization, 85 patients (74·6 %) were treated with dexamethasone. The win-ratio of canakinumab vs placebo was 1·08 (95 % CI 0·69-1·69; p = 0·72). During four weeks, in the canakinumab vs placebo group 4 (7·0%) vs 7 (12·3%) participants died, 11 (20·0 %) vs 16 (28·1%) patients were on ICU, 12 (23·5 %) vs 11 (21·6%) were hospitalised for more than 3 weeks, respectively. Median ventilation time at four weeks in the canakinumab vs placebo group was 10 [IQR 6.0, 16.5] and 16 days [IQR 14.0, 23.0], respectively. There was no statistically significant difference in HbA1c after four weeks despite a lower number of anti-diabetes drug administered in patients treated with canakinumab. Finally, high-sensitive CRP and IL-6 was lowered by canakinumab. Serious adverse events were reported in 13 patients (11·4%) in each group. INTERPRETATION In patients with type 2 diabetes who were hospitalised with COVID-19, treatment with canakinumab in addition to standard-of-care did not result in a statistically significant improvement of the primary composite outcome. Patients treated with canakinumab required significantly less anti-diabetes drugs to achieve similar glycaemic control. Canakinumab was associated with a prolonged reduction of systemic inflammation. FUNDING Swiss National Science Foundation grant #198415 and University of Basel. Novartis supplied study medication.
Collapse
Affiliation(s)
- Matthias Hepprich
- University Hospital Basel, Division of Endocrinology, Diabetes and Metabolism, Basel, Switzerland
| | - Jonathan M. Mudry
- University Hospital Basel, Division of Endocrinology, Diabetes and Metabolism, Basel, Switzerland
| | - Claudia Gregoriano
- Medical University Department of Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Francois R. Jornayvaz
- Division of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Geneva University Hospital, Genève, Switzerland
| | - Sebastian Carballo
- Sevice of General Internal Medicine, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Anne Wojtusciszyn
- Service d'Endocrinologie Diabète et Métabolisme, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Pierre-Alexandre Bart
- Service of Internal Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Jean-Daniel Chiche
- Department of Intensive Care Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Stefan Fischli
- Department of Endocrinology, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Thomas Baumgartner
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zürich, Switzerland
| | - Claudia Cavelti-Weder
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zürich, Switzerland
| | - Dominique L. Braun
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland and Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Huldrych F. Günthard
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland and Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Felix Beuschlein
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zürich, Switzerland
| | - Anna Conen
- Division of Infectious Diseases and Infection Prevention, Kantonsspital Aarau, Aarau, Switzerland
| | - Emily West
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland and Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Egon Isenring
- Medical University Department of Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Stefan Zechmann
- University Hospital Basel, Division of Endocrinology, Diabetes and Metabolism, Basel, Switzerland
| | - Gabriela Bucklar
- University Hospital Basel, Division of Endocrinology, Diabetes and Metabolism, Basel, Switzerland
| | - Yoann Aubry
- University Hospital Basel, Division of Endocrinology, Diabetes and Metabolism, Basel, Switzerland
| | - Ludovic Dey
- Hôpital du Jura, Site de Delémont, Delémont, Switzerland
| | - Beat Müller
- Medical University Department of Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Patrick Hunziker
- Intensive Care Unit, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Philipp Schütz
- Medical University Department of Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Marco Cattaneo
- Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Marc Y. Donath
- University Hospital Basel, Division of Endocrinology, Diabetes and Metabolism, Basel, Switzerland
- Corresponding author.
| |
Collapse
|
25
|
Obi ON, Saketkoo LA, Russell AM, Baughman RP. Sarcoidosis: Updates on therapeutic drug trials and novel treatment approaches. Front Med (Lausanne) 2022; 9:991783. [PMID: 36314034 PMCID: PMC9596775 DOI: 10.3389/fmed.2022.991783] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/17/2022] [Indexed: 12/04/2022] Open
Abstract
Sarcoidosis is a systemic granulomatous inflammatory disease of unknown etiology. It affects the lungs in over 90% of patients yet extra-pulmonary and multi-organ involvement is common. Spontaneous remission of disease occurs commonly, nonetheless, over 50% of patients will require treatment and up to 30% of patients will develop a chronic progressive non-remitting disease with marked pulmonary fibrosis leading to significant morbidity and death. Guidelines outlining an immunosuppressive treatment approach to sarcoidosis were recently published, however, the strength of evidence behind many of the guideline recommended drugs is weak. None of the drugs currently used for the treatment of sarcoidosis have been rigorously studied and prescription of these drugs is often based on off-label” indications informed by experience with other diseases. Indeed, only two medications [prednisone and repository corticotropin (RCI) injection] currently used in the treatment of sarcoidosis are approved by the United States Food and Drug Administration. This situation results in significant reimbursement challenges especially for the more advanced (and often more effective) drugs that are favored for severe and refractory forms of disease causing an over-reliance on corticosteroids known to be associated with significant dose and duration dependent toxicities. This past decade has seen a renewed interest in developing new drugs and exploring novel therapeutic pathways for the treatment of sarcoidosis. Several of these trials are active randomized controlled trials (RCTs) designed to recruit relatively large numbers of patients with a goal to determine the safety, efficacy, and tolerability of these new molecules and therapeutic approaches. While it is an exciting time, it is also necessary to exercise caution. Resources including research dollars and most importantly, patient populations available for trials are limited and thus necessitate that several of the challenges facing drug trials and drug development in sarcoidosis are addressed. This will ensure that currently available resources are judiciously utilized. Our paper reviews the ongoing and anticipated drug trials in sarcoidosis and addresses the challenges facing these and future trials. We also review several recently completed trials and draw lessons that should be applied in future.
Collapse
Affiliation(s)
- Ogugua Ndili Obi
- Division of Pulmonary Critical Care and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, United States,*Correspondence: Ogugua Ndili Obi,
| | - Lesley Ann Saketkoo
- New Orleans Scleroderma and Sarcoidosis Patient Care and Research Center, New Orleans, LA, United States,University Medical Center—Comprehensive Pulmonary Hypertension Center and Interstitial Lung Disease Clinic Programs, New Orleans, LA, United States,Section of Pulmonary Medicine, Louisiana State University School of Medicine, New Orleans, LA, United States,Department of Undergraduate Honors, Tulane University School of Medicine, New Orleans, LA, United States
| | - Anne-Marie Russell
- Exeter Respiratory Institute University of Exeter, Exeter, United Kingdom,Royal Devon and Exeter NHS Foundation Trust, Devon, United Kingdom,Faculty of Medicine, Imperial College and Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Robert P. Baughman
- Department of Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
26
|
Abbate A, Wohlford GF, Del Buono MG, Chiabrando JG, Markley R, Turlington J, Kadariya D, Trankle CR, Biondi-Zoccai G, Lipinski MJ, Van Tassell BW. Interleukin-1 blockade with anakinra and heart failure following ST-segment elevation myocardial infarction: results from a pooled analysis of the VCUART clinical trials. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2022; 8:503-510. [PMID: 34617567 PMCID: PMC9366639 DOI: 10.1093/ehjcvp/pvab075] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/17/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022]
Abstract
AIMS ST-segment elevation myocardial infarction (STEMI) is associated with an intense acute inflammatory response and an increased risk of death and heart failure (HF). In this study, we sought to evaluate the effect of anakinra, a recombinant interleukin-1 receptor antagonist, on the incidence of HF. METHODS AND RESULTS We performed a pooled analysis of three early phase randomized clinical trials. The endpoints included the composite of all-cause death and new-onset HF, and the composite of all-cause death and hospitalization for HF at 1-year follow-up. Safety events, including injection site reaction and serious infections, were also recorded. We analysed 139 patients with STEMI from three separate trials: VCUART (N = 10), VCUART2 (N = 30), and VCUART3 (N = 99). Of these, 84 (60%) patients were randomized to anakinra and 55 (40%) to placebo. Treatment with anakinra significantly reduced the incidence of all-cause death or new-onset HF (7 [8.2%] vs. 16 [29.1%], log-rank P = 0.002) and of all-cause death or HF hospitalization (0 [0] vs. 5 [9.1%], log-rank P = 0.007). Patients treated with anakinra had significantly higher injection site reactions (19 [22.6%] vs. 3 [5.5%], P = 0.016) without a significant difference in the incidence of serious infections (11 [13.1%] vs. 7 [12.7%], P = 0.435). Treatment with anakinra significantly reduced the area under the curve for high-sensitivity C-reactive protein between baseline and 14 days (75.48 [41.7-147.47] vs. 222.82 [117.22-399.28] mg day/L, P < 0.001). CONCLUSION IL-1 blockade with anakinra for 14 days in patients with STEMI reduces the incidence of new-onset HF or hospitalization for HF at 1 year following STEMI.
Collapse
Affiliation(s)
- Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - George F Wohlford
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | - Roshanak Markley
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jeremy Turlington
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Dinesh Kadariya
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Cory R Trankle
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Michael J Lipinski
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | |
Collapse
|
27
|
Van Tassell B, Mihalick V, Thomas G, Marawan A, Talasaz AH, Lu J, Kang L, Ladd A, Damonte JI, Dixon DL, Markley R, Turlington J, Federmann E, Del Buono MG, Biondi-Zoccai G, Canada JM, Arena R, Abbate A. Rationale and design of interleukin-1 blockade in recently decompensated heart failure (REDHART2): a randomized, double blind, placebo controlled, single center, phase 2 study. J Transl Med 2022; 20:270. [PMID: 35706006 PMCID: PMC9198622 DOI: 10.1186/s12967-022-03466-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/30/2022] [Indexed: 01/01/2023] Open
Abstract
Background Heart failure (HF) is a global leading cause of mortality despite implementation of guideline directed therapy which warrants a need for novel treatment strategies. Proof-of-concept clinical trials of anakinra, a recombinant human Interleukin-1 (IL-1) receptor antagonist, have shown promising results in patients with HF. Method We designed a single center, randomized, placebo controlled, double-blind phase II randomized clinical trial. One hundred and two adult patients hospitalized within 2 weeks of discharge due to acute decompensated HF with reduced ejection fraction (HFrEF) and systemic inflammation (high sensitivity of C-reactive protein > 2 mg/L) will be randomized in 2:1 ratio to receive anakinra or placebo for 24 weeks. The primary objective is to determine the effect of anakinra on peak oxygen consumption (VO2) measured at cardiopulmonary exercise testing (CPX) after 24 weeks of treatment, with placebo-corrected changes in peak VO2 at CPX after 24 weeks (or longest available follow up). Secondary exploratory endpoints will assess the effects of anakinra on additional CPX parameters, structural and functional echocardiographic data, noninvasive hemodynamic, quality of life questionnaires, biomarkers, and HF outcomes. Discussion The current trial will assess the effects of IL-1 blockade with anakinra for 24 weeks on cardiorespiratory fitness in patients with recent hospitalization due to acute decompensated HFrEF. Trial registration: The trial was registered prospectively with ClinicalTrials.gov on Jan 8, 2019, identifier NCT03797001.
Collapse
Affiliation(s)
- Benjamin Van Tassell
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA. .,Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA, USA.
| | - Virginia Mihalick
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Georgia Thomas
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Amr Marawan
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Azita H Talasaz
- Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Juan Lu
- Division of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, USA
| | - Le Kang
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy Ladd
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Juan Ignacio Damonte
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Dave L Dixon
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA.,Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Roshanak Markley
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Jeremy Turlington
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Emily Federmann
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Marco Giuseppe Del Buono
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Mediterranea Cardiocentro, Naples, Italy
| | - Justin M Canada
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Ross Arena
- Department of Physical Therapy, College of Applied Science, University of Illinois Chicago, Chicago, IL, USA
| | - Antonio Abbate
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| |
Collapse
|
28
|
Wang C, Wu Y, Su Y, Mao B, Luo Y, Yan Y, Hu K, Lu Y, Che W, Wan M. Elevated levels of sIL-2R, TNF-α and hs-CRP are independent risk factors for post percutaneous coronary intervention coronary slow flow in patients with non-ST segment elevation acute coronary syndrome. Int J Cardiovasc Imaging 2022; 38:1191-1202. [PMID: 35182256 PMCID: PMC11143008 DOI: 10.1007/s10554-022-02529-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/12/2022] [Indexed: 11/27/2022]
Abstract
To evaluate the association between circulating levels of inflammatory cytokines and the occurrence of post-percutaneous coronary intervention (PCI) coronary slow flow (CSF) in patients with non-ST segment elevation acute coronary syndrome (NSTE-ACS). CSF after PCI commonly occurs and implies poor outcomes, while the determinants of post-PCI CSF in patients with NSTE-ACS remain controversial. In this multicenter case control study, 176 patients diagnosed with NSTE-ACS and with post-PCI CSF occurred composed of CSF group, while 352 matched NSTE-ACS patients composed control group. Corrected thrombolysis in myocardial infarction frame count (cTFC), circulating levels of inflammatory cytokines and PCI related parameters were analyzed using Logistic regression models. Among 528 patients with median age of 67 (59-76) and male proportion of 65.5%, 176 (35.0%) patients had occurrence of post-PCI CSF defined as cTFC ≥ 24. Patients with CSF presented more intense inflammatory activity revealed by higher levels of white blood cell, high-sensitivity C-reactive protein (hs-CRP), interleukin-1ß (IL-1ß), soluble IL-2 receptor (sIL-2R), IL-6, IL-8, IL-10 and tumor necrosis factor-α (TNF-α), while PCI related parameters were comparable. Correlation analysis showed cTFC was positively correlated with those inflammatory cytokines. Logistic regression model indicates that hs-CRP (odds ratio (OR) = 3.038, 95% confidence interval (CI) 1.545-5.975), sIL-2R (OR = 2.103, 95% CI 1.959-4.026) and TNF-α (OR = 3.708, 95% CI 1.426-9.641) were valuable predictors for CSF occurrence. Elevated circulating levels of inflammatory cytokine including hs-CRP, sIL-2R and TNF-α rather than PCI related parameters could predict post-PCI CSF in patients with NSTE-ACS.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Yan Wu
- Department of Cardiology, Shanghai Putuo District Liqun Hospital, Shanghai, 200333, China
| | - Yang Su
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai, 200072, China
| | - Bin Mao
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Yihong Luo
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Yexiang Yan
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Kun Hu
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Yi Lu
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Wenliang Che
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China.
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai, 200072, China.
| | - Minying Wan
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China.
| |
Collapse
|
29
|
Del Buono MG, Damonte JI, Chiabrando JG, Markley R, Turlington J, Trankle CR, Kang L, Biondi-Zoccai G, Van Tassell BW, Abbate A. Effect of IL-1 Blockade With Anakinra on Heart Failure Outcomes in Patients With Anterior Versus Nonanterior ST Elevation Myocardial Infarction. J Cardiovasc Pharmacol 2022; 79:774-780. [PMID: 35170493 PMCID: PMC9177574 DOI: 10.1097/fjc.0000000000001240] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/22/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Patients with ST elevation myocardial infarction (STEMI) are at risk of future heart failure (HF), particularly those with anterior STEMI. Interleukin-1 (IL-1) is a key mediator of the inflammatory response, and its blockade has emerged as a potential therapeutic strategy to prevent HF events. The aim of this analysis was to explore the effects of anakinra, an IL-1 receptor antagonist, on HF outcomes based on anterior versus nonanterior location STEMI and to explore whether this effect is mediated through the amelioration of left ventricular systolic function and cardiac remodeling. We pooled data from 3 early phase randomized clinical trials. The primary end point was a composite of all-cause death and new-onset HF at 1-year follow-up. The left anterior descending coronary artery as culprit vessel was used to identify anterior STEMI. We included 139 patients, 47 (34%) with anterior STEMI and 92 (66%) with nonanterior STEMI. Anakinra significantly reduced the combined end point of death or new-onset HF in patients with anterior STEMI [4 (13%) vs. 7 (42%), log-rank P value = 0.049] and in patients with nonanterior STEMI [3 (6%) vs. 9 (24%), log-rank P value = 0.014]. We found no significant differences comparing anakinra versus placebo in interval changes in left ventricular ejection fraction and volumes in anterior and nonanterior STEMI. In conclusion, anakinra is associated with a reduction of HF events in patients with STEMI, irrespective of anterior or nonanterior location, or of changes in left ventricular ejection fraction or cardiac remodeling.
Collapse
Affiliation(s)
- Marco Giuseppe Del Buono
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Juan Ignacio Damonte
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Argentina
| | - Juan Guido Chiabrando
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Argentina
| | - Roshanak Markley
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jeremy Turlington
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Cory R. Trankle
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Le Kang
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Benjamin W. Van Tassell
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
30
|
Liao Y, Liu K, Zhu L. Emerging Roles of Inflammasomes in Cardiovascular Diseases. Front Immunol 2022; 13:834289. [PMID: 35464402 PMCID: PMC9021369 DOI: 10.3389/fimmu.2022.834289] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/07/2022] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular diseases are known as the leading cause of morbidity and mortality worldwide. As an innate immune signaling complex, inflammasomes can be activated by various cardiovascular risk factors and regulate the activation of caspase-1 and the production and secretion of proinflammatory cytokines such as IL-1β and IL-18. Accumulating evidence supports that inflammasomes play a pivotal role in the progression of atherosclerosis, myocardial infarction, and heart failure. The best-known inflammasomes are NLRP1, NLRP3, NLRC4, and AIM2 inflammasomes, among which NLRP3 inflammasome is the most widely studied in the immune response and disease development. This review focuses on the activation and regulation mechanism of inflammasomes, the role of inflammasomes in cardiovascular diseases, and the research progress of targeting NLRP3 inflammasome and IL-1β for related disease intervention.
Collapse
Affiliation(s)
- Yingnan Liao
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Kui Liu
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, China
| | - Liyuan Zhu
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
31
|
Heart Failure After ST-Elevation Myocardial Infarction: Beyond Left Ventricular Adverse Remodeling. Curr Probl Cardiol 2022:101215. [PMID: 35460680 DOI: 10.1016/j.cpcardiol.2022.101215] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/13/2022] [Indexed: 12/11/2022]
Abstract
ST-segment elevation myocardial infarction (STEMI) remains a significant source of morbidity and mortality worldwide. Despite advances in treatment leading to a significant reduction in the early complications and in-hospital mortality, a significant proportion of STEMI survivors develop heart failure (HF) at follow-up. The classic paradigm of HF after STEMI is one characterized by left ventricular adverse remodeling (LVAR) and encompasses the process of regional and global structural and functional changes that occur in the heart as a consequence of loss of viable myocardium, increased wall stress and neurohormonal activation, and results in HF with reduced ejection fraction (HFrEF). More recently, however, with further improvements in the treatment of STEMI the incidence and entity of LVAR appear to be largely reduced, yet the risk for HF following STEMI is not abolished and remains substantial, identifying a new paradigm by which patients with STEMI present with HF and preserved EF (HFpEF) characterized by reduction of diastolic or systolic reserve independent of LVAR.
Collapse
|
32
|
Armbruster AL, Campbell KB, Kahanda MG, Cuculich PS. The role of inflammation in the pathogenesis and treatment of arrhythmias. Pharmacotherapy 2022; 42:250-262. [PMID: 35098555 DOI: 10.1002/phar.2663] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022]
Abstract
The pathogenesis of arrhythmias is complex and multifactorial. The role of inflammation in the pathogenesis of both atrial and ventricular arrhythmias (VA) has been explored. However, developing successful pharmacotherapy regimens based on those pathways has proven more of a challenge. This narrative review provides an overview of five common arrhythmias impacted by inflammation, including atrial fibrillation (AF), myocardial infarction, arrhythmogenic cardiomyopathy, cardiac sarcoidosis, and QT prolongation, and the potential role for anti-inflammatory therapy in their management. We identified arrhythmias and arrhythmogenic disease states with the most evidence linking pathogenesis to inflammation and conducted comprehensive searches of United States National Library of Medicine MEDLINE® and PubMed databases. Although a variety of agents have been studied for the management of AF, primarily in an effort to reduce postoperative AF following cardiac surgery, no standard anti-inflammatory agents are used in clinical practice at this time. Although inflammation following myocardial infarction may contribute to the development of VA, there is no clear benefit with the use of anti-inflammatory agents at this time. Similarly, although inflammation is clearly linked to the development of arrhythmias in arrhythmogenic cardiomyopathy, data demonstrating a benefit with anti-inflammatory agents are limited. Cardiac sarcoidosis, an infiltrative disease eliciting an immune response, is primarily treated by immunosuppressive therapy and steroids, despite a lack of primary literature to support such regimens. In this case, anti-inflammatory agents are frequently used in clinical practice. The pathophysiology of arrhythmias is complex, and inflammation likely plays a role in both onset and duration, however, for most arrhythmias the role of pharmacotherapy targeting inflammation remains unclear.
Collapse
Affiliation(s)
- Anastasia L Armbruster
- St. Louis College of Pharmacy, University of Health Sciences and Pharmacy in St. Louis, St. Louis, Missouri, USA
| | | | - Milan G Kahanda
- Cardiovascular Division, Department of Internal Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Phillip S Cuculich
- Cardiovascular Division, Department of Internal Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
33
|
Inflammation as a mechanism and therapeutic target in peripheral artery disease. Can J Cardiol 2022; 38:588-600. [PMID: 35114347 DOI: 10.1016/j.cjca.2022.01.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/24/2022] Open
Abstract
Peripheral artery disease is one of three major clinical manifestations of atherosclerosis, the other two being coronary artery and cerebrovascular disease. Despite progress in surgery, antithrombotic therapy and therapies that modify conventional risk factors (lipid-, blood pressure-, and glucose-lowering interventions), patients with peripheral artery disease have unacceptably high risk of vascular complications. Additional strategies to reduce this residual risk are needed. The accumulated evidence that inflammation plays an important role in the pathogenesis of atherosclerosis has spurred recent efforts to evaluate anti-inflammatory agents as an additional therapeutic approach for atherothrombosis prevention and treatment. In this review, we examine the evidence supporting the role of inflammation in atherosclerosis, review recent trials evaluating anti-inflammatory approaches to reduce cardiovascular complications, and offer insights into the opportunities for novel anti-inflammatory strategies to reduce the burden of cardiovascular and limb complications in patients with peripheral artery disease.
Collapse
|
34
|
Lin J, Li Q, Jin T, Wang J, Gong Y, Lv Q, Wang M, Chen J, Shang M, Zhao Y, Fu G. Cardiomyocyte IL-1R2 protects heart from ischemia/reperfusion injury by attenuating IL-17RA-mediated cardiomyocyte apoptosis. Cell Death Dis 2022; 13:90. [PMID: 35087030 PMCID: PMC8795442 DOI: 10.1038/s41419-022-04533-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 12/17/2021] [Accepted: 01/13/2022] [Indexed: 12/30/2022]
Abstract
Myocardial ischemia reperfusion (I/R) injury is a complex process with intense inflammatory response and cardiomyocyte apoptosis. As a decoy receptor of IL-1β, Interleukin-1 receptor type 2 (IL-1R2) inhibits IL-1β signaling. However, its role in I/R injury remains unknown. Here we found that the serum levels of IL-1R2 were significantly increased in patients with acute myocardial infarction (AMI) following interventional therapy. Similarly, after myocardial I/R surgery, IL-1R2 expression was significantly increased in heart of wild-type mice. In addition, IL-1R2-deficient mice heart showed enlarged infarct size, increased cardiomyocyte apoptosis together with reduced cardiac systolic function. Following exposure to hypoxia and reoxygenation (H/R), neonatal rat ventricular myocytes (NRVM) significantly increased IL-1R2 expression relying on NF-κB activation. Consistently, IL-1R2-deficient mice increased immune cells infiltrating into heart after surgery, which was relevant with cardiac damage. Additionally, IL-1R2 overexpression in cardiomyocyte protected cardiomyocyte against apoptosis through reducing the IL-17RA expression both in vivo and in vitro. Our results indicate that IL-1R2 protects cardiomyocytes from apoptosis, which provides a therapeutic approach to turn down myocardial I/R injury.
Collapse
Affiliation(s)
- Jun Lin
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Qinfeng Li
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Tingting Jin
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Jiacheng Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Yingchao Gong
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Qingbo Lv
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Meihui Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Jiawen Chen
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Min Shang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China.
| | - Yanbo Zhao
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China.
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
35
|
Del Buono MG, Damonte JI, Trankle CR, Kadariya D, Carbone S, Thomas G, Turlington J, Markley R, Canada JM, Biondi-Zoccai GG, Kontos MC, Van Tassell BW, Abbate A. Effect of interleukin-1 blockade with anakinra on leukocyte count in patients with ST-segment elevation acute myocardial infarction. Sci Rep 2022; 12:1254. [PMID: 35075216 PMCID: PMC8786840 DOI: 10.1038/s41598-022-05374-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/11/2022] [Indexed: 02/08/2023] Open
Abstract
Leukocytosis is a common finding in patients with ST elevation myocardial infarction (STEMI) and portends a poor prognosis. Interleukin 1-β regulates leukopoiesis and pre-clinical studies suggest that anakinra (recombinant human interleukin-1 [IL-1] receptor antagonist) suppresses leukocytosis in myocardial infarction. However, the effect of IL-1 blockade with anakinra on leukocyte count in patients with STEMI is unknown. We reviewed the white blood cell (WBC) and differential count of 99 patients enrolled in a clinical trial of anakinra (n = 64) versus placebo (n = 35) for 14 days after STEMI. A complete blood cell count with differential count were obtained at admission, and after 72 h, 14 days and 3 months. After 72 h from treatment, anakinra compared to placebo led to a statistically significant greater percent reduction in total WBC count (− 35% [− 48 to − 24] vs. − 21% [− 34 to − 10], P = 0.008), absolute neutrophil count (− 48% [− 60 to − 22] vs. − 27% [− 46 to − 5], P = 0.004) and to an increase in absolute eosinophil count (+ 50% [0 to + 100] vs. 0% [− 50 to + 62], P = 0.022). These changes persisted while on treatment at 14 days and were no longer apparent at 3 months after treatment discontinuation. We found that in patients with STEMI IL-1 blockade with anakinra accelerates resolution of leukocytosis and neutrophilia. This modulation may represent one of the mechanisms by which IL-1 blockade improves clinical outcomes.
Collapse
Affiliation(s)
- Marco Giuseppe Del Buono
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA.,Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Juan Ignacio Damonte
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA.,Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Cory R Trankle
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA
| | - Dinesh Kadariya
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA
| | - Salvatore Carbone
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA.,Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Georgia Thomas
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA
| | - Jeremy Turlington
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA
| | - Roshanak Markley
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA
| | - Justin M Canada
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA
| | - Giuseppe G Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Mediterranea Cardiocentro, Naples, Italy
| | - Michael C Kontos
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA
| | - Benjamin W Van Tassell
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA.,Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, West Hospital, West Wing 5-020, 1200 E Broad Street, P.O. Box 980204, Richmond, VA, 23298, USA.
| |
Collapse
|
36
|
Rohm TV, Meier DT, Olefsky JM, Donath MY. Inflammation in obesity, diabetes, and related disorders. Immunity 2022; 55:31-55. [PMID: 35021057 PMCID: PMC8773457 DOI: 10.1016/j.immuni.2021.12.013] [Citation(s) in RCA: 626] [Impact Index Per Article: 313.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 01/13/2023]
Abstract
Obesity leads to chronic, systemic inflammation and can lead to insulin resistance (IR), β-cell dysfunction, and ultimately type 2 diabetes (T2D). This chronic inflammatory state contributes to long-term complications of diabetes, including non-alcoholic fatty liver disease (NAFLD), retinopathy, cardiovascular disease, and nephropathy, and may underlie the association of type 2 diabetes with other conditions such as Alzheimer's disease, polycystic ovarian syndrome, gout, and rheumatoid arthritis. Here, we review the current understanding of the mechanisms underlying inflammation in obesity, T2D, and related disorders. We discuss how chronic tissue inflammation results in IR, impaired insulin secretion, glucose intolerance, and T2D and review the effect of inflammation on diabetic complications and on the relationship between T2D and other pathologies. In this context, we discuss current therapeutic options for the treatment of metabolic disease, advances in the clinic and the potential of immune-modulatory approaches.
Collapse
Affiliation(s)
- Theresa V. Rohm
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel T. Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Jerrold M. Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marc Y. Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland.,Correspondence:
| |
Collapse
|
37
|
Chan YH, Ramji DP. Key Roles of Inflammation in Atherosclerosis: Mediators Involved in Orchestrating the Inflammatory Response and Its Resolution in the Disease Along with Therapeutic Avenues Targeting Inflammation. Methods Mol Biol 2022; 2419:21-37. [PMID: 35237956 DOI: 10.1007/978-1-0716-1924-7_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Inflammation is a critical driver of all stages of atherosclerosis, from lesion development to plaque rupture. Cytokines are mediators of the immune response and in atherosclerosis, the balance of anti- and pro-inflammatory cytokines is tipped in favor of the latter, resulting in persistent and unresolved inflammation. Although reducing plasma cholesterol levels mainly via the use of statins has positively impacted patient outcomes and reduced mortality rates, the presence of significant residual inflammation and cardiovascular risk posttherapy emphasizes the prevailing risk of primary and secondary events driven by inflammation independently of hyperlipidemia. Given the dominant role of inflammation in driving pathogenesis, alternative therapeutic avenues beyond targeting lowering of plasma lipids are required. This chapter will discuss the role of inflammation and pro-inflammatory cytokines in driving atherogenesis and disease progression, the therapeutic potential of targeting cytokines for atherosclerosis and promising avenues in this area.
Collapse
Affiliation(s)
- Yee-Hung Chan
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK.
| | - Dipak P Ramji
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
38
|
Toldo S, Mezzaroma E, Buckley LF, Potere N, Di Nisio M, Biondi-Zoccai G, Van Tassell BW, Abbate A. Targeting the NLRP3 inflammasome in cardiovascular diseases. Pharmacol Ther 2021; 236:108053. [PMID: 34906598 PMCID: PMC9187780 DOI: 10.1016/j.pharmthera.2021.108053] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/21/2021] [Accepted: 12/06/2021] [Indexed: 02/05/2023]
Abstract
The NACHT, leucine-rich repeat (LRR), and pyrin domain (PYD)-containing protein 3 (NLRP3) inflammasome is an intracellular sensing protein complex that plays a major role in innate immunity. Following tissue injury, activation of the NLRP3 inflammasome results in cytokine production, primarily interleukin(IL)-1β and IL-18, and, eventually, inflammatory cell death - pyroptosis. While a balanced inflammatory response favors damage resolution and tissue healing, excessive NLRP3 activation causes detrimental effects. A key involvement of the NLRP3 inflammasome has been reported across a wide range of cardiovascular diseases (CVDs). Several pharmacological agents selectively targeting the NLRP3 inflammasome system have been developed and tested in animals and early phase human studies with overall promising results. While the NLRP3 inhibitors are in clinical development, multiple randomized trials have demonstrated the safety and efficacy of IL-1 blockade in atherothrombosis, heart failure and recurrent pericarditis. Furthermore, the non-selective NLRP3 inhibitor colchicine has been recently shown to significantly reduce cardiovascular events in patients with chronic coronary disease. In this review, we will outline the mechanisms driving NLRP3 assembly and activation, and discuss the pathogenetic role of the NLRP3 inflammasome in CVDs, providing an overview of the current and future therapeutic approaches targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Stefano Toldo
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Eleonora Mezzaroma
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Pharmacotherapy and Outcome Studies, Virginia Commonwealth University, Richmond, VA, USA
| | - Leo F Buckley
- Department of Pharmacy, Brigham and Women's Hospital, Boston, MA, USA
| | - Nicola Potere
- Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Marcello Di Nisio
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Mediterranea Cardiocentro, Napoli, Italy
| | - Benjamin W Van Tassell
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Pharmacotherapy and Outcome Studies, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
39
|
Kron J, Crawford T, Mihalick V, Bogun F, Jordan JH, Koelling T, Syed H, Syed A, Iden T, Polly K, Federmann E, Bray K, Lathkar-Pradhan S, Jasti S, Rosenfeld L, Birnie D, Smallfield M, Kang L, Fowler AB, Ladd A, Ellenbogen K, Van Tassell B, Gregory Hundley W, Abbate A. Interleukin-1 blockade in cardiac sarcoidosis: study design of the multimodality assessment of granulomas in cardiac sarcoidosis: Anakinra Randomized Trial (MAGiC-ART). J Transl Med 2021; 19:460. [PMID: 34749739 PMCID: PMC8575149 DOI: 10.1186/s12967-021-03130-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/23/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Sarcoidosis is an inflammatory disease characterized by the formation of granulomas, which involve the heart in up to 25% of patients. Cardiac sarcoidosis can lead to life threatening arrhythmias and heart failure. While corticosteroids have been used as a treatment for over 50 years, they are associated with hypertension, diabetes, and weight gain, further increasing cardiovascular risk. Interleukin-1 (IL-1) is the prototypical proinflammatory cytokine that works to activate the nuclear transcription factor NF-kB, one of the targets of glucocorticoids. IL-1 also plays an important role also in the pathophysiology of heart disease including atherosclerosis, myocardial infarction, and myocarditis. METHODS Building on a network of research collaborators developed in the Cardiac Sarcoidosis Consortium, we will investigate the feasibility and tolerability of treatment of CS with anakinra at two National Institute of Health Clinical and Translational Science Award (CTSA) hubs with expertise in cardiac sarcoidosis. In this pilot study, up to 28 patients with cardiac sarcoidosis will be recruited to compare the administration of an IL-1 blocker, anakinra, 100 mg daily on top of standard of care versus standard of care only for 28 days and followed for 180 days. Utilizing surrogate endpoints of changes in systemic inflammatory biomarkers and cardiac imaging, we aim to determine whether IL-1 blockade with anakinra can combat systemic and cardiac inflammation in patients with cardiac sarcoidosis. DISCUSSION The current trial demonstrates an innovative collaborative approach to clinical trial development in a rare, understudied disease that disproportionately affects females and minorities. Trial Registration The trial was registered prospectively with ClinicalTrials.gov on July 12, 2019, identifier NCT04017936.
Collapse
Affiliation(s)
- Jordana Kron
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA.
| | - Thomas Crawford
- Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Virginia Mihalick
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA
| | - Frank Bogun
- Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer H Jordan
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA.,Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Todd Koelling
- Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Huzaefah Syed
- Division of Rheumatology, Virginia Commonwealth University, Allergy, and Immunology, Richmond, VA, USA
| | - Aamer Syed
- Division of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, VA, USA
| | - Thomas Iden
- Division of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, VA, USA
| | - Kelly Polly
- Division of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, VA, USA
| | - Emily Federmann
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA
| | - Kirsta Bray
- Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | | | - Shilpa Jasti
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA
| | - Lynda Rosenfeld
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - David Birnie
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Melissa Smallfield
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA
| | - Le Kang
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Alpha Berry Fowler
- Division of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy Ladd
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA
| | - Kenneth Ellenbogen
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA
| | - Benjamin Van Tassell
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA.,Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, USA
| | - W Gregory Hundley
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Virginia Commonwealth University Medical Center, P.O. Box 980053, Richmond, VA, 23298-0053, USA.,Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
40
|
Chen W, Xing J, Liu X, Wang S, Xing D. The role and transformative potential of IL-19 in atherosclerosis. Cytokine Growth Factor Rev 2021; 62:70-82. [PMID: 34600839 DOI: 10.1016/j.cytogfr.2021.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/19/2022]
Abstract
Atherosclerotic cardiovascular disease is the leading cause of death worldwide. Traditionally, IL-19 was thought to be expressed in only immune cells, but studies revealed that IL-19 is also expressed in multiple atherosclerotic plaque cell types, but not normal arteries, in humans and mice. IL-19 reduces the development of atherosclerosis via multiple mechanisms, including balancing cholesterol metabolism; enhancing Th2 immune cell polarization; reducing the inflammatory response; and reducing the proliferation, migration and chemotaxis of vascular smooth muscle cells (VSMCs). Clinical and/or animal studies have primarily aimed to achieve regression and/or stabilization of atherosclerotic plaques, with regression in particular indicating a very good drug response. Most antiatherosclerotic drugs in current clinical use, including atorvastatin and alirocumab, target hyperlipidemia. Several other drugs have also been investigated in clinical trials as anti-inflammatory agents; the development of some of these agents has been terminated (canakinumab, darapladib, varespladib, losmapimod, atreleuton, setileuton, PF-04191834, veliflapon, and methotrexate), but others remain in development (ziltivekimab, tocilizumab, Somalix, IFM-2427, anakinra, mesenchymal stem cells (MSCs), colchicine, everolimus, allopurinol, and montelukast). Most of the tested drugs have shown a limited ability to reverse atherosclerosis in animal studies. Interestingly, recombinant IL-19 (rIL-19) was shown to reduce atherosclerosis development in a time- and dose-dependent manner. A low dose of rIL-19 (1 ng/g/day) reduced aortic arch and root plaque areas by 70.1% and 32.1%, respectively, in LDLR-/- mice. At 10 ng/g/day, rIL-19 completely eliminated atherosclerotic plaques. There were no sex differences in the effects of rIL-19 on atherosclerotic mice. Thus, low-dose rIL-19 is an effective antiatherosclerotic agent, in addition to its efficacy in intimal hyperplasia, spinal cord injury, stroke, and multiple sclerosis. We propose that IL-19 is a promising biomarker and target for the diagnosis and treatment of atherosclerosis. This review considers the role and mechanism of action of IL-19 in atherosclerosis and discusses whether IL-19 is a potential therapeutic target for this condition.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Jiyao Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Xinlin Liu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Shuai Wang
- School of Medical Imaging, Radiotherapy Department, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
41
|
Interleukin 1 receptor antagonism abrogates acute pressure-overload induced murine heart failure. Ann Thorac Surg 2021; 114:98-107. [PMID: 34419440 DOI: 10.1016/j.athoracsur.2021.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Recent clinical trials have suggested that blockade of interleukin-1 can favorably impact patients with myocardial infarction and heart failure. However, the mechanism of how antagonism of this specific cytokine in mediating cardiac disease remains unclear. Hence, we sought to determine the influence of IL-1 blockade on acute hypertensive remodeling. METHODS Transverse aortic constriction (TAC) was performed in C57BL mice with or without intraperitoneal administration of interleukin 1 receptor antagonism (IL-1ra). Function, structure, and molecular diagnostics were subsequently performed and analyzed. RESULTS Six weeks after TAC, a progressive decline of ejection fraction and increases in LV mass and dimensions was effectively mitigated with IL-1ra. TAC resulted in an expected profile of hypertrophic markers including myosin heavy chain, atrial natriuretic peptide, and skeletal muscle actin which were all significantly lower in IL-1ra treated mice. While trichrome staining 2-weeks post TAC demonstrated similar levels of fibrosis, IL-1ra reduced expression of collagen-1, TIMP1, and periostin. Investigating the angiogenic response to pressure overload, similar levels of VEGF were observed, but IL-1ra was associated with more SDF-1. Immune cell infiltration (macrophages and lymphocytes) was also decreased in IL-1ra treated mice. Similarly, cytokine concentrations of IL-1, IL-18, and IL-6 were all reduced in IL-1ra-treated animals. CONCLUSIONS IL-1ra prevents the progression towards heart failure associated with acute pressure overload. This functional response was associated with reductions in mediators of fibrosis, cellular infiltration, and cytokine production. These results provide mechanistic insight into recent clinical trials and could springboard future investigations in patients with pressure-overload based cardiomyopathies.
Collapse
|
42
|
Kloner RA. Treating Acute Myocardial Infarctions With Anti-Inflammatory Agents. J Cardiovasc Pharmacol Ther 2021; 26:736-738. [PMID: 34328816 DOI: 10.1177/10742484211033711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Robert A Kloner
- Cardiovascular Research Institute, 6465Huntington Medical Research Institutes, Pasadena, CA, USA.,Cardiovascular Division, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
43
|
Morris G, Berk M, Walder K, O'Neil A, Maes M, Puri BK. The lipid paradox in neuroprogressive disorders: Causes and consequences. Neurosci Biobehav Rev 2021; 128:35-57. [PMID: 34118292 DOI: 10.1016/j.neubiorev.2021.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 04/27/2021] [Accepted: 06/06/2021] [Indexed: 02/07/2023]
Abstract
Chronic systemic inflammation is associated with an increased risk of cardiovascular disease in an environment of low low-density lipoprotein (LDL) and low total cholesterol and with the pathophysiology of neuroprogressive disorders. The causes and consequences of this lipid paradox are explored. Circulating activated neutrophils can release inflammatory molecules such as myeloperoxidase and the pro-inflammatory cytokines interleukin-1 beta, interleukin-6 and tumour necrosis factor-alpha. Since activated neutrophils are associated with atherosclerosis and cardiovascular disease and with major depressive disorder, bipolar disorder and schizophrenia, it seems reasonable to hypothesise that the inflammatory molecules released by them may act as mediators of the link between systemic inflammation and the development of atherosclerosis in neuroprogressive disorders. This hypothesis is tested by considering the association at a molecular level of systemic inflammation with increased LDL oxidation; increased small dense LDL levels; increased lipoprotein (a) concentration; secretory phospholipase A2 activation; cytosolic phospholipase A2 activation; increased platelet activation; decreased apolipoprotein A1 levels and function; decreased paroxonase-1 activity; hyperhomocysteinaemia; and metabolic endotoxaemia. These molecular mechanisms suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand
| | | |
Collapse
|
44
|
Tucker B, Vaidya K, Cochran BJ, Patel S. Inflammation during Percutaneous Coronary Intervention-Prognostic Value, Mechanisms and Therapeutic Targets. Cells 2021; 10:cells10061391. [PMID: 34199975 PMCID: PMC8230292 DOI: 10.3390/cells10061391] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/17/2022] Open
Abstract
Periprocedural myocardial injury and myocardial infarction (MI) are not infrequent complications of percutaneous coronary intervention (PCI) and are associated with greater short- and long-term mortality. There is an abundance of preclinical and observational data demonstrating that high levels of pre-, intra- and post-procedural inflammation are associated with a higher incidence of periprocedural myonecrosis as well as future ischaemic events, heart failure hospitalisations and cardiac-related mortality. Beyond inflammation associated with the underlying coronary pathology, PCI itself elicits an acute inflammatory response. PCI-induced inflammation is driven by a combination of direct endothelial damage, liberation of intra-plaque proinflammatory debris and reperfusion injury. Therefore, anti-inflammatory medications, such as colchicine, may provide a novel means of improving PCI outcomes in both the short- and long-term. This review summarises periprocedural MI epidemiology and pathophysiology, evaluates the prognostic value of pre-, intra- and post-procedural inflammation, dissects the mechanisms involved in the acute inflammatory response to PCI and discusses the potential for periprocedural anti-inflammatory treatment.
Collapse
Affiliation(s)
- Bradley Tucker
- Heart Research Institute, 7 Eliza St., Newtown 2042, Australia;
- Sydney Medical School, University of Sydney, Camperdown 2050, Australia;
- School of Medical Sciences, University of New South Wales, Kensington 2052, Australia;
| | - Kaivan Vaidya
- Sydney Medical School, University of Sydney, Camperdown 2050, Australia;
- Royal Prince Alfred Hospital, Camperdown 2050, Australia
| | - Blake J. Cochran
- School of Medical Sciences, University of New South Wales, Kensington 2052, Australia;
| | - Sanjay Patel
- Heart Research Institute, 7 Eliza St., Newtown 2042, Australia;
- Sydney Medical School, University of Sydney, Camperdown 2050, Australia;
- Royal Prince Alfred Hospital, Camperdown 2050, Australia
- Correspondence: ; Tel.: +61-2-9515-6111
| |
Collapse
|
45
|
Fragoulis GE, Soulaidopoulos S, Sfikakis PP, Dimitroulas T, D Kitas G. Effect of Biologics on Cardiovascular Inflammation: Mechanistic Insights and Risk Reduction. J Inflamm Res 2021; 14:1915-1931. [PMID: 34017189 PMCID: PMC8131071 DOI: 10.2147/jir.s282691] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/14/2021] [Indexed: 12/17/2022] Open
Abstract
It is increasingly recognized that atherosclerosis and consequently cardiovascular disease (CVD) are closely linked with inflammatory processes. The latter is in the center of the pathogenic mechanism underlying autoimmune rheumatic diseases (ARD). It follows then, that optimal control of inflammation in ARDs may lead to a decrease of the accompanied CVD risk. Major trials (eg, CANTOS, CIRT), aimed at examining the possible benefits of immunomodulatory treatments in CVD, demonstrated conflicting results. On the other hand, substantial evidence is accumulating about the possible beneficial effects of biologic disease modifying antirheumatic drugs (bDMARDs) in patients with ARDs, particularly those with rheumatoid arthritis (RA). It seems that bDMARDs (some more than others) alter the lipid profile in RA patients but do not adversely affect, in most cases, the TC/HDL ratio. Favorable effects are noted for arterial stiffness and endothelial function. This is reflected in the lower risk for CVD events, seen in observational studies of RA patients treated with bDMARDs. It should be stressed that more data exist for the TNF-inhibitors than for other bDMARDs, such as tocilizumab, abatacept and rituximab. As regards the spondyloarthropathies (SpA), data are less robust. For TNF-inhibitors, effects appear to be on par with those seen in RA but no conclusions can be drawn for newer biologic drugs used in SpA (eg, IL-17 blockers). Finally, there is accumulating evidence for a beneficial effect of immunosuppressive treatment in cardiac inflammation and function in several ARDs. Introduction of newer therapeutic options in clinical practice seem to have a positive impact on CVD in the setting of ARD. This is probably due to better control of inflammation, but direct improvement in vascular pathology is also a valid hypothesis. Most data are derived from observational studies and, therefore, randomized controlled trials are needed to assess the possible favorable effect of bDMARDs on CVD outcomes.
Collapse
Affiliation(s)
- George E Fragoulis
- Rheumatology Unit, Joint Rheumatology Program, Medical School, First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens, "Laiko" General Hospital, Athens, 115 27, Greece
| | - Stergios Soulaidopoulos
- First Department of Cardiology, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, 115 27, Greece
| | - Petros P Sfikakis
- Rheumatology Unit, Joint Rheumatology Program, Medical School, First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens, "Laiko" General Hospital, Athens, 115 27, Greece
| | - Theodoros Dimitroulas
- Fourth Department of Internal Medicine, Hippokration Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 546 41, Greece
| | - George D Kitas
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS FT, Dudley, DY1 2HQ, UK.,Arthritis Research UK Epidemiology Unit, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
46
|
Xu M, Zhang K, Song J. Targeted Therapy in Cardiovascular Disease: A Precision Therapy Era. Front Pharmacol 2021; 12:623674. [PMID: 33935716 PMCID: PMC8085499 DOI: 10.3389/fphar.2021.623674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Targeted therapy refers to exploiting the specific therapeutic drugs against the pathogenic molecules (a protein or a gene) or cells. The drug specifically binds to disease-causing molecules or cells without affecting normal tissue, thus enabling personalized and precision treatment. Initially, therapeutic drugs included antibodies and small molecules, (e.g. nucleic acid drugs). With the advancement of the biology technology and immunotherapy, the gene editing and cell editing techniques are utilized for the disease treatment. Currently, targeted therapies applied to treat cardiovascular diseases (CVDs) mainly include protein drugs, gene editing technologies, nucleic acid drugs and cell therapy. Although targeted therapy has demonstrated excellent efficacy in pre-clinical and clinical trials, several limitations need to be recognized and overcome in clinical application, (e.g. off-target events, gene mutations, etc.). This review introduces the mechanisms of different targeted therapies, and mainly describes the targeted therapy applied in the CVDs. Furthermore, we made comparative analysis to clarify the advantages and disadvantages of different targeted therapies. This overview is expected to provide a new concept to the treatment of the CVDs.
Collapse
Affiliation(s)
- Mengda Xu
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kailun Zhang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Kailun Zhang, ; Jiangping Song,
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,*Correspondence: Kailun Zhang, ; Jiangping Song,
| |
Collapse
|
47
|
Pasin L, Cavalli G, Navalesi P, Sella N, Landoni G, Yavorovskiy AG, Likhvantsev VV, Zangrillo A, Dagna L, Monti G. Anakinra for patients with COVID-19: a meta-analysis of non-randomized cohort studies. Eur J Intern Med 2021; 86:34-40. [PMID: 33581979 PMCID: PMC7862887 DOI: 10.1016/j.ejim.2021.01.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Severe COVID-19 cases have a detrimental hyper-inflammatory host response and different cytokine-blocking biologic agents were explored to improve outcomes. Anakinra blocks the activity of both IL-1α and IL‑1β and is approved for different autoinflammatory disorders, but it is used off-label for conditions characterized by an excess of cytokine production. Several studies on anakinra in COVID-19 patients reported positive effects. We performed a meta-analysis of all published evidence on the use of anakinra in COVID19 to investigate its effect on survival and need for mechanical ventilation. METHODS We searched for any study performed on adult patients with acute hypoxemic failure related to 2019-nCoV infection, receiving anakinra versus any comparator. Primary endpoint was mortality at the longest available follow-up. Adverse effects, need for mechanical ventilation and discharge at home with no limitations were also analysed. RESULTS Four observational studies involving 184 patients were included. Overall mortality of patients treated with anakinra was significantly lower than mortality in the control group (95% CI 0.14-0.48, p<0.0001). Moreover, patients treated with anakinra had a significantly lower risk of need for mechanical ventilation than controls (95% CI 0.250.74, p=0.002). No difference in adverse events and discharge at home with no limitations was observed. The Trial Sequential Analysis z-cumulative line reached the monitoring boundary for benefit and the required sample size. CONCLUSIONS Administration of anakinra in COVID-19 patients was safe and might be associated with reductions in both mortality and need for mechanical ventilation. Randomized clinical trials are warranted to confirm these findings.
Collapse
Affiliation(s)
- Laura Pasin
- Institute of Anesthesia and Intensive Care, Azienda Ospedaliera Universitaria di Padova, Padova (Italy)
| | - Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy, and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Navalesi
- Institute of Anesthesia and Intensive Care, Azienda Ospedaliera Universitaria di Padova, Padova (Italy); Department of Medicine (DIMED), University of Padova, Padova (Italy)
| | - Nicolò Sella
- Institute of Anesthesia and Intensive Care, Azienda Ospedaliera Universitaria di Padova, Padova (Italy)
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan (Italy).
| | - Andrey G Yavorovskiy
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian
| | - Valery V Likhvantsev
- V. Negovsky Reanimatology Research Institute, Petrovka str, 25, b.2, Moscow, Russia; Department of Anesthesiology and Intensive Care, First Moscow State Medical University, Moscow, Russia
| | - Alberto Zangrillo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan (Italy)
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy, and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Faculty of Medicine, Vita-Salute San Raffaele University, Milan (Italy)
| | - Giacomo Monti
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan (Italy)
| |
Collapse
|
48
|
Randomized Trial of Interleukin-6 Receptor Inhibition in Patients With Acute ST-Segment Elevation Myocardial Infarction. J Am Coll Cardiol 2021; 77:1845-1855. [PMID: 33858620 DOI: 10.1016/j.jacc.2021.02.049] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/11/2021] [Accepted: 02/22/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Prompt myocardial revascularization with percutaneous coronary intervention (PCI) reduces infarct size and improves outcomes in patients with ST-segment elevation myocardial infarction (STEMI). However, as much as 50% of the loss of viable myocardium may be attributed to the reperfusion injury and the associated inflammatory response. OBJECTIVES This study sought to evaluate the effect of the interleukin-6 receptor inhibitor tocilizumab on myocardial salvage in acute STEMI. METHODS The ASSAIL-MI trial was a randomized, double-blind, placebo-controlled trial conducted at 3 high-volume PCI centers in Norway. Patients admitted with STEMI within 6 h of symptom onset were eligible. Consenting patients were randomized in a 1:1 fashion to promptly receive a single infusion of 280 mg tocilizumab or placebo. The primary endpoint was the myocardial salvage index as measured by magnetic resonance imaging after 3 to 7 days. RESULTS We randomized 101 patients to tocilizumab and 98 patients to placebo. The myocardial salvage index was larger in the tocilizumab group than in the placebo group (adjusted between-group difference 5.6 [95% confidence interval: 0.2 to 11.3] percentage points, p = 0.04). Microvascular obstruction was less extensive in the tocilizumab arm, but there was no significant difference in the final infarct size between the tocilizumab arm and the placebo arm (7.2% vs. 9.1% of myocardial volume, p = 0.08). Adverse events were evenly distributed across the treatment groups. CONCLUSIONS Tocilizumab increased myocardial salvage in patients with acute STEMI. (ASSessing the effect of Anti-IL-6 treatment in Myocardial Infarction [ASSAIL-MI]; NCT03004703).
Collapse
|
49
|
Liberale L, Ministrini S, Carbone F, Camici GG, Montecucco F. Cytokines as therapeutic targets for cardio- and cerebrovascular diseases. Basic Res Cardiol 2021; 116:23. [PMID: 33770265 PMCID: PMC7997823 DOI: 10.1007/s00395-021-00863-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Despite major advances in prevention and treatment, cardiac and cerebral atherothrombotic complications still account for substantial morbidity and mortality worldwide. In this context, inflammation is involved in the chronic process leading atherosclerotic plaque formation and its complications, as well as in the maladaptive response to acute ischemic events. For this reason, modulation of inflammation is nowadays seen as a promising therapeutic strategy to counteract the burden of cardio- and cerebrovascular disease. Being produced and recognized by both inflammatory and vascular cells, the complex network of cytokines holds key functions in the crosstalk of these two systems and orchestrates the progression of atherothrombosis. By binding to membrane receptors, these soluble mediators trigger specific intracellular signaling pathways eventually leading to the activation of transcription factors and a deep modulation of cell function. Both stimulatory and inhibitory cytokines have been described and progressively reported as markers of disease or interesting therapeutic targets in the cardiovascular field. Nevertheless, cytokine inhibition is burdened by harmful side effects that will most likely prevent its chronic use in favor of acute administrations in well-selected subjects at high risk. Here, we summarize the current state of knowledge regarding the modulatory role of cytokines on atherosclerosis, myocardial infarction, and stroke. Then, we discuss evidence from clinical trials specifically targeting cytokines and the potential implication of these advances into daily clinical practice.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952, Schlieren, Switzerland. .,First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.
| | - Stefano Ministrini
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952, Schlieren, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
50
|
Weber ANR. Targeting the NLRP3 Inflammasome via BTK. Front Cell Dev Biol 2021; 9:630479. [PMID: 33718366 PMCID: PMC7947255 DOI: 10.3389/fcell.2021.630479] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/14/2021] [Indexed: 12/28/2022] Open
Abstract
The NLRP3 inflammasome represents a critical inflammatory machinery driving pathology in many acute (e. g., myocardial infarction or stroke) and chronic (Alzheimer's disease, atherosclerosis) human disorders linked to the activity of IL-1 cytokines. Although the therapeutic potential of NLRP3 is undisputed, currently no clinically approved therapies exist to target the NLRP3 inflammasome directly. The recent discovery of BTK as a direct and positive regulator of the NLRP3 inflammasome has, however, raised the intriguing possibility of targeting the NLRP3 inflammasome via existing or future BTK inhibitors. Here, I review the mechanistic basis for this notion and discuss the molecular and cellular role of BTK in the inflammasome process. Specific attention will be given to cell-type dependent characteristics and differences that may be relevant for targeting approaches. Furthermore, I review recent (pre-)clinical evidence for effects of BTK inhibitors on NLRP3 activity and highlight and discuss open questions and future research directions. Collectively, the concept of targeting BTK to target NLRP3-dependent inflammation will be explored comprehensively at the molecular, cellular and therapeutic levels.
Collapse
Affiliation(s)
- Alexander N. R. Weber
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- iFIT – Cluster of Excellence (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- CMFI – Cluster of Excellence (EXC 2124) “Controlling Microbes to Fight Infection”, University of Tübingen, Tübingen, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK; German Cancer Consortium), Partner Site Tübingen, Department of Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|