1
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
2
|
Xu H, Fu J, Tu Q, Shuai Q, Chen Y, Wu F, Cao Z. The SGLT2 inhibitor empagliflozin attenuates atherosclerosis progression by inducing autophagy. J Physiol Biochem 2024; 80:27-39. [PMID: 37792168 DOI: 10.1007/s13105-023-00974-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/10/2023] [Indexed: 10/05/2023]
Abstract
Cardiovascular disease due to atherosclerosis is one of the leading causes of death worldwide; however, the underlying mechanism has yet to be defined. The sodium-dependent glucose transporter 2 inhibitor (SGLT2i) empagliflozin is a new type of hypoglycemic drug. Recent studies have shown that empagliflozin not only reduces high glucose levels but also exerts cardiovascular-protective effects and slows the process of atherosclerosis. The purpose of this study was to elucidate the mechanism by which empagliflozin ameliorates atherosclerosis. Male apolipoprotein E-deficient (ApoE-/-) mice were fed a high-fat Western diet to establish an atherosclerosis model. The area and size of atherosclerotic lesions in ApoE-/- mice were then assessed by performing hematoxylin-eosin (HE) staining after empagliflozin treatment. Concurrently, oxidized low-density lipoprotein (oxLDL) was used to mimic atherosclerosis in three different types of cells. Then, following empagliflozin treatment of macrophage cells (RAW264.7), human aortic smooth muscle cells (HASMCs), and human umbilical vein endothelial cells (HUVECs), western blotting was applied to measure the levels of autophagy-related proteins and proinflammatory cytokines, and green fluorescent protein (GFP)-light chain 3 (LC3) puncta were detected using confocal microscopy to confirm autophagosome formation. Oil Red O staining was performed to detect the foaming of macrophages and HASMCs, and flow cytometry was used for the cell cycle analysis. 5-ethynyl-2'-deoxyuridine (EdU), cell counting kit-8 (CCK-8), and scratch assays were also performed to examine the proliferation and migration of HASMCs. Empagliflozin suppressed the progression of atherosclerotic lesions in ApoE-/- mice. Empagliflozin also induced autophagy in RAW246.7 cells, HASMCs, and HUVECs via the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway, and it significantly increased the levels of the Beclin1 protein, the LC3B-II/I ratio, and p-AMPK protein. In addition, empagliflozin decreased the expression of P62 and the protein levels of inflammatory cytokines, and it inhibited the foaming of RAW246.7 cells and HASMCs, as well as the expression of inflammatory factors by inducing autophagy. Empagliflozin activated autophagy through the AMPK signaling pathway to delay the progression of atherosclerosis. Furthermore, the results of flow cytometry, EdU assays, CCK-8 cell viability assays, and scratch assays indicated that empagliflozin blocked HASMCs proliferation and migration. Empagliflozin activates autophagy through the AMPK signaling pathway to delay the evolution of atherosclerosis, indicating that it may represent a new and effective drug for the clinical treatment of atherosclerosis.
Collapse
Affiliation(s)
- Hualin Xu
- Postgraduate Training Basement of Jinzhou Medical University, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Jie Fu
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Qiang Tu
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Qingyun Shuai
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Yizhi Chen
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Fuyun Wu
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| | - Zheng Cao
- Postgraduate Training Basement of Jinzhou Medical University, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| |
Collapse
|
3
|
Buske M, Desch S, Heusch G, Rassaf T, Eitel I, Thiele H, Feistritzer HJ. Reperfusion Injury: How Can We Reduce It by Pre-, Per-, and Postconditioning. J Clin Med 2023; 13:159. [PMID: 38202166 PMCID: PMC10779793 DOI: 10.3390/jcm13010159] [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: 11/28/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
While early coronary reperfusion via primary percutaneous coronary intervention (pPCI) is established as the most efficacious therapy for minimizing infarct size (IS) in acute ST-elevation myocardial infarction (STEMI), the restoration of blood flow also introduces myocardial ischemia-reperfusion injury (IRI), leading to cardiomyocyte death. Among diverse methods, ischemic conditioning (IC), achieved through repetitive cycles of ischemia and reperfusion, has emerged as the most promising method to mitigate IRI. IC can be performed by applying the protective stimulus directly to the affected myocardium or indirectly to non-affected tissue, which is known as remote ischemic conditioning (RIC). In clinical practice, RIC is often applied by serial inflations and deflations of a blood pressure cuff on a limb. Despite encouraging preclinical studies, as well as clinical studies demonstrating reductions in enzymatic IS and myocardial injury on imaging, the observed impact on clinical outcome has been disappointing so far. Nevertheless, previous studies indicate a potential benefit of IC in high-risk STEMI patients. Additional research is needed to evaluate the impact of IC in such high-risk cohorts. The objective of this review is to summarize the pathophysiological background and preclinical and clinical data of IRI reduction by IC.
Collapse
Affiliation(s)
- Maria Buske
- Department of Cardiology, Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, 04289 Leipzig, Germany; (M.B.); (S.D.)
| | - Steffen Desch
- Department of Cardiology, Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, 04289 Leipzig, Germany; (M.B.); (S.D.)
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, 45122 Essen, Germany;
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany;
| | - Ingo Eitel
- Medical Clinic II, Clinic for Cardiology, Angiology and Intensive Care Medicine, University Heart Center Lübeck, 23538 Lübeck, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23538 Lübeck, Germany
| | - Holger Thiele
- Department of Cardiology, Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, 04289 Leipzig, Germany; (M.B.); (S.D.)
| | - Hans-Josef Feistritzer
- Department of Cardiology, Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, 04289 Leipzig, Germany; (M.B.); (S.D.)
| |
Collapse
|
4
|
Bergman I, Boyle D, Braver O, Gelikas S, Wexler Y, Omelchenko A, Assali A, Nussinovitch U. Ischemic Postconditioning Confers No Benefit to Left Ventricular Systolic Function: A Meta-Analysis of Cardiac Magnetic Resonance Imaging Results. Am J Cardiol 2023; 208:126-133. [PMID: 37837795 DOI: 10.1016/j.amjcard.2023.09.039] [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: 07/10/2023] [Revised: 09/05/2023] [Accepted: 09/09/2023] [Indexed: 10/16/2023]
Abstract
Ischemic postconditioning (IPoC) is a technique suggested to reduce reperfusion injury in patients suffering acute ST-elevation myocardial infarction (STEMI), although its use is highly controversial. This meta-analysis aimed to evaluate the effect of IPoC with percutaneous coronary intervention in patients with acute STEMI, as measured by follow-up left ventricular ejection fraction (LVEF) on cardiac magnetic resonance imaging. The investigators searched PubMed, Embase, and Web of Science for all randomized controlled trials published during the last 2 decades. After the removal of duplicates, 2,021 articles from online databases had been identified using relevant search criteria. The included randomized controlled trials had studied patients with acute STEMI and Thrombolysis in Myocardial Infarction flow 0 to 1 at presentation and had measured follow-up LVEF using cardiac magnetic resonance imaging. Overall, 11 studies (n = 1,339 patients) qualified for inclusion. In each study, the control group did not differ significantly from the experimental group. The pooled data from included studies were analyzed using standardized mean difference between IPoC and control groups, and the 95% confidence interval for LVEF; the results were visualized using a forest plot. Bivariate regression analyses and 1-way analyses of LVEF coefficient ratios were done to isolate for various clinical and procedural parameters. An analysis of pooled data of the IPoC (n = 674) and control (n = 665) groups showed that IPoC did not significantly impact follow-up LVEF (using standardized mean difference 0.10, 95% confidence interval 0.00 to 0.21). Further analysis showed that IPoC did not improve follow-up LVEF when isolating for relevant clinical and procedural parameters. In conclusion, the use of IPoC as an adjunctive therapy to percutaneous coronary intervention seemingly provides no benefit to left ventricular systolic function, as quantified with cardiac magnetic resonance imaging, in patients with acute STEMI with Thrombolysis in Myocardial Infarction flow 0 to 1.
Collapse
Affiliation(s)
- Idan Bergman
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Rabin Medical Center, Petach Tikva, Israel
| | | | - Omri Braver
- Department of Cardiology, Barzilai Medical Center, Ashkelon, Israel
| | - Shaul Gelikas
- The Trauma and Combat Medicine Branch, Surgeon General's Headquarters, Israel Defense Forces, Ramat Gan, Israel
| | - Yehuda Wexler
- Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Alexander Omelchenko
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Cardiology, Meir Medical Center, Kfar Saba, Israel
| | - Abid Assali
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Cardiology, Meir Medical Center, Kfar Saba, Israel
| | - Udi Nussinovitch
- Heart Institute at the Edith Wolfson Medical Center, Holon, Israel.
| |
Collapse
|
5
|
Heusch G, Andreadou I, Bell R, Bertero E, Botker HE, Davidson SM, Downey J, Eaton P, Ferdinandy P, Gersh BJ, Giacca M, Hausenloy DJ, Ibanez B, Krieg T, Maack C, Schulz R, Sellke F, Shah AM, Thiele H, Yellon DM, Di Lisa F. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol 2023; 67:102894. [PMID: 37839355 PMCID: PMC10590874 DOI: 10.1016/j.redox.2023.102894] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The present review summarizes the beneficial and detrimental roles of reactive oxygen species in myocardial ischemia/reperfusion injury and cardioprotection. In the first part, the continued need for cardioprotection beyond that by rapid reperfusion of acute myocardial infarction is emphasized. Then, pathomechanisms of myocardial ischemia/reperfusion to the myocardium and the coronary circulation and the different modes of cell death in myocardial infarction are characterized. Different mechanical and pharmacological interventions to protect the ischemic/reperfused myocardium in elective percutaneous coronary interventions and coronary artery bypass grafting, in acute myocardial infarction and in cardiotoxicity from cancer therapy are detailed. The second part keeps the focus on ROS providing a comprehensive overview of molecular and cellular mechanisms involved in ischemia/reperfusion injury. Starting from mitochondria as the main sources and targets of ROS in ischemic/reperfused myocardium, a complex network of cellular and extracellular processes is discussed, including relationships with Ca2+ homeostasis, thiol group redox balance, hydrogen sulfide modulation, cross-talk with NAPDH oxidases, exosomes, cytokines and growth factors. While mechanistic insights are needed to improve our current therapeutic approaches, advancements in knowledge of ROS-mediated processes indicate that detrimental facets of oxidative stress are opposed by ROS requirement for physiological and protective reactions. This inevitable contrast is likely to underlie unsuccessful clinical trials and limits the development of novel cardioprotective interventions simply based upon ROS removal.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert Bell
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Edoardo Bertero
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genova, Genova, Italy
| | - Hans-Erik Botker
- Department of Cardiology, Institute for Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - James Downey
- Department of Physiology, University of South Alabama, Mobile, AL, USA
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Heart Centre, Charterhouse Square, London, United Kingdom
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, National Heart Research Institute Singapore, National Heart Centre, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), IIS-Fundación Jiménez Díaz University Hospital, and CIBERCV, Madrid, Spain
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig -Universität, Giessen, Germany
| | - Frank Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Fabio Di Lisa
- Dipartimento di Scienze Biomediche, Università degli studi di Padova, Padova, Italy.
| |
Collapse
|
6
|
Algoet M, Janssens S, Himmelreich U, Gsell W, Pusovnik M, Van den Eynde J, Oosterlinck W. Myocardial ischemia-reperfusion injury and the influence of inflammation. Trends Cardiovasc Med 2023; 33:357-366. [PMID: 35181472 DOI: 10.1016/j.tcm.2022.02.005] [Citation(s) in RCA: 145] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 12/20/2022]
Abstract
Acute myocardial infarction is caused by a sudden coronary artery occlusion and leads to ischemia in the corresponding myocardial territory which generally results in myocardial necrosis. Without restoration of coronary perfusion, myocardial scar formation will cause adverse remodelling of the myocardium and heart failure. Successful introduction of percutaneous coronary intervention and surgical coronary artery bypass grafting made it possible to achieve early revascularisation/reperfusion, hence limiting the ischemic zone of myocardium. However, reperfusion by itself paradoxically triggers an exacerbated and accelerated injury in the myocardium, called ischemia-reperfusion (I/R) injury. This mechanism is partially driven by inflammation through multiple interacting pathways. In this review we summarize the current insights in mechanisms of I/R injury and the influence of altered inflammation. Multiple pharmacological and interventional therapeutic strategies (ischemic conditioning) have proven to be beneficial during I/R in preclinical models but were notoriously unsuccessful upon clinical translation. In this review we focus on common mechanisms of I/R injury, altered inflammation and potential therapeutic strategies. We hypothesize that a dual approach may be of value because I/R injury patients are predestined with multiple comorbidities and systemic low-grade inflammation, which requires targeted intervention before other strategies can be fully effective.
Collapse
Affiliation(s)
- Michiel Algoet
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.
| | - Stefan Janssens
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Matic Pusovnik
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jef Van den Eynde
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Helen B. Taussig Heart Center, The Johns Hopkins Hospital and School of Medicine, Baltimore, United States
| | | |
Collapse
|
7
|
A Novel Reperfusion Strategy for Primary Percutaneous Coronary Intervention in Patients with Acute ST-Segment Elevation Myocardial Infarction: A Prospective Case Series. J Clin Med 2023; 12:jcm12020433. [PMID: 36675362 PMCID: PMC9864309 DOI: 10.3390/jcm12020433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Ischemia reperfusion injury (IRI) remains a major problem in patients with acute ST-segment elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (PCI). We have developed a novel reperfusion strategy for PCI and named it "volume-controlled reperfusion (VCR)". The aim of the current study was to assess the safety and feasibility of VCR in patients with STEMI. METHODS Consecutive patients admitted to Beijing Chaoyang Hospital with STEMI were prospectively enrolled. The feasibility endpoint was procedural success. The safety endpoints included death from all causes, major vascular complications, and major adverse cardiac event (MACE), i.e., a composite of cardiac death, myocardial reinfarction, target vessel revascularization (TVR), and heart failure. RESULTS A total of 30 patients were finally included. Procedural success was achieved in 28 (93.3%) patients. No patients died during the study and no major vascular complications or MACE occurred during hospitalization. With the exception of one patient (3.3%) who underwent TVR three months after discharge, no patient encountered death (0.0%), major vascular complications (0.0%), or and other MACEs (0.0%) during the median follow-up of 16 months. CONCLUSION The findings of the pilot study suggest that VCR has favorable feasibility and safety in patients with STEMI. Further larger randomized trials are required to evaluate the effectiveness of VCR in STEMI patients.
Collapse
|
8
|
Ferdinandy P, Andreadou I, Baxter GF, Bøtker HE, Davidson SM, Dobrev D, Gersh BJ, Heusch G, Lecour S, Ruiz-Meana M, Zuurbier CJ, Hausenloy DJ, Schulz R. Interaction of Cardiovascular Nonmodifiable Risk Factors, Comorbidities and Comedications With Ischemia/Reperfusion Injury and Cardioprotection by Pharmacological Treatments and Ischemic Conditioning. Pharmacol Rev 2023; 75:159-216. [PMID: 36753049 PMCID: PMC9832381 DOI: 10.1124/pharmrev.121.000348] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/07/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
Preconditioning, postconditioning, and remote conditioning of the myocardium enhance the ability of the heart to withstand a prolonged ischemia/reperfusion insult and the potential to provide novel therapeutic paradigms for cardioprotection. While many signaling pathways leading to endogenous cardioprotection have been elucidated in experimental studies over the past 30 years, no cardioprotective drug is on the market yet for that indication. One likely major reason for this failure to translate cardioprotection into patient benefit is the lack of rigorous and systematic preclinical evaluation of promising cardioprotective therapies prior to their clinical evaluation, since ischemic heart disease in humans is a complex disorder caused by or associated with cardiovascular risk factors and comorbidities. These risk factors and comorbidities induce fundamental alterations in cellular signaling cascades that affect the development of ischemia/reperfusion injury and responses to cardioprotective interventions. Moreover, some of the medications used to treat these comorbidities may impact on cardioprotection by again modifying cellular signaling pathways. The aim of this article is to review the recent evidence that cardiovascular risk factors as well as comorbidities and their medications may modify the response to cardioprotective interventions. We emphasize the critical need for taking into account the presence of cardiovascular risk factors as well as comorbidities and their concomitant medications when designing preclinical studies for the identification and validation of cardioprotective drug targets and clinical studies. This will hopefully maximize the success rate of developing rational approaches to effective cardioprotective therapies for the majority of patients with multiple comorbidities. SIGNIFICANCE STATEMENT: Ischemic heart disease is a major cause of mortality; however, there are still no cardioprotective drugs on the market. Most studies on cardioprotection have been undertaken in animal models of ischemia/reperfusion in the absence of comorbidities; however, ischemic heart disease develops with other systemic disorders (e.g., hypertension, hyperlipidemia, diabetes, atherosclerosis). Here we focus on the preclinical and clinical evidence showing how these comorbidities and their routine medications affect ischemia/reperfusion injury and interfere with cardioprotective strategies.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Ioanna Andreadou
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gary F Baxter
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Hans Erik Bøtker
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Sean M Davidson
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Dobromir Dobrev
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Bernard J Gersh
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gerd Heusch
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Sandrine Lecour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Marisol Ruiz-Meana
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Coert J Zuurbier
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Derek J Hausenloy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| |
Collapse
|
9
|
Gao W, Zhong X, Ma Y, Huang D, Wang R, Zhao S, Yang S, Qian J, Ge J. A randomized multicenter trial to evaluate early invasive strategy for patients with acute ST-segment elevation myocardial infarction presenting 24-48 hours from symptom onset: Protocol of the RESCUE-MI study. Am Heart J 2022; 251:54-60. [PMID: 35525262 DOI: 10.1016/j.ahj.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND For ST-segment elevation myocardial infarction (STEMI) patients presenting 24 to 48 hours from symptom onset, whether early invasive strategy should be performed still remains controversial. METHODS This is a prospective, open-label, multicenter, investigator initiated, randomized controlled trial (NCT04962178) to evaluate the efficacy of early invasive strategy for STEMI patients within 24 to 48 hours of symptom onset. A total of 366 patients will be included from 10 hospitals in mainland China. They will be randomly (1:1) divided into 2 groups: the early invasive strategy group (primary percutaneous coronary intervention, PPCI) and conservative strategy group (optimal medical therapy with primary PCI not performed). All patients will be followed for 1 month. The primary end point is myocardial infarction size on cardiac magnetic resonance (CMR). The secondary end points are as follows: (1) major adverse cardiovascular events (MACE), which is defined as a composite of cardiac death, recurrent myocardial infarction, ischemic driven target vessel revascularization and stroke; (2) other CMR end points, including microvascular obstruction, intramyocardial hemorrhage, myocardial area at risk, left ventricular ejection fraction, left ventricular end diastolic volume and left ventricular end systolic volume. DISCUSSION This study is designed to evaluate the efficacy of early invasive strategy for STEMI patients within 24 to 48 hours of symptom onset and will add more evidence for clinical practice. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04962178. Registered on July 14, 2021.
Collapse
Affiliation(s)
- Wei Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xin Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yuanji Ma
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Dong Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Ruochen Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Shihai Zhao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Shan Yang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China.
| |
Collapse
|
10
|
Cardioprotective Effects of Physical Activity: Focus on Ischemia and Reperfusion. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2022. [DOI: 10.2478/sjecr-2022-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
This review aimed to revisit the old and introduce some of the new various cardioprotective effects of physical exercise, focusing on ischemia-reperfusion injury. A wealth of data shows that regular physical exercise is necessary to prevent cardiovascular diseases. In the last few years, a number of new training regimes, usually modified variations of high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) protocols, have been rising in popularity among people of all ages. Since exercising is not limited to only healthy people, our study emphasized the benefits of HIIT and MICT in preventing or mitigating cardiac ischemia-reperfusion injury. Different kinds of research are being performed, studying the various positive and side effects of these training regimes, all in hopes of finding the most optimal ones. So far, all of them have shown that exercising to any extent, even for a short period of time, is beneficial in one way or another, and outweighs the possible risks it might have. We also revisited some of the known molecular mechanisms responsible for many of the effects of physical exercise and introduced some new findings related to them. Lastly, we summarized and compared the benefits of different HIIT and MICT protocols to narrow down the search for the most efficient training method.
Collapse
|
11
|
Penna C, Comità S, Tullio F, Alloatti G, Pagliaro P. Challenges facing the clinical translation of cardioprotection: 35 years after the discovery of ischemic preconditioning. Vascul Pharmacol 2022; 144:106995. [PMID: 35470102 DOI: 10.1016/j.vph.2022.106995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/17/2022] [Accepted: 04/16/2022] [Indexed: 12/19/2022]
|
12
|
Mewton N, Roubille F, Bresson D, Prieur C, Bouleti C, Bochaton T, Ivanes F, Dubreuil O, Biere L, Hayek A, Derimay F, Akodad M, Alos B, Haider L, El Jonhy N, Daw R, De Bourguignon C, Dhelens C, Finet G, Bonnefoy-Cudraz E, Bidaux G, Boutitie F, Maucort-Boulch D, Croisille P, Rioufol G, Prunier F, Angoulvant D. Effect of Colchicine on Myocardial Injury in Acute Myocardial Infarction. Circulation 2021; 144:859-869. [PMID: 34420373 PMCID: PMC8462445 DOI: 10.1161/circulationaha.121.056177] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Inflammation is a key factor of myocardial damage in reperfused ST-segment–elevation myocardial infarction. We hypothesized that colchicine, a potent anti-inflammatory agent, may reduce infarct size (IS) and left ventricular (LV) remodeling at the acute phase of ST-segment–elevation myocardial infarction. Methods: In this double-blind multicenter trial, we randomly assigned patients admitted for a first episode of ST-segment–elevation myocardial infarction referred for primary percutaneous coronary intervention to receive oral colchicine (2-mg loading dose followed by 0.5 mg twice a day) or matching placebo from admission to day 5. The primary efficacy outcome was IS determined by cardiac magnetic resonance imaging at 5 days. The relative LV end-diastolic volume change at 3 months and IS at 3 months assessed by cardiac magnetic resonance imaging were among the secondary outcomes. Results: We enrolled 192 patients, 101 in the colchicine group and 91 in the control group. At 5 days, the gadolinium enhancement–defined IS did not differ between the colchicine and placebo groups with a mean of 26 interquartile range (IQR) [16–44] versus 28.4 IQR [14–40] g of LV mass, respectively (P=0.87). At 3 months follow-up, there was no significant difference in LV remodeling between the colchicine and placebo groups with a +2.4% (IQR, –8.3% to 11.1%) versus –1.1% (IQR, –8.0% to 9.9%) change in LV end-diastolic volume (P=0.49). Infarct size at 3 months was also not significantly different between the colchicine and placebo groups (17 IQR [10–28] versus 18 IQR [10–27] g of LV mass, respectively; P=0.92). The incidence of gastrointestinal adverse events during the treatment period was greater with colchicine than with placebo (34% versus 11%, respectively; P=0.0002). Conclusions: In this randomized, placebo-controlled trial, oral administration of high-dose colchicine at the time of reperfusion and for 5 days did not reduce IS assessed by cardiac magnetic resonance imaging. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03156816.
Collapse
Affiliation(s)
- Nathan Mewton
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - François Roubille
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Cardiology Department, CHU de Montpellier, France (F.R., M.A.)
| | - Didier Bresson
- Cardiology Division, University Hospital of Mulhouse, Hôpital Emile Muller, Mulhouse, France (D.B.)
| | - Cyril Prieur
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - Claire Bouleti
- Université de Poitiers, CIC Inserm 1402n CHU de Poitiers, France (C.B., B.A.)
| | - Thomas Bochaton
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - Fabrice Ivanes
- Cardiology Department CHRU de Tours and EA4245 T2i Tours University, France (F.I., D.A.)
| | - Olivier Dubreuil
- Centre Hospitalier Saint-Joseph Saint-Luc, Invasive Cardiology Department, Lyon, France (O.D.)
| | - Loïc Biere
- Institut MITOVASC, CNRS 6015 INSERM U1083, Université d'Angers, Cardiology Division, CHU Angers, France (L.B., F.P.)
| | - Ahmad Hayek
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - François Derimay
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - Mariama Akodad
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Cardiology Department, CHU de Montpellier, France (F.R., M.A.)
| | - Benjamin Alos
- Université de Poitiers, CIC Inserm 1402n CHU de Poitiers, France (C.B., B.A.)
| | - Lamis Haider
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - Naoual El Jonhy
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - Rachel Daw
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - Charles De Bourguignon
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - Carole Dhelens
- Pharmacy Department, FRIPHARM-RC (C.D.), Hospices Civils de Lyon, France
| | - Gérard Finet
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - Eric Bonnefoy-Cudraz
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | | | - Florent Boutitie
- UMR 5558 CNRS UCBL Biostatistics Departement (F.B., D.M.-B.), Hospices Civils de Lyon, France.,INSERM CarMeN 1060, IRIS Team, Claude Bernard University, Lyon, France (F.B.)
| | - Delphine Maucort-Boulch
- UMR 5558 CNRS UCBL Biostatistics Departement (F.B., D.M.-B.), Hospices Civils de Lyon, France
| | - Pierre Croisille
- CREATIS CNRS 5220 INSERM U1206 Research Lab, Radiology Department, University Hospital/CHU Saint Etienne, France (P.C.)
| | - Gilles Rioufol
- Hôpital Cardiovasculaire Louis Pradel, Clinical Investigation Center, INSERM 1407 and INSERM CarMeN 1060, Hospices Civils de Lyon and Claude Bernard University, Lyon, France (N.M., C.P., T.B., A.H., F.D., L.H., N.E.J, R.D., C.D.B., G.F., E.B.-C., G.R.)
| | - Fabrice Prunier
- Institut MITOVASC, CNRS 6015 INSERM U1083, Université d'Angers, Cardiology Division, CHU Angers, France (L.B., F.P.)
| | - Denis Angoulvant
- Cardiology Department CHRU de Tours and EA4245 T2i Tours University, France (F.I., D.A.)
| |
Collapse
|
13
|
Myocardial preservation during primary percutaneous intervention: It's time to rethink? Indian Heart J 2021; 73:395-403. [PMID: 34474749 PMCID: PMC8424360 DOI: 10.1016/j.ihj.2021.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
14
|
Mir T, Uddin M, Changal KH, Perveiz E, Kaur J, Sattar Y, Ullah W, Sheikh M. Long-term outcomes of ischemic post-conditioning primary PCI and conventional primary PCI in acute STEMI: a meta-analysis of randomized trials. Expert Rev Cardiovasc Ther 2021; 19:673-680. [PMID: 34115566 DOI: 10.1080/14779072.2021.1941874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Data regarding ischemic postconditioning during percutaneous coronary intervention (PCI) as compared conventional PCI alone has yielded conflicting results. METHODS Online databases comparing use of ischemic postconditioning percutaneous coronary intervention (ICP-PPCI) in STEMI patients with conventional PPCI were selected. Mortality, heart failure (HF), myocardial infarction (MI), and major adverse cardiac events (MACE) were evaluated. The primary outcome was composite of HF, MI, and mortality. Pooled risk ratio (RR) with 95% confidence interval (CI) were computed using random-effects model. RESULTS Eight studies consisting of 2,566 patients (ICP-PPCI n = 1,228; PPCI n = 1,278) were included. The mean age for PPCI group was 61.38 ± 7.86 years (51% men) and for PCI 59.83 ± 8.94 years (47% men). There were no differences in outcome between ICP-PPCI and PPCI in terms of HF (RR 0.87 95% CI0.51-1.48; p = 0.29), MI (RR 1.28, 95%CI0.74-2.20; p = 0.20), mortality (RR 0.93, 95%CI0.64-1.34; p = 0.58), and MACE (RR 0.89, 95%CI0.74-1.07; p = 0.22). The results for composite event for the ICP-PPCI and PPIC procedures, at ≥1 year follow-up duration, were comparable (RR 1.00 95%CI0.82-1.22; p = 1). CONCLUSION Ischemic postconditioning post percutaneous coronary intervention in STEMI patients has no long-term benefits over conventional PCI.
Collapse
Affiliation(s)
- Tanveer Mir
- Internal Medicine, Detroit Medical Center, Wayne State University, Detroit, MI, USA
| | - Mohammed Uddin
- Internal Medicine, Detroit Medical Center, Wayne State University, Detroit, MI, USA
| | | | - Eskara Perveiz
- Internal Medicine, Detroit Medical Center, Wayne State University, Detroit, MI, USA
| | - Jasmeet Kaur
- Department of Internal Medicine, Saint Joseph Mercy Oakland, Pontiac, MI, USA
| | - Yasar Sattar
- Internal Medicine, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Waqas Ullah
- Internal Medicine, Abington Jefferson Health, Abington, PA, USA
| | - Mujeeb Sheikh
- Department of Cardiovascular Medicine and Interventional Cardiology, Promedica Toledo Hospital, Ohio, USA
| |
Collapse
|
15
|
Taha HSED, Shaker MM. Percutaneous management of reperfusion arrhythmias during primary percutaneous coronary intervention: a case report. Egypt Heart J 2021; 73:30. [PMID: 33765245 PMCID: PMC7994471 DOI: 10.1186/s43044-021-00158-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/16/2021] [Indexed: 12/02/2022] Open
Abstract
Background Myocardial reperfusion may cause profound electrophysiological alterations and can lead to serious reperfusion arrhythmias (RA). Management of RA and the accompanying electrical storm that may occur remains a problem. To our knowledge, the role of balloon re-inflation of the infarct-related artery (IRA) has never been addressed as a treatment modality for RA presenting as ventricular tachycardia (VT) with pulse or supraventricular tachycardia (SVT). Case presentation Six patients presenting with ST elevation myocardial infarction (STEMI) in the first 12 h, who underwent successful primary percutaneous coronary intervention (PCI), developed RA in the cathlab after restoration of flow in the IRA. The RA was in the form of VT with pulse, except in one patient who had SVT. In four patients, the RA was associated with hemodynamic instability. The mean age of the studied patients was 59.16 ± 7.94 years, and four were males. Coronary artery disease risk factors were prevalent, with four patients being hypertensive, two dyslipidemic, one diabetic, and 2 current smokers. One patient had a history of prior myocardial infarction (MI), and none had a history of congestive heart failure. The coronary angiography showed 100% occlusion of IRA in all patients and 2–3-vessel disease was present in 50%. PCI was successful with restoration of thrombolysis in myocardial infarction (TIMI) 2–3 flow in IRA in all cases. The mean time to revascularization from the onset of chest pain was 4.88 ± 2.68 h. In all cases, balloon re-inflation was successful in terminating the arrhythmias. None of the patients needed direct current cardioversion or anti-arrhythmic drugs for management of the acute arrhythmia. Conclusion Balloon re-inflation of IRA was successful in terminating RA that develop in the form of VT with pulse or SVT.
Collapse
Affiliation(s)
- Hesham Salah El Din Taha
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562, Egypt.
| | - Mirna Mamdouh Shaker
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562, Egypt
| |
Collapse
|
16
|
Comparison of infarction size, complete ST-segment resolution incidence, mortality and re-infarction and target vessel revascularization between remote ischemic conditioning and ischemic postconditioning in ST-segment elevation myocardial infarction patients undergoing primary percutaneous coronary intervention. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2020; 16:278-286. [PMID: 33597992 PMCID: PMC7863805 DOI: 10.5114/aic.2020.99262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/06/2020] [Indexed: 11/30/2022] Open
Abstract
Introduction Due to higher morbidity and mortality, ST-segment elevation myocardial infarction (STEMI) causes many public health problems. Aim To observe effects of remote ischemic conditioning (RIC) and ischemic postconditioning (IPC) on patients diagnosed as STEMI undergoing primary percutaneous coronary intervention (pPCI). Material and methods This meta-analysis was conducted using indirect comparison by conducting a network meta-analysis (NMA). We conducted searches by utilizing PubMed and the other databases to identify randomized controlled trials (RCTs) that described IPC or RIC treated patients diagnosed with STEMI during processes of pPCI. Enzymatic infarct size and infarction size were evaluated and cardiac events were assessed during the follow-up. Results Pooled results showed that lower enzymatic infarction size was associated with the RIC group compared to the IPC group (IPC vs. RIC: standardized mean difference (SMD) = 1.126; 95% confidence interval (CI): 0.756–1.677). Compared with IPC, RIC significantly reduced infarction size, which was assessed using cardiac magnetic resonance (CMR) (SMD = 1.113; 95% CI: 0.674–1.837). We noted a potential toward greater complete ST-segment resolution in RIC patients compared with IPC patients (odds ratio (OR) = 0.821; 95% CI: 0.166–4.051). No significant difference existed in all-cause mortality (OR = 2.211; 95% CI: 0.845–5.784), Target vessel revascularization (TVR) (OR = 0.045; 95% CI: 0.001–.662) or re-infarction (OR = 1.763; 95% CI: 0.741–4.193). Conclusions This meta-analysis suggested RIC was correlated with significantly smaller infarction size compared to IPC. No significant superiority between RIC and IPC has been observed in this study on cSTR incidence, mortality and re-infarction or TVR.
Collapse
|
17
|
Pezel T, Viallon M, Croisille P, Sebbag L, Bochaton T, Garot J, Lima JAC, Mewton N. Imaging Interstitial Fibrosis, Left Ventricular Remodeling, and Function in Stage A and B Heart Failure. JACC Cardiovasc Imaging 2020; 14:1038-1052. [PMID: 32828781 DOI: 10.1016/j.jcmg.2020.05.036] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/11/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023]
Abstract
Myocardial interstitial fibrosis is part of the advanced disease stage of most cardiovascular pathologies. It has been characterized histologically in various disease settings from hypertensive heart disease and diabetic cardiomyopathy to severe aortic stenosis. It is also involved in the process of aging. In cardiovascular medicine, myocardial interstitial fibrosis is associated with several adverse outcomes, especially heart failure (HF) and sudden cardiac death. Until recently, clinical measures of interstitial fibrosis could only be made by invasive myocardial biopsy. The availability of cardiac magnetic resonance (CMR) T1 mapping techniques allows for the indirect measurement of interstitial space characteristics and extracellular volume size, which is closely correlated with collagen content and interstitial infiltration by amyloid and other molecules. There has been significant improvement in the accuracy and reproducibility of T1 acquisition sequences in the last decade; however, the correct use of this technique requires a solid CMR expertise in daily imaging practice. CMR has become the gold standard to assess left ventricular (LV) remodeling and functional features associated with interstitial fibrosis. These features can be detected in the early stages of HF. The main objective of this paper is to review the relevant results of preclinical and clinical observational studies that demonstrate the prognostic impact of interstitial fibrosis assessed by T1 mapping, as well as adverse left ventricular remodeling, as determinants of HF. Therefore, this review focuses on the pathological mechanisms underlying LV remodeling and interstitial fibrosis, in addition to the technical considerations involved in the assessment of interstitial LV fibrosis by CMR. It provides a thorough review of clinical evidence that demonstrates the association of interstitial fibrosis and other-CMR derived LV phenotypes with Stages A and B HF.
Collapse
Affiliation(s)
- Theo Pezel
- Department of Cardiology, Paris University, Lariboisiere Hospital, AP-HP, INSERM, UMRS 942, Paris, France; Division of Cardiology, Johns Hopkins University, Baltimore, Maryland
| | - Magalie Viallon
- University Lyon, UJM-Saint-Etienne, INSA, CNRS UMR 5520, INSERM U1206, CREATIS, Saint-Etienne, France
| | - Pierre Croisille
- University Lyon, UJM-Saint-Etienne, INSA, CNRS UMR 5520, INSERM U1206, CREATIS, Saint-Etienne, France
| | - Laurent Sebbag
- Heart Failure and Transplant Department, Hospices Civils de Lyon, Hôpital Louis Pradel, Bron, France
| | - Thomas Bochaton
- Hospices Civils de Lyon, Hôpital Louis Pradel, Cardiac Intensive Care Unit, Bron, France
| | - Jerome Garot
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay-Générale de Santé, Massy, France
| | - Joao A C Lima
- Division of Cardiology, Johns Hopkins University, Baltimore, Maryland
| | - Nathan Mewton
- Cardiovascular Hospital Louis Pradel, Clinical Investigation Center and Heart Failure Department, INSERM 1407, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France.
| |
Collapse
|
18
|
Chadet S, Ternant D, Roubille F, Bejan-Angoulvant T, Prunier F, Mewton N, Paintaud G, Ovize M, Dupuy AM, Angoulvant D, Ivanes F. Kinetic modelling of myocardial necrosis biomarkers offers an easier, reliable and more acceptable assessment of infarct size. Sci Rep 2020; 10:13597. [PMID: 32788683 PMCID: PMC7423884 DOI: 10.1038/s41598-020-70501-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/28/2020] [Indexed: 11/30/2022] Open
Abstract
Infarct size is a major prognostic factor in ST-segment elevation myocardial infarction (STEMI). It is often assessed using repeated blood sampling and the estimation of biomarker area under the concentration versus time curve (AUC) in translational research. We aimed at developing limited sampling strategies (LSS) to accurately estimate biomarker AUC using only a limited number of blood samples in STEMI patients. This retrospective study was carried out on pooled data from five clinical trials of STEMI patients (TIMI blood flow 0/1) studies where repeated blood samples were collected within 72 h after admission to assess creatine kinase (CK), cardiac troponin I (cTnI) and muscle-brain CK (CK-MB). Biomarker kinetics was assessed using previously described biomarker kinetic models. A number of LSS models including combinations of 1 to 3 samples were developed to identify sampling times leading to the best estimation of AUC. Patients were randomly assigned to either learning (2/3) or validation (1/3) subsets. Descriptive and predictive performances of LSS models were compared using learning and validation subsets, respectively. An external validation cohort was used to validate the model and its applicability to different cTnI assays, including high-sensitive (hs) cTnI. 132 patients had full CK and cTnI dataset, 49 patients had CK-MB. For each biomarker, 180 LSS models were tested. Best LSS models were obtained for the following sampling times: T4-16 for CK, T8-T20 for cTnI and T8-T16 for CK-MB for 2-sample LSS; and T4-T16-T24 for CK, T4-T12-T20 for cTnI and T8-T16-T20 for CK-MB for 3-sample LSS. External validation was achieved on 103 anterior STEMI patients (TIMI flow 0/1), and the cTnI model applicability to recommended hs cTnI confirmed. Biomarker kinetics can be assessed with a limited number of samples using kinetic modelling. This opens the way for substantial simplification of future cardioprotection studies, more acceptable for the patients.
Collapse
Affiliation(s)
- Stéphanie Chadet
- Loire Valley Cardiovascular Collaboration, Université de Tours, EA 4245 T2I & FHU SUPORT, Tours, France
| | - David Ternant
- Loire Valley Cardiovascular Collaboration, Université de Tours, EA 4245 T2I & FHU SUPORT, Tours, France
- Laboratory of Pharmacology-Toxicology, CHRU de Tours, Tours, France
| | - François Roubille
- Department of Cardiology, PhyMedExp, Université de Montpellier, INSERM U1046, CNRS UMR 9214, CHU de Montpellier, Montpellier, France
| | - Theodora Bejan-Angoulvant
- Loire Valley Cardiovascular Collaboration, Université de Tours, EA 4245 T2I & FHU SUPORT, Tours, France
- Department of Clinical Pharmacology, CHRU de Tours, Tours, France
| | | | - Nathan Mewton
- Université Claude Bernard Lyon 1, INSERM U1060 CarMeN, Lyon, France
| | - Gilles Paintaud
- Loire Valley Cardiovascular Collaboration, Université de Tours, EA 4245 T2I & FHU SUPORT, Tours, France
- Laboratory of Pharmacology-Toxicology, CHRU de Tours, Tours, France
| | - Michel Ovize
- Université Claude Bernard Lyon 1, INSERM U1060 CarMeN, Lyon, France
| | - Anne Marie Dupuy
- Department of Cardiology, PhyMedExp, Université de Montpellier, INSERM U1046, CNRS UMR 9214, CHU de Montpellier, Montpellier, France
| | - Denis Angoulvant
- Loire Valley Cardiovascular Collaboration, Université de Tours, EA 4245 T2I & FHU SUPORT, Tours, France
- Department of Cardiology & FACT, CHRU de Tours, Tours, France
| | - Fabrice Ivanes
- Loire Valley Cardiovascular Collaboration, Université de Tours, EA 4245 T2I & FHU SUPORT, Tours, France.
- Department of Cardiology & FACT, CHRU de Tours, Tours, France.
| |
Collapse
|
19
|
Abstract
Despite the increasing use and success of interventional coronary reperfusion strategies, morbidity and mortality from acute myocardial infarction are still substantial. Myocardial infarct size is a major determinant of prognosis in these patients. Therefore, cardioprotective strategies aim to reduce infarct size. However, a perplexing gap exists between the many preclinical studies reporting infarct size reduction with mechanical and pharmacological interventions and the poor translation into better clinical outcomes in patients. This Review revisits the pathophysiology of myocardial ischaemia-reperfusion injury, including the role of autophagy and forms of cell death such as necrosis, apoptosis, necroptosis and pyroptosis. Other cellular compartments in addition to cardiomyocytes are addressed, notably the coronary microcirculation. Preclinical and clinical research developments in mechanical and pharmacological approaches to induce cardioprotection, and their signal transduction pathways, are discussed. Additive cardioprotective interventions are advocated. For clinical translation into treatments for patients with acute myocardial infarction, who typically are of advanced age, have comorbidities and are receiving several medications, not only infarct size reduction but also attenuation of coronary microvascular obstruction, as well as longer-term targets including infarct repair and reverse remodelling, must be considered to improve patient outcomes. Future clinical trials must focus on patients who really need adjunct cardioprotection, that is, those with severe haemodynamic alterations.
Collapse
|
20
|
Meng Z, Gai W, Song D. Postconditioning with Nitrates Protects Against Myocardial Reperfusion Injury: A New Use for an Old Pharmacological Agent. Med Sci Monit 2020; 26:e923129. [PMID: 32516304 PMCID: PMC7299064 DOI: 10.12659/msm.923129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Early reperfusion remains the key therapy to salvage viable myocardium and must be applied as soon as possible following an acute myocardial infarction (AMI) to attenuate the ischemic insult. However, reperfusion injury may develop following reintroduction of blood and oxygen to vulnerable myocytes, which results in more severe cell death than in the preceding ischemic episode. Ischemic postconditioning (I-PostC) provides a cardioprotective effect in combination with pharmacological agents. Although nitrates have been tested in many experimental and clinical studies of acute AMI to evaluate the cardioprotective effect, few investigations have been focused on nitrates postconditioning in patients undergoing percutaneous coronary intervention (PCI). This review presents the manifestations of myocardial reperfusion injury (RI) and potential mechanisms underlying it, and provides the mechanisms involved in the cardioprotection of I-PostC. We also present a new therapeutic approach to attenuate RI by use of an ‘old’ agent – nitrates – in AMI patients.
Collapse
Affiliation(s)
- Zhu Meng
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland)
| | - Weili Gai
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland)
| | - Dalin Song
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland)
| |
Collapse
|
21
|
Management of No-Reflow. Microcirculation 2020. [DOI: 10.1007/978-3-030-28199-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
22
|
Traverse JH, Swingen CM, Henry TD, Fox J, Wang YL, Chavez IJ, Lips DL, Lesser JR, Pedersen WR, Burke NM, Pai A, Lindberg JL, Garberich RF. NHLBI-Sponsored Randomized Trial of Postconditioning During Primary Percutaneous Coronary Intervention for ST-Elevation Myocardial Infarction. Circ Res 2019; 124:769-778. [PMID: 30602360 DOI: 10.1161/circresaha.118.314060] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
RATIONALE Postconditioning at the time of primary percutaneous coronary intervention (PCI) for ST-segment-elevation myocardial infarction may reduce infarct size and improve myocardial salvage. However, clinical trials have shown inconsistent benefit. OBJECTIVE We performed the first National Heart, Lung, and Blood Institute-sponsored trial of postconditioning in the United States using strict enrollment criteria to optimize the early benefits of postconditioning and assess its long-term effects on left ventricular (LV) function. METHODS AND RESULTS We randomized 122 ST-segment-elevation myocardial infarction patients to postconditioning (4, 30 seconds PTCA [percutaneous transluminal coronary angioplasty] inflations/deflations)+PCI (n=65) versus routine PCI (n=57). All subjects had an occluded major epicardial artery (thrombolysis in myocardial infarction=0) with ischemic times between 1 and 6 hours with no evidence of preinfarction angina or collateral blood flow. Cardiac magnetic resonance imaging measured at 2 days post-PCI showed no difference between the postconditioning group and control in regards to infarct size (22.5±14.5 versus 24.0±18.5 g), myocardial salvage index (30.3±15.6% versus 31.5±23.6%), or mean LV ejection fraction. Magnetic resonance imaging at 12 months showed a significant recovery of LV ejection fraction in both groups (61.0±11.4% and 61.4±9.1%; P<0.01). Subjects randomized to postconditioning experienced more favorable remodeling over 1 year (LV end-diastolic volume =157±34 to 150±38 mL) compared with the control group (157±40 to 165±45 mL; P<0.03) and reduced microvascular obstruction ( P=0.05) on baseline magnetic resonance imaging and significantly less adverse LV remodeling compared with control subjects with microvascular obstruction ( P<0.05). No significant adverse events were associated with the postconditioning protocol and all patients but one (hemorrhagic stroke) survived through 1 year of follow-up. CONCLUSIONS We found no early benefit of postconditioning on infarct size, myocardial salvage index, and LV function compared with routine PCI. However, postconditioning was associated with improved LV remodeling at 1 year of follow-up, especially in subjects with microvascular obstruction. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov . Unique identifier: NCT01324453.
Collapse
Affiliation(s)
- Jay H Traverse
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.).,Cardiovascular Division, The University of Minnesota School of Medicine, Minneapolis (J.H.T., C.M.S.)
| | - Cory M Swingen
- Cardiovascular Division, The University of Minnesota School of Medicine, Minneapolis (J.H.T., C.M.S.)
| | - Timothy D Henry
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Jane Fox
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Yale L Wang
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Ivan J Chavez
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Daniel L Lips
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - John R Lesser
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Wesley R Pedersen
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Nicholas M Burke
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Akila Pai
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Jana L Lindberg
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Ross F Garberich
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| |
Collapse
|
23
|
Rios-Navarro C, Marcos-Garces V, Bayes-Genis A, Husser O, Nuñez J, Bodi V. Microvascular Obstruction in ST-Segment Elevation Myocardial Infarction: Looking Back to Move Forward. Focus on CMR. J Clin Med 2019; 8:E1805. [PMID: 31661823 PMCID: PMC6912395 DOI: 10.3390/jcm8111805] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/14/2022] Open
Abstract
After a myocardial infarction (MI), despite the resolution of the coronary occlusion, the deterioration of myocardial perfusion persists in a considerable number of patients. This phenomenon is known as microvascular obstruction (MVO). Initially, the focus was placed on re-establishing blood flow in the epicardial artery. Then, the observation that MVO has profound negative structural and prognostic repercussions revived interest in microcirculation. In the near future, the availability of co-adjuvant therapies (beyond timely coronary reperfusion) aimed at preventing, minimizing, and repairing MVOs and finding convincing answers to questions regarding what, when, how, and where to administer these therapies will be of utmost importance. The objective of this work is to review the state-of-the-art concepts on pathophysiology, diagnostic methods, and structural and clinical implications of MVOs in patients with ST-segment elevation MIs. Based on this knowledge we discuss previously-tested and future opportunities for the prevention and repair of MVO.
Collapse
Affiliation(s)
| | | | - Antoni Bayes-Genis
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain.
- Cardiology Department and Heart Failure Unit, Hospital Universitari Germans Trias i Pujol (Badalona) and Department of Medicine Universitat Autonoma de Barcelona, 08916 Barcelona, Spain.
| | - Oliver Husser
- Department of Cardiology, St-Johannes Hospital, 44137 Dortmund, Germany.
| | - Julio Nuñez
- Institute of Health Research INCLIVA, 46010 Valencia, Spain.
- Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain.
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain.
- Department of Medicine, Universidad de Valencia, 46010 Valencia, Spain.
| | - Vicente Bodi
- Institute of Health Research INCLIVA, 46010 Valencia, Spain.
- Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain.
- Centro de Investigación Biomédica en Red-Cardiovascular (CIBER-CV), 28029 Madrid, Spain.
- Department of Medicine, Universidad de Valencia, 46010 Valencia, Spain.
| |
Collapse
|
24
|
Heusch G. Coronary microvascular obstruction: the new frontier in cardioprotection. Basic Res Cardiol 2019; 114:45. [DOI: 10.1007/s00395-019-0756-8] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 12/16/2022]
|
25
|
Niccoli G, Montone RA, Ibanez B, Thiele H, Crea F, Heusch G, Bulluck H, Hausenloy DJ, Berry C, Stiermaier T, Camici PG, Eitel I. Optimized Treatment of ST-Elevation Myocardial Infarction. Circ Res 2019; 125:245-258. [PMID: 31268854 DOI: 10.1161/circresaha.119.315344] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Primary percutaneous coronary intervention is nowadays the preferred reperfusion strategy for patients with acute ST-segment-elevation myocardial infarction, aiming at restoring epicardial infarct-related artery patency and achieving microvascular reperfusion as early as possible, thus limiting the extent of irreversibly injured myocardium. Yet, in a sizeable proportion of patients, primary percutaneous coronary intervention does not achieve effective myocardial reperfusion due to the occurrence of coronary microvascular obstruction (MVO). The amount of infarcted myocardium, the so-called infarct size, has long been known to be an independent predictor for major adverse cardiovascular events and adverse left ventricular remodeling after myocardial infarction. Previous cardioprotection studies were mainly aimed at protecting cardiomyocytes and reducing infarct size. However, several clinical and preclinical studies have reported that the presence and extent of MVO represent another important independent predictor of adverse left ventricular remodeling, and recent evidences support the notion that MVO may be more predictive of major adverse cardiovascular events than infarct size itself. Although timely and complete reperfusion is the most effective way of limiting myocardial injury and subsequent ventricular remodeling, the translation of effective therapeutic strategies into improved clinical outcomes has been largely disappointing. Of importance, despite the presence of a large number of studies focused on infarct size, only few cardioprotection studies addressed MVO as a therapeutic target. In this review, we provide a detailed summary of MVO including underlying causes, diagnostic techniques, and current therapeutic approaches. Furthermore, we discuss the hypothesis that simultaneously addressing infarct size and MVO may help to translate cardioprotective strategies into improved clinical outcome following ST-segment-elevation myocardial infarction.
Collapse
Affiliation(s)
- Giampaolo Niccoli
- From the Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (G.N., R.A.M., F.C.).,Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy (G.N., F.C.)
| | - Rocco A Montone
- From the Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (G.N., R.A.M., F.C.)
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (B.I.).,Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain(B.I.).,CIBER de enfermedades CardioVasculares (CIBERCV), Madrid, Spain (B.I.)
| | - Holger Thiele
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig and Leipzig Heart Institute, Germany (H.T.)
| | - Filippo Crea
- From the Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (G.N., R.A.M., F.C.).,Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy (G.N., F.C.)
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Germany (G.H.)
| | - Heerajnarain Bulluck
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, United Kingdom (H.B., D.J.H.)
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, United Kingdom (H.B., D.J.H.).,Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School (D.J.H.).,National Heart Research Institute Singapore, National Heart Centre (D.J.H.).,Yong Loo Lin School of Medicine, National University Singapore (D.J.H.).,The Hatter Cardiovascular Institute, University College London, United Kingdom (D.J.H.).,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research and Development, United Kingdom (D.J.H.).,Department of Cardiology, Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Nuevo Leon, Mexico (D.J.H.)
| | - Colin Berry
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, United Kingdom (C.B.).,British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (C.B.)
| | - Thomas Stiermaier
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine) and German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany (T.S., I.E.)
| | - Paolo G Camici
- Vita-Salute University and San Raffaele Hospital, Milan, Italy (P.G.C.)
| | - Ingo Eitel
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine) and German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany (T.S., I.E.)
| |
Collapse
|
26
|
Relaxin mitigates microvascular damage and inflammation following cardiac ischemia-reperfusion. Basic Res Cardiol 2019; 114:30. [PMID: 31218471 DOI: 10.1007/s00395-019-0739-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 06/14/2019] [Indexed: 02/07/2023]
Abstract
Microvascular obstruction (MVO) and leakage (MVL) forms a pivotal part of microvascular damage following cardiac ischemia-reperfusion (IR). We tested the effect of relaxin therapy on MVO and MVL in mice following cardiac IR injury including severity of MVO and MVL, opening capillaries, infarct size, regional inflammation, cardiac function and remodelling, and permeability of cultured endothelial monolayer. Compared to vehicle group, relaxin treatment (50 μg/kg) reduced no-reflow area by 38% and the content of Evans blue as a permeability tracer by 56% in jeopardized myocardium (both P < 0.05), effects associated with increased opening capillaries. Relaxin also decreased leukocyte density, gene expression of cytokines, and mitigated IR-induced decrease in protein content of VE-cadherin and relaxin receptor. Infarct size was comparable between the two groups. At 2 weeks post-IR, relaxin treatment partially preserved cardiac contractile function and limited chamber dilatation versus untreated controls by echocardiography. Endothelial cell permeability assay demonstrated that relaxin attenuated leakage induced by hypoxia-reoxygenation, H2O2, or cytokines, action that was independent of nitric oxide but associated with the preservation of VE-cadherin. In conclusion, relaxin therapy attenuates IR-induced MVO and MVL and endothelial leakage. This protection was associated with reduced regional inflammatory responses and consequently led to alleviated adverse cardiac remodeling.
Collapse
|
27
|
Hausenloy DJ, Chilian W, Crea F, Davidson SM, Ferdinandy P, Garcia-Dorado D, van Royen N, Schulz R, Heusch G. The coronary circulation in acute myocardial ischaemia/reperfusion injury: a target for cardioprotection. Cardiovasc Res 2019; 115:1143-1155. [PMID: 30428011 PMCID: PMC6529918 DOI: 10.1093/cvr/cvy286] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 10/15/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022] Open
Abstract
The coronary circulation is both culprit and victim of acute myocardial infarction. The rupture of an epicardial atherosclerotic plaque with superimposed thrombosis causes coronary occlusion, and this occlusion must be removed to induce reperfusion. However, ischaemia and reperfusion cause damage not only in cardiomyocytes but also in the coronary circulation, including microembolization of debris and release of soluble factors from the culprit lesion, impairment of endothelial integrity with subsequently increased permeability and oedema formation, platelet activation and leucocyte adherence, erythrocyte stasis, a shift from vasodilation to vasoconstriction, and ultimately structural damage to the capillaries with eventual no-reflow, microvascular obstruction (MVO), and intramyocardial haemorrhage (IMH). Therefore, the coronary circulation is a valid target for cardioprotection, beyond protection of the cardiomyocyte. Virtually all of the above deleterious endpoints have been demonstrated to be favourably influenced by one or the other mechanical or pharmacological cardioprotective intervention. However, no-reflow is still a serious complication of reperfused myocardial infarction and carries, independently from infarct size, an unfavourable prognosis. MVO and IMH can be diagnosed by modern imaging technologies, but still await an effective therapy. The current review provides an overview of strategies to protect the coronary circulation from acute myocardial ischaemia/reperfusion injury. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.
Collapse
Affiliation(s)
- Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, UK
- The National Institute of Health Research, University College London Hospitals Biomedical Research Centre, Research & Development, London, UK
- Department of Cardiology, Barts Heart Centre, St Bartholomew’s Hospital, London, UK
| | - William Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, USA
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, F. Policlinico Gemelli—IRCCS, Università Cattolica Sacro Cuore, Roma, Italy
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - David Garcia-Dorado
- Department of Cardiology, Vascular Biology and Metabolism Area, Vall d’Hebron University Hospital and Research Institute (VHIR), Universitat Autónoma de Barcelona, Barcelona, Spain
- Instituto CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| |
Collapse
|
28
|
Lou B, Zhou C. Letter by Lou and Zhou Regarding Article, "NHLBI-Sponsored Randomized Trial of Postconditioning During Primary Percutaneous Coronary Intervention for ST-Elevation Myocardial Infarction". Circ Res 2019; 124:e55-e56. [PMID: 30973802 DOI: 10.1161/circresaha.119.314734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Baohui Lou
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Chenghui Zhou
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Coronary artery no-reflow phenomenon is an incidental outcome of percutaneous coronary intervention in patients presenting with acute myocardial infarction. Despite advances in pharmacologic and non-pharmacologic therapies, coronary no-reflow phenomenon occurs more commonly than desired. It often results in poor clinical outcomes and remains as a relevant consideration in the cardiac catheterization laboratory. In this systematic review, we have sought to discuss the topic in detail, and to relay the most recent discoveries and data on management of this condition. RECENT FINDINGS We discuss several pharmacologic and non-pharmacologic treatments used in the prevention and management of coronary no-reflow and microvascular obstruction. Covered topics include the understanding of pharmacologic mechanisms of current and future agents, and recent discoveries that may result in the development of future treatment options. We conclude that the pathophysiology of coronary no-reflow phenomenon and microvascular obstruction still remains incompletely understood, although several plausible theories have led to the current standard of care for its management. We also conclude that coronary no-reflow phenomenon and microvascular obstruction must be recognized as a multifactorial condition that has certain predispositions and characteristics, therefore its prevention and treatment must begin pre-procedurally and be multi-faceted including certain medications and operator techniques in the cardiac catheterization laboratory.
Collapse
Affiliation(s)
- Ahmadreza Karimianpour
- Department of Cardiovascular Diseases, Heart & Vascular Institute, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC, 29425, USA.
| | - Anbukarasi Maran
- Department of Cardiovascular Diseases, Heart & Vascular Institute, Medical University of South Carolina, 171 Ashley Ave, Charleston, SC, 29425, USA
| |
Collapse
|
30
|
Abstract
Rapid admission and acute interventional treatment combined with modern antithrombotic pharmacologic therapy have improved outcomes in patients with ST elevation myocardial infarction. The next major target to further advance outcomes needs to address ischemia-reperfusion injury, which may contribute significantly to the final infarct size and hence mortality and postinfarction heart failure. Mechanical conditioning strategies including local and remote ischemic pre-, per-, and postconditioning have demonstrated consistent cardioprotective capacities in experimental models of acute ischemia-reperfusion injury. Their translation to the clinical scenario has been challenging. At present, the most promising mechanical protection strategy of the heart seems to be remote ischemic conditioning, which increases myocardial salvage beyond acute reperfusion therapy. An additional aspect that has gained recent focus is the potential of extended conditioning strategies to improve physical rehabilitation not only after an acute ischemia-reperfusion event such as acute myocardial infarction and cardiac surgery but also in patients with heart failure. Experimental and preliminary clinical evidence suggests that remote ischemic conditioning may modify cardiac remodeling and additionally enhance skeletal muscle strength therapy to prevent muscle waste, known as an inherent component of a postoperative period and in heart failure. Blood flow restriction exercise and enhanced external counterpulsation may represent cardioprotective corollaries. Combined with exercise, remote ischemic conditioning or, alternatively, blood flow restriction exercise may be of aid in optimizing physical rehabilitation in populations that are not able to perform exercise practice at intensity levels required to promote optimal outcomes.
Collapse
Affiliation(s)
- Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital , Aarhus , Denmark
| | | | | |
Collapse
|
31
|
Heusch G, Gersh BJ. The pathophysiology of acute myocardial infarction and strategies of protection beyond reperfusion: a continual challenge. Eur Heart J 2018; 38:774-784. [PMID: 27354052 DOI: 10.1093/eurheartj/ehw224] [Citation(s) in RCA: 273] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/12/2016] [Indexed: 12/15/2022] Open
Abstract
The incidence of ST segment elevation myocardial infarction (STEMI) has decreased over the last two decades in developed countries, but mortality from STEMI despite widespread access to reperfusion therapy is still substantial as is the development of heart failure, particularly among an expanding older population. In developing countries, the incidence of STEMI is increasing and interventional reperfusion is often not available. We here review the pathophysiology of acute myocardial infarction and reperfusion, notably the temporal and spatial evolution of ischaemic and reperfusion injury, the different modes of cell death, and the resulting coronary microvascular dysfunction. We then go on to briefly characterize the cardioprotective phenomena of ischaemic preconditioning, ischaemic postconditioning, and remote ischaemic conditioning and their underlying signal transduction pathways. We discuss in detail the attempts to translate conditioning strategies and drug therapy into the clinical setting. Most attempts have failed so far to reduce infarct size and improve clinical outcomes in STEMI patients, and we discuss potential reasons for such failure. Currently, it appears that remote ischaemic conditioning and a few drugs (atrial natriuretic peptide, exenatide, metoprolol, and esmolol) reduce infarct size, but studies with clinical outcome as primary endpoint are still underway.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Hufelandstr. 55, 45122 Essen, Germany
| | - Bernard J Gersh
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
32
|
Bochaton T, Mewton N, Thiam ND, Lavocat F, Baetz D, Dufay N, Prieur C, Bonnefoy-Cudraz E, Miossec P, Ovize M. Early kinetics of serum Interleukine-17A and infarct size in patients with reperfused acute ST-elevated myocardial infarction. PLoS One 2017; 12:e0188202. [PMID: 29166391 PMCID: PMC5699816 DOI: 10.1371/journal.pone.0188202] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 11/02/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Recently, it was shown that interleukin-17A (IL-17A) is involved in the pathophysiology of reperfusion injury and associated with infarct size (IS) in experimental models of myocardial infarction. Our aim was to evaluate whether the IL-17A serum level and the IL-17A active fraction was correlated with IS in humans. METHODS 101 patients presenting with a ST-elevated Myocardial Infarction (STEMI) referred for primary percutaneous coronary intervention (PPCI) and 10 healthy controls were included. For each participant, blood samples at admission (H0) and 4 hours after admission (H4) were collected. IL-17A serum levels were assessed using ELISA and the active fraction was assessed with a functional test. IS was determined by peak troponin and peak CK levels for every patient and by contrast-enhanced cardiac magnetic resonance (ce-CMR) for 20 patients. RESULTS The IL-17A serum level was significantly increased in STEMI patients compared to healthy controls, (0.9 pg/mL IQR [0.0-3.2] at H0 and 1.0 pg/mL IQR [0.2-2.8] at H4 versus 0.2 pg/mL IQR [0.0-0.7] for healthy controls; p<0.005). At either time points, IL-17A levels did not correlate with IS as measured by peak troponin, peak CK pr ce-CMR. Also, no correlation was found between the active fraction of IL-17A and IS. CONCLUSION Serum IL-17A level is significantly increased in patients at the early phase of acute MI compared to healthy controls. However, the level of IL-17A in the early hours after reperfusion does not correlate with IS.
Collapse
Affiliation(s)
- Thomas Bochaton
- INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bron, France
- Unité de Soins Intensifs Cardiologiques, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| | - Nathan Mewton
- Service d’explorations fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
- Centre d’investigation clinique de Lyon, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| | - NDieme Thiam
- Immunogénomique et Inflammation, Hôpital Edouard Herriot Pavillon P, Lyon, France
| | - Fabien Lavocat
- Immunogénomique et Inflammation, Hôpital Edouard Herriot Pavillon P, Lyon, France
| | - Delphine Baetz
- INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bron, France
| | - Nathalie Dufay
- NeuroBioTec, Groupement Hospitalier Est, Hôpital Neurologique Pierre Wertheimer, Lyon, France
| | - Cyril Prieur
- Unité de Soins Intensifs Cardiologiques, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| | - Eric Bonnefoy-Cudraz
- INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bron, France
- Unité de Soins Intensifs Cardiologiques, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| | - Pierre Miossec
- Immunogénomique et Inflammation, Hôpital Edouard Herriot Pavillon P, Lyon, France
| | - Michel Ovize
- INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bron, France
- Service d’explorations fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
- Centre d’investigation clinique de Lyon, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
33
|
Management of No-Reflow Phenomenon in the Catheterization Laboratory. JACC Cardiovasc Interv 2017; 10:215-223. [PMID: 28183461 DOI: 10.1016/j.jcin.2016.11.059] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/07/2016] [Accepted: 11/29/2016] [Indexed: 12/29/2022]
Abstract
At the conclusion of a primary percutaneous coronary intervention for ST-segment elevation myocardial infarction, and after the cardiologist makes certain that there is no residual stenosis following stenting, assessment of coronary flow becomes the top priority. The presence of no-reflow is a serious prognostic sign. No-reflow can result in poor healing of the infarct and adverse left ventricular remodeling, increasing the risk for major adverse cardiac events, including congestive heart failure and death. To achieve normal flow, features associated with a high incidence of no-reflow must be anticipated, and measures must be undertaken to prevent its occurrence. In this review, the authors discuss various preventive strategies for no-reflow as well as pharmacological and nonpharmacological interventions that improve coronary blood flow, such as intracoronary adenosine and nitroprusside. Nonpharmacological therapies, such as induced hypothermia, were successful in animal studies, but their effectiveness in reducing no-reflow in humans remains to be determined.
Collapse
|
34
|
Ternant D, Ivanes F, Prunier F, Mewton N, Bejan-Angoulvant T, Paintaud G, Ovize M, Angoulvant D. Revisiting myocardial necrosis biomarkers: assessment of the effect of conditioning therapies on infarct size by kinetic modelling. Sci Rep 2017; 7:10709. [PMID: 28878319 PMCID: PMC5587689 DOI: 10.1038/s41598-017-11352-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/23/2017] [Indexed: 11/09/2022] Open
Abstract
Infarct size is a major predictor of subsequent cardiovascular events following ST-segment elevation myocardial infarction (STEMI) and is frequently used in clinical trials focused on cardioprotection. Approximately assessed through serial blood sampling, it can be accurately measured by imaging techniques, e.g. cardiac magnetic resonance imaging, which is the actual gold standard for infarct size determination but with limited availability in daily practice. We developed a mathematical biomarker kinetic model based on pharmacokinetic compartment models to easily and accurately estimate infarct size using individual data from five clinical trials evaluating the impact of conditioning therapies in STEMI between 2005 and 2013. Serial blood sampling was available in all studies with data regarding creatine kinase (CK), CK specific of cardiomyocytes (CK-MB) and cardiac troponin I. Our model allowed an accurate estimation of biomarker release as a surrogate marker of infarct size and a powerful assessment of conditioning treatments. This biomarker kinetic modelling approach identified CK-MB as the most accurate biomarker in determining infarct size and supports the development of limited sampling strategies that estimate total biomarker amount released with a lower number of samples. It will certainly be a useful add-on to future studies in the field of STEMI and cardioprotection.
Collapse
Affiliation(s)
- David Ternant
- Université François Rabelais de Tours, CNRS, UMR 7292 GICC, Tours, France.,CHRU de Tours, Laboratory of Pharmacology-Toxicology, Tours, France
| | - Fabrice Ivanes
- Université François Rabelais de Tours, EA 4245 CDG & FHU SUPORT, Tours, France. .,CHRU de Tours, Department of Cardiology & FACT, Tours, France.
| | | | - Nathan Mewton
- Université Claude Bernard Lyon 1, INSERM U1060 CarMeN, Lyon, France
| | - Theodora Bejan-Angoulvant
- Université François Rabelais de Tours, CNRS, UMR 7292 GICC, Tours, France.,CHRU de Tours, Department of Clinical Pharmacology, Tours, France
| | - Gilles Paintaud
- Université François Rabelais de Tours, CNRS, UMR 7292 GICC, Tours, France.,CHRU de Tours, Laboratory of Pharmacology-Toxicology, Tours, France
| | - Michel Ovize
- Université Claude Bernard Lyon 1, INSERM U1060 CarMeN, Lyon, France
| | - Denis Angoulvant
- Université François Rabelais de Tours, EA 4245 CDG & FHU SUPORT, Tours, France.,CHRU de Tours, Department of Cardiology & FACT, Tours, France
| |
Collapse
|
35
|
Abstract
The atherosclerotic coronary vasculature is not only the culprit but also a victim of myocardial ischemia/reperfusion injury. Manifestations of such injury are increased vascular permeability and edema, endothelial dysfunction and impaired vasomotion, microembolization of atherothrombotic debris, stasis with intravascular cell aggregates, and finally, in its most severe form, capillary destruction with hemorrhage. In animal experiments, local and remote ischemic pre- and postconditioning not only reduce infarct size but also these manifestations of coronary vascular injury, as do drugs which recruit signal transduction steps of conditioning. Clinically, no-reflow is frequently seen after interventional reperfusion, and it carries an adverse prognosis. The translation of cardioprotective interventions to clinical practice has been difficult to date. Only 4 drugs (brain natriuretic peptide, exenatide, metoprolol, and esmolol) stand unchallenged to date in reducing infarct size in patients with reperfused acute myocardial infarction; unfortunately, for these drugs, no information on their impact on the ischemic/reperfused coronary circulation is available.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, University of Essen, Essen, Germany.
| |
Collapse
|
36
|
Baranyai T, Giricz Z, Varga ZV, Koncsos G, Lukovic D, Makkos A, Sárközy M, Pávó N, Jakab A, Czimbalmos C, Vágó H, Ruzsa Z, Tóth L, Garamvölgyi R, Merkely B, Schulz R, Gyöngyösi M, Ferdinandy P. In vivo MRI and ex vivo histological assessment of the cardioprotection induced by ischemic preconditioning, postconditioning and remote conditioning in a closed-chest porcine model of reperfused acute myocardial infarction: importance of microvasculature. J Transl Med 2017; 15:67. [PMID: 28364777 PMCID: PMC5376486 DOI: 10.1186/s12967-017-1166-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 03/15/2017] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Cardioprotective value of ischemic post- (IPostC), remote (RIC) conditioning in acute myocardial infarction (AMI) is unclear in clinical trials. To evaluate cardioprotection, most translational animal studies and clinical trials utilize necrotic tissue referred to the area at risk (AAR) by magnetic resonance imaging (MRI). However, determination of AAR by MRI' may not be accurate, since MRI-indices of microvascular damage, i.e., myocardial edema and microvascular obstruction (MVO), may be affected by cardioprotection independently from myocardial necrosis. Therefore, we assessed the effect of IPostC, RIC conditioning and ischemic preconditioning (IPreC; positive control) on myocardial necrosis, edema and MVO in a clinically relevant, closed-chest pig model of AMI. METHODS AND RESULTS Acute myocardial infarction was induced by a 90-min balloon occlusion of the left anterior descending coronary artery (LAD) in domestic juvenile female pigs. IPostC (6 × 30 s ischemia/reperfusion after 90-min occlusion) and RIC (4 × 5 min hind limb ischemia/reperfusion during 90-min LAD occlusion) did not reduce myocardial necrosis as assessed by late gadolinium enhancement 3 days after reperfusion and by ex vivo triphenyltetrazolium chloride staining 3 h after reperfusion, however, the positive control, IPreC (3 × 5 min ischemia/reperfusion before 90-min LAD occlusion) did. IPostC and RIC attenuated myocardial edema as measured by cardiac T2-weighted MRI 3 days after reperfusion, however, AAR measured by Evans blue staining was not different among groups, which confirms that myocardial edema is not a measure of AAR, IPostC and IPreC but not RIC decreased MVO. CONCLUSION We conclude that IPostC and RIC interventions may protect the coronary microvasculature even without reducing myocardial necrosis.
Collapse
Affiliation(s)
- Tamás Baranyai
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Giricz
- 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
| | - Gábor Koncsos
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Dominika Lukovic
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - András Makkos
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Márta Sárközy
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - Noémi Pávó
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - András Jakab
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Csilla Czimbalmos
- The Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Hajnalka Vágó
- The Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Zoltán Ruzsa
- The Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Levente Tóth
- Institute of Diagnostic Imaging and Radiation Oncology, University of Kaposvár, Kaposvár, Hungary
- Department of Radiology, University of Pécs, Pecs, Hungary
| | - Rita Garamvölgyi
- Institute of Diagnostic Imaging and Radiation Oncology, University of Kaposvár, Kaposvár, Hungary
| | - Béla Merkely
- The Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University, Giessen, Germany
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Department of Biochemistry, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
37
|
Mentias A, Mahmoud AN, Elgendy IY, Elgendy AY, Barakat AF, Abuzaid AS, Saad M, Kapadia SR. Ischemic postconditioning during primary percutaneous coronary intervention. Catheter Cardiovasc Interv 2017; 90:1059-1067. [PMID: 28296005 DOI: 10.1002/ccd.26965] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/10/2016] [Accepted: 01/16/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Although some studies have shown potential benefit for ischemic postconditioning (IPoC) during primary percutaneous coronary intervention (PCI) in improving surrogate markers of reperfusion and infarction size, the benefit of this approach on clinical outcomes remains unknown. METHODS AND RESULTS Electronic databases were searched for randomized clinical trials that compared IPoC versus conventional treatment during primary PCI. Random effects DerSimonian-Laird risk ratios (RR) were calculated for different clinical and surrogate outcomes. The main outcome of this analysis was all-cause mortality. A total of 25 trials involving 3,619 patients were included in the analysis. At a mean follow up of 14 months (95% confidence interval (CI) 8.6-19.4 months), the incidence of all-cause mortality was 4.9% [95% CI 3.8-6.0%] in the IPoC group versus 3.8% [95% CI 1.9-5.7%] in the control group (RR 0.92, 95% CI 0.68-1.24, P = 0.74). The risk of reinfarction (2.7% [95% CI 1.1-4.3%] vs. 2.3% [0.6-4.0%]; RR 1.29, 95% CI 0.62-2.68, P = 0.72), heart failure (3.6% [95% CI 2.0-5.1%] vs. 5.7% [95% CI 3.3-8.2%]; RR 0.77, 95% CI 0.58-1.06, P = 0.24), target vessel revascularization (3.2% [95% CI 1.7-4.7%] vs. 2.4% [95% CI 1.4-3.3%]; RR 1.40, 95% CI 0.90-2.20, P = 0.20), and stent thrombosis (2.4% [95% CI 1.1-3.8%] vs. 1.8% [95% CI 0.5-3.2%]); RR 1.50, 95% CI 0.60-3.70, P = 0.40) was similar in both groups. CONCLUSIONS IPoC does not appear to reduce the risk of clinical adverse events in patients with ST-elevation myocardial infarction undergoing primary PCI. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amgad Mentias
- Department of Medicine, Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Ahmed N Mahmoud
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Islam Y Elgendy
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Akram Y Elgendy
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Amr F Barakat
- Department of Internal Medicine, Cleveland Clinic, Cleveland, Ohio
| | - A Sami Abuzaid
- Division of Cardiovascular medicine, Sidney Kimmel Medical College at Thomas Jefferson University/Christiana Care Health System, Newark, Delaware
| | - Marwan Saad
- Department of Medicine, Division of cardiovascular diseases, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Samir R Kapadia
- Cleveland Clinic, Heart and Vascular Institute, Cleveland, Ohio
| |
Collapse
|
38
|
Abstract
The mortality from acute myocardial infarction (AMI) remains significant, and the prevalence of post-myocardial infarction heart failure is increasing. Therefore, cardioprotection beyond timely reperfusion is needed. Conditioning procedures are the most powerful cardioprotective interventions in animal experiments. However, ischemic preconditioning cannot be used to reduce infarct size in patients with AMI because its occurrence is not predictable; several studies in patients undergoing surgical coronary revascularization report reduced release of creatine kinase and troponin. Ischemic postconditioning reduces infarct size in most, but not all, studies in patients undergoing interventional reperfusion of AMI, but may require direct stenting and exclusion of patients with >6 hours of symptom onset to protect. Remote ischemic conditioning reduces infarct size in patients undergoing interventional reperfusion of AMI, elective percutaneous or surgical coronary revascularization, and other cardiovascular surgery in many, but not in all, studies. Adequate dose-finding phase II studies do not exist. There are only 2 phase III trials, both on remote ischemic conditioning in patients undergoing cardiovascular surgery, both with neutral results in terms of infarct size and clinical outcome, but also both with major problems in trial design. We discuss the difficulties in translation of cardioprotection from animal experiments and proof-of-concept trials to clinical practice. Given that most studies on ischemic postconditioning and all studies on remote ischemic preconditioning in patients with AMI reported reduced infarct size, it would be premature to give up on cardioprotection.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology (G.H.) and Clinic for Cardiology (T.R.), West German Heart and Vascular Center, University School of Medicine Essen, Essen, Germany
| | - Tienush Rassaf
- From the Institute for Pathophysiology (G.H.) and Clinic for Cardiology (T.R.), West German Heart and Vascular Center, University School of Medicine Essen, Essen, Germany
| |
Collapse
|
39
|
Reinstadler SJ, Stiermaier T, Fuernau G, de Waha S, Desch S, Metzler B, Thiele H, Eitel I. The challenges and impact of microvascular injury in ST-elevation myocardial infarction. Expert Rev Cardiovasc Ther 2016; 14:431-43. [DOI: 10.1586/14779072.2016.1135055] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
40
|
Bouleti C, Mewton N, Germain S. The no-reflow phenomenon: State of the art. Arch Cardiovasc Dis 2015; 108:661-74. [PMID: 26616729 DOI: 10.1016/j.acvd.2015.09.006] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 08/28/2015] [Accepted: 09/07/2015] [Indexed: 02/06/2023]
Abstract
Primary percutaneous coronary intervention (PCI) is the best available reperfusion strategy for acute ST-segment elevation myocardial infarction (STEMI), with nearly 95% of occluded coronary vessels being reopened in this setting. Despite re-establishing epicardial coronary vessel patency, primary PCI may fail to restore optimal myocardial reperfusion within the myocardial tissue, a failure at the microvascular level known as no-reflow (NR). NR has been reported to occur in up to 60% of STEMI patients with optimal coronary vessel reperfusion. When it does occur, it significantly attenuates the beneficial effect of reperfusion therapy, leading to poor outcomes. The pathophysiology of NR is complex and incompletely understood. Many phenomena are known to contribute to NR, including leukocyte infiltration, vasoconstriction, activation of inflammatory pathways and cellular oedema. Vascular damage and haemorrhage may also play important roles in the establishment of NR. In this review, we describe the pathophysiological mechanisms of NR and the tools available for diagnosing it. We also describe the microvasculature and the endothelial mechanisms involved in NR, which may provide relevant therapeutic targets for reducing NR and improving the prognosis for patients.
Collapse
Affiliation(s)
- Claire Bouleti
- Service de cardiologie, hôpital Bichat, AP-HP, Paris, France; DHU FIRE, université Paris Diderot, Paris, France; Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris, France; CNRS/UMR 7241, Paris, France; Inserm U 1050, Paris, France
| | - Nathan Mewton
- Hôpital cardiovasculaire Louis-Pradel, centre d'investigation clinique unité, hospices civils de Lyon, Bron, France; Inserm U 1407, Lyon, France
| | - Stéphane Germain
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris, France; CNRS/UMR 7241, Paris, France; Inserm U 1050, Paris, France.
| |
Collapse
|
41
|
Gao J, Luo J, Liu F, Zheng Y, Chen B, Chen Q, Yang Y. Short-and long-term effects of ischemic postconditioning in STEMI patients: a meta-analysis. Lipids Health Dis 2015; 14:147. [PMID: 26573572 PMCID: PMC4647593 DOI: 10.1186/s12944-015-0151-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/06/2015] [Indexed: 01/28/2023] Open
Abstract
Background Compelling evidence from large randomized trials demonstrates the salutary effects of ischemic postconditioning on cardioprotection against ischemic/reperfusion injury. However, some studies appear negative findings. This study was designed to assess the short-and long-term effects of postconditioning (Poc) in studies including evolving ST-elevation myocardial infarction (STEMI). Methods Relevant studies were identified through an electronic literature search from the PubMed, Library of Congress, Embase, Cochrane Central Register of Controlled Trials, and ISI Web of Science. Studies published up to December 2014 were eligible for inclusion. Patients older than 18 years presenting within 12 h of the first STEMI and eligible for angioplasty were considered for the study. Results The 25 trials allocated 1136 patients to perform locational postconditioning cycles at the onset of reperfusion and 1153 patients to usual percutaneous coronary intervention (PCI). Ischemic postconditioning demonstrated a decrease in serum cardiac enzymes creatine kinase (CK) and CK-MB (P < 0.00001 and P =0.25, respectively) in the subgroup analysis based on direct stenting. Reduction in infarct size by imaging was showed during7 days after myocardial infarction (P =0.01), but not in the longterm (P = 0.08). The wall motion score index was improved in both the short term within 7 days (P = 0.009) and the long term over 6 months after receiving Poc (P = 0.02). All included studies were limited by the high risk of performance and publication bias. Conclusions Ischemic postconditioning by brief interruptions of coronary blood flow at the onset of reperfusion after PCI appears to be superior to PCI alone in reducing myocardial injury and improving left ventricular function, especially in patients who have received direct stenting in PCI.
Collapse
Affiliation(s)
- Jing Gao
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, P.R., China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, P.R., China.,Department of endocrinology, Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, P.R.China
| | - Junyi Luo
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, P.R., China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, P.R., China
| | - Fen Liu
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, P.R., China
| | - Yingying Zheng
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, P.R., China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, P.R., China
| | - Bangdang Chen
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, P.R., China
| | - Qingjie Chen
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, P.R., China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, P.R., China
| | - Yining Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, P.R., China. .,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, P.R., China.
| |
Collapse
|
42
|
Ferdinandy P, Hausenloy DJ, Heusch G, Baxter GF, Schulz R. Interaction of risk factors, comorbidities, and comedications with ischemia/reperfusion injury and cardioprotection by preconditioning, postconditioning, and remote conditioning. Pharmacol Rev 2015; 66:1142-74. [PMID: 25261534 DOI: 10.1124/pr.113.008300] [Citation(s) in RCA: 461] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pre-, post-, and remote conditioning of the myocardium are well described adaptive responses that markedly enhance the ability of the heart to withstand a prolonged ischemia/reperfusion insult and provide therapeutic paradigms for cardioprotection. Nevertheless, more than 25 years after the discovery of ischemic preconditioning, we still do not have established cardioprotective drugs on the market. Most experimental studies on cardioprotection are still undertaken in animal models, in which ischemia/reperfusion is imposed in the absence of cardiovascular risk factors. However, ischemic heart disease in humans is a complex disorder caused by, or associated with, cardiovascular risk factors and comorbidities, including hypertension, hyperlipidemia, diabetes, insulin resistance, heart failure, altered coronary circulation, and aging. These risk factors induce fundamental alterations in cellular signaling cascades that affect the development of ischemia/reperfusion injury per se and responses to cardioprotective interventions. Moreover, some of the medications used to treat these risk factors, including statins, nitrates, and antidiabetic drugs, may impact cardioprotection by modifying cellular signaling. The aim of this article is to review the recent evidence that cardiovascular risk factors and their medication may modify the response to cardioprotective interventions. We emphasize the critical need to take into account the presence of cardiovascular risk factors and concomitant medications when designing preclinical studies for the identification and validation of cardioprotective drug targets and clinical studies. This will hopefully maximize the success rate of developing rational approaches to effective cardioprotective therapies for the majority of patients with multiple risk factors.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Derek J Hausenloy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gerd Heusch
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gary F Baxter
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| |
Collapse
|
43
|
Eitel I, Stiermaier T, Rommel KP, Fuernau G, Sandri M, Mangner N, Linke A, Erbs S, Lurz P, Boudriot E, Mende M, Desch S, Schuler G, Thiele H. Cardioprotection by combined intrahospital remote ischaemic perconditioning and postconditioning in ST-elevation myocardial infarction: the randomized LIPSIA CONDITIONING trial. Eur Heart J 2015; 36:3049-57. [PMID: 26385956 DOI: 10.1093/eurheartj/ehv463] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/19/2015] [Indexed: 11/13/2022] Open
Abstract
AIMS Remote ischaemic conditioning (RIC) and postconditioning (PostC) are both potent activators of innate protection against ischaemia-reperfusion injury and have demonstrated cardioprotection in experimental and clinical ST-elevation myocardial infarction (STEMI) trials. However, their combined effects have not been studied in detail. The aim of this study was to evaluate if the co-application of intrahospital RIC and PostC has a more powerful effect on myocardial salvage compared with either PostC alone or control. METHODS AND RESULTS This prospective, controlled, single-centre study randomized 696 STEMI patients to one of the following three groups: (i) combined intrahospital RIC + PostC in addition to primary percutaneous coronary intervention (PCI); (ii) PostC in addition to PCI; and (iii) conventional PCI (control). The primary endpoint myocardial salvage index was assessed by cardiac magnetic resonance (CMR) imaging within 3 days after infarction. Secondary endpoints included infarct size and microvascular obstruction (MVO) assessed by CMR. The combined clinical endpoint consisted of death, reinfarction, and new congestive heart failure within 6 months. The primary endpoint myocardial salvage index was significantly greater in the combined RIC + PostC group when compared with the control group (49 [interquartile range 30-72] vs. 40 [interquartile range 16-68], P = 0.02). Postconditioning alone failed to improve myocardial salvage when compared with conventional PCI (P = 0.39). The secondary endpoints, including infarct size and MVO, showed no significant differences between groups. Clinical follow-up at 6 months revealed no differences in the combined clinical endpoint between groups (P = 0.44). CONCLUSION Combined intrahospital RIC + PostC in conjunction with PCI in STEMI significantly improves myocardial salvage in comparison with control and PostC. CLINICALTRIALSGOV NCT02158468.
Collapse
Affiliation(s)
- Ingo Eitel
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Thomas Stiermaier
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Karl P Rommel
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Georg Fuernau
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Marcus Sandri
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Norman Mangner
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Axel Linke
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Sandra Erbs
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Phillip Lurz
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Enno Boudriot
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Meinhard Mende
- Clinical Trial Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Steffen Desch
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Gerhard Schuler
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Holger Thiele
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| |
Collapse
|
44
|
Yetgin T, van Kranenburg M, Ten Cate T, Duncker DJ, de Boer MJ, Diletti R, van Geuns RJM, Zijlstra F, Manintveld OC. Ischemic Postconditioning After Routine Thrombus Aspiration During Primary Percutaneous Coronary Intervention: Rationale and Design of the POstconditioning Rotterdam Trial. Catheter Cardiovasc Interv 2015; 88:508-514. [PMID: 27022882 DOI: 10.1002/ccd.26239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/31/2015] [Accepted: 08/24/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Whether ischemic postconditioning (IPOC) immediately after routine thrombus aspiration (TA) reduces infarct size (IS) in patients with ST-segment elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (PPCI) has not been established. STUDY DESIGN The POstconditioning Rotterdam Trial (PORT) is a dual-center, prospective, open-label, randomized trial with blinded endpoint evaluation enrolling 72 subjects with first-time STEMI, and an occluded infarct-related artery (IRA) without collaterals undergoing PPCI. Subjects are randomized 1:1 to a strategy of IPOC immediately after TA followed by stenting of the IRA or to conventional percutaneous coronary intervention (PCI), including TA followed by stenting of the IRA (controls). Cardiac magnetic resonance imaging (MRI) is performed at 3-5 days after STEMI and at 3 months. The primary endpoint is IS at 3 months measured by delayed enhancement MRI. Other secondary endpoints include MRI-derived microvascular obstruction (MVO), left ventricular ejection fraction, myocardial salvage index, enzymatic IS, ST-segment resolution, myocardial blush grade, microcirculatory resistance, inflammation markers, and clinical events through 3-month follow-up. CONCLUSIONS PORT is testing the hypothesis that adding IPOC (against lethal reperfusion injury) to TA (against distal embolization and MVO) is cardioprotective and reduces ultimate IS in STEMI patients undergoing PPCI (Dutch Trial Register identifier: NTR4040). © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tuncay Yetgin
- Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands.,Interuniversity Cardiology Institute of the Netherlands, ICIN-KNAW, Utrecht, the Netherlands
| | | | - Tim Ten Cate
- Department of Cardiology, UMC St. Radboud, Nijmegen, the Netherlands
| | - Dirk J Duncker
- Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands.,Interuniversity Cardiology Institute of the Netherlands, ICIN-KNAW, Utrecht, the Netherlands
| | - Menko-Jan de Boer
- Department of Cardiology, UMC St. Radboud, Nijmegen, the Netherlands
| | - Roberto Diletti
- Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
| | - Robert-Jan M van Geuns
- Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands.,Department of Radiology, Erasmus MC, Rotterdam, the Netherlands
| | - Felix Zijlstra
- Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands.,Interuniversity Cardiology Institute of the Netherlands, ICIN-KNAW, Utrecht, the Netherlands
| | | |
Collapse
|
45
|
Uitterdijk A, Yetgin T, te Lintel Hekkert M, Sneep S, Krabbendam-Peters I, van Beusekom HMM, Fischer TM, Cornelussen RN, Manintveld OC, Merkus D, Duncker DJ. Vagal nerve stimulation started just prior to reperfusion limits infarct size and no-reflow. Basic Res Cardiol 2015; 110:508. [PMID: 26306761 PMCID: PMC4549380 DOI: 10.1007/s00395-015-0508-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 07/22/2015] [Accepted: 08/11/2015] [Indexed: 12/26/2022]
Abstract
Vagal nerve stimulation (VNS) started prior to, or during, ischemia has been shown to reduce infarct size. Here, we investigated the effect of VNS when started just prior to, and continued during early, reperfusion on infarct size and no-reflow and studied the underlying mechanisms. For this purpose, swine (13 VNS, 10 sham) underwent 45 min mid-LAD occlusion followed by 120 min of reperfusion. VNS was started 5 min prior to reperfusion and continued until 15 min of reperfusion. Area at risk, area of no-reflow (% of infarct area) and infarct size (% of area at risk), circulating cytokines, and regional myocardial leukocyte influx were assessed after 120 min of reperfusion. VNS significantly reduced infarct size from 67 ± 2 % in sham to 54 ± 5 % and area of no-reflow from 54 ± 6 % in sham to 32 ± 6 %. These effects were accompanied by reductions in neutrophil (~40 %) and macrophage (~60 %) infiltration in the infarct area (all p < 0.05), whereas systemic circulating plasma levels of TNFα and IL6 were not affected. The degree of cardioprotection could not be explained by the VNS-induced bradycardia or the VNS-induced decrease in the double product of heart rate and left ventricular systolic pressure. In the presence of NO-synthase inhibitor LNNA, VNS no longer attenuated infarct size and area of no-reflow, which was paralleled by similarly unaffected regional leukocyte infiltration. In conclusion, VNS is a promising novel adjunctive therapy that limits reperfusion injury in a large animal model of acute myocardial infarction.
Collapse
Affiliation(s)
- André Uitterdijk
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000, CA, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mechanical post-conditioning in STEMI patients undergoing primary percutaneous coronary intervention. J Saudi Heart Assoc 2015; 27:192-200. [PMID: 26136633 PMCID: PMC4481425 DOI: 10.1016/j.jsha.2014.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/05/2014] [Accepted: 11/11/2014] [Indexed: 11/21/2022] Open
Abstract
Although early myocardial reperfusion via primary percutaneous coronary intervention (PCI) allows the preservation of left ventricular function and improves outcome, the acute restoration of blood flow may contribute to the pathophysiology of infarction, a complex phenomenon called reperfusion injury. First described in animal models of coronary obstruction, mechanical post-conditioning, a sequence of repetitive interruption of coronary blood flow applied immediately after reopening of the occluded vessel, was able to reduce the infarct size. However, evidence of its real benefit remains controversial. This review describes the mechanisms of post-conditioning action and the different protocols employed focusing on its impact on primary PCI outcome.
Collapse
|
47
|
He H, Li N, Zhao Z, Han F, Wang X, Zeng Y. Ischemic postconditioning improves the expression of cellular membrane connexin 43 and attenuates the reperfusion injury in rat acute myocardial infarction. Biomed Rep 2015; 3:668-674. [PMID: 26405543 DOI: 10.3892/br.2015.485] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 05/21/2015] [Indexed: 11/05/2022] Open
Abstract
To investigate the effects of cellular membrane connexin 43 (Cx43) and the potential details in ischemic postconditioning (IPOC)-induced cardioprotection, ischemia/reperfusion (IR) models were generated in 8-week-old male Sprague-Dawley rats by ligating the left coronary artery anterior descending branch. The serum levels of myocardial creatases, nitric oxide (NO) and malondialdehyde (MDA) levels, infarct size, arrhythmia events, expression and distribution of Cx43, ultrastructure and apoptosis in the myocardium in different treatments with IR, IR + IPOC, IR + diazoxide or IR + IPOC + 5-hydroxydecanoate acid (5-HD) were investigated. Consequently, IPOC decreased infarct size (10.9 vs. 43.3%, P<0.01) and the levels of myocardial creatases, NO and MDA, and improved the expression (16.8 vs. 25.2% and 6.4 vs. 32.8%, after 1- and 3-h reperfusion, respectively; P<0.01) and distribution of Cx43, ultrastructure and apoptosis (19.2 vs. 42.9%, P<0.01) significantly. Diazoxide partly simulated the effects, and 5-HD attenuated but not completely abolished the effects of IPOC. In addition, the phosphorylated Cx43 (p-Cx43) level in the IR + IPOC group was lower than that in the IR + diazoxide group after 1-h reperfusion (26.1 vs. 29.4%, P>0.05); however, it was reversed after 3-h reperfusion and the p-Cx43 level in the IR + IPOC group was significantly higher than that in the IR + diazoxide group (32.8 vs. 18.7%, P<0.01). In conclusion, cell membrane Cx43 is also involved in the process of IPOC-induced cardioprotection and the improvement of membrane Cx43 is more dependent on mitochondrial KATP in the earlier phase of IPOC compared to the late phase of IPOC.
Collapse
Affiliation(s)
- Hua He
- Department of Emergency Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Nan Li
- Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Zhihong Zhao
- Department of Cardiology, Pudong New Area District Zhoupu Hospital, Shanghai 201318, P.R. China
| | - Fusheng Han
- Department of Emergency Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Xifu Wang
- Department of Emergency Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Yujie Zeng
- Department of Emergency Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| |
Collapse
|
48
|
Schaaf MJ, Mewton N, Rioufol G, Angoulvant D, Cayla G, Delarche N, Jouve B, Guerin P, Vanzetto G, Coste P, Morel O, Roubille F, Elbaz M, Roth O, Prunier F, Cung TT, Piot C, Sanchez I, Bonnefoy-Cudraz E, Revel D, Giraud C, Croisille P, Ovize M. Pre-PCI angiographic TIMI flow in the culprit coronary artery influences infarct size and microvascular obstruction in STEMI patients. J Cardiol 2015; 67:248-53. [PMID: 26116981 DOI: 10.1016/j.jjcc.2015.05.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/18/2015] [Accepted: 05/03/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The influence of initial-thrombolysis in myocardial infarction (i-TIMI) coronary flow in the culprit coronary artery on myocardial infarct and microvascular obstruction (MVO) size is unclear. We assessed the impact on infarct size of i-TIMI flow in the culprit coronary artery, as well as on MVO incidence and size, by contrast-enhanced cardiac magnetic resonance (ce-CMR). METHODS In a prospective, multicenter study, pre-percutaneous coronary intervention (PCI) coronary occlusion was defined by an i-TIMI flow ≤1, and patency was defined by an i-TIMI flow ≥2. Infarct size, as well as MVO presence and size, were measured on ce-CMR 72h after admission. RESULTS A total of 140 patients presenting with ST-elevated myocardial infarction referred for primary PCI were included. There was no significant difference in final post-PCI TIMI flow between the groups (2.95±0.02 vs. 2.97±0.02, respectively; p=0.44). In the i-TIMI flow ≤1 group, infarct size was significantly larger (32±17g vs. 21±17g, respectively; p=0.002), MVO was significantly more frequent (74% vs. 53%, respectively; p=0.012), and MVO size was significantly larger [1.3 IQR (0; 7.1) vs. 0 IQR (0; 1.6)], compared to in the i-TIMI ≥2 patient group. CONCLUSION Initial angiographic TIMI flow in the culprit coronary artery prior to any PCI predicted final infarct size and MVO size: the better was the i-TIMI flow, the smaller were the infarct and MVO size.
Collapse
Affiliation(s)
- Mathieu Julien Schaaf
- Cardiology Division, Centre d'Investigation Clinique de Lyon (CIC), Groupement Hospitalier Est, Hôpital Louis Pradel, 28 avenue Doyen Lépine, 69677 Bron, Hospices Civils de Lyon, France.
| | - Nathan Mewton
- Cardiology Division, Centre d'Investigation Clinique de Lyon (CIC), Groupement Hospitalier Est, Hôpital Louis Pradel, 28 avenue Doyen Lépine, 69677 Bron, Hospices Civils de Lyon, France; INSERM UMR-1060, CarMeN Laboratory, Université Claude Bernard Lyon1, Faculté de Médecine Lyon Est, F-69373 Lyon, France
| | - Gilles Rioufol
- Cardiology Division, Centre d'Investigation Clinique de Lyon (CIC), Groupement Hospitalier Est, Hôpital Louis Pradel, 28 avenue Doyen Lépine, 69677 Bron, Hospices Civils de Lyon, France
| | - Denis Angoulvant
- University Hospital of Tours, Hopital Trousseau, Cardiology Division, Université François Rabelais, Tours, France
| | - Guillaume Cayla
- University Hospital of Nîmes, Hôpital Universitaire Carémeau, Cardiology Division, Nîmes, France
| | | | - Bernard Jouve
- Regional Hospital of Aix-en-Provence, Cardiology Division, Aix en Provence, France
| | - Patrice Guerin
- Thorax Institute, Invasive Cardiology Department, University Hospital of Nantes, Nantes, France
| | - Gerald Vanzetto
- University Hospital of Grenoble, Hôpital La Tronche, Cardiology Division, Grenoble, France
| | - Pierre Coste
- University Hospital of Bordeaux, Groupe Hospitalier Sud Pessac, Bordeaux, France
| | - Olivier Morel
- University Hospital of Strasbourg, Nouvel Hôpital Civil, Cardiology Division, Strasbourg, France
| | - François Roubille
- University Hospital of Montpellier, Cardiology Division, UMR5203, UMR661, Universités Montpellier 1 and 2, Montpellier, France
| | - Meyer Elbaz
- University Hospital of Toulouse, Hôpital Rangeuil, Université Paul Sabatier, Toulouse, France
| | - Olivier Roth
- Regional Hospital of Mulhouse, Hôpital Emile Müller, Cardiology Division, Mulhouse, France
| | - Fabrice Prunier
- University Hospital of Angers, Cardiology Division, Angers, France
| | - Thien Tri Cung
- University Hospital of Montpellier, Cardiology Division, UMR5203, UMR661, Universités Montpellier 1 and 2, Montpellier, France
| | - Christophe Piot
- University Hospital of Montpellier, Cardiology Division, UMR5203, UMR661, Universités Montpellier 1 and 2, Montpellier, France
| | - Ingrid Sanchez
- Cardiology Division, Centre d'Investigation Clinique de Lyon (CIC), Groupement Hospitalier Est, Hôpital Louis Pradel, 28 avenue Doyen Lépine, 69677 Bron, Hospices Civils de Lyon, France
| | - Eric Bonnefoy-Cudraz
- Cardiology Division, Centre d'Investigation Clinique de Lyon (CIC), Groupement Hospitalier Est, Hôpital Louis Pradel, 28 avenue Doyen Lépine, 69677 Bron, Hospices Civils de Lyon, France
| | - Didier Revel
- Radiology Department, CREATIS-LRMN, CNRS UMR 5220 - INSERM U630 - Université Claude Bernard Lyon 1, Lyon, France
| | - Céline Giraud
- Cardiology Division, Centre d'Investigation Clinique de Lyon (CIC), Groupement Hospitalier Est, Hôpital Louis Pradel, 28 avenue Doyen Lépine, 69677 Bron, Hospices Civils de Lyon, France
| | - Pierre Croisille
- Radiology Department, CREATIS-LRMN, CNRS UMR 5220 - INSERM U630 - Université Claude Bernard Lyon 1, Lyon, France; University Hospital of Saint-Etienne, Radiology Department, Saint-Etienne, France
| | - Michel Ovize
- Cardiology Division, Centre d'Investigation Clinique de Lyon (CIC), Groupement Hospitalier Est, Hôpital Louis Pradel, 28 avenue Doyen Lépine, 69677 Bron, Hospices Civils de Lyon, France; INSERM UMR-1060, CarMeN Laboratory, Université Claude Bernard Lyon1, Faculté de Médecine Lyon Est, F-69373 Lyon, France
| |
Collapse
|
49
|
Rationale and design of the Cyclosporine to ImpRove Clinical oUtcome in ST-elevation myocardial infarction patients (the CIRCUS trial). Am Heart J 2015; 169:758-766.e6. [PMID: 26027612 DOI: 10.1016/j.ahj.2015.02.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/28/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND Both acute myocardial ischemia and reperfusion contribute to cardiomyocyte death in ST-elevation myocardial infarction (STEMI). The final infarct size is the principal determinant of subsequent clinical outcome in STEMI patients. In a proof-of-concept phase II trial, the administration of cyclosporine prior to primary percutaneous coronary intervention (PPCI) has been associated with a reduction of infarct size in STEMI patients. METHODS CIRCUS is an international, prospective, multicenter, randomized, double-blinded, placebo-controlled trial. The study is designed to compare the efficacy and safety of cyclosporine versus placebo, in addition to revascularization by PPCI, in patients presenting with acute anterior myocardial infarction within 12 hours of symptoms onset and initial TIMI flow ≤1 in the culprit left anterior descending coronary artery. Patients are randomized in a 1:1 fashion to 2.5 mg/kg intravenous infusion of cyclosporine or matching placebo performed in the minutes preceding PCI. The primary efficacy end point of CIRCUS is a composite of 1-year all-cause mortality, rehospitalization for heart failure or heart failure worsening during initial hospitalization, and left ventricular adverse remodeling as determined by sequential transthoracic echochardiography. Secondary outcomes will be tested using a hierarchical sequence of left ventricular (LV) ejection fraction and absolute measurements of LV volumes. The composite of death and rehospitalization for heart failure or heart failure worsening during initial hospitalization will be further assessed at three years after the initial infarction. RESULTS Recruitment lasted from April 2011 to February 2014. The CIRCUS trial has recruited 975 patients with acute anterior myocardial infarction. The 12-months results are expected to be available in 2015. CONCLUSIONS The CIRCUS trial is testing the hypothesis that cyclosporine in addition to early revascularization with PPCI compared to placebo in patients with acute anterior myocardial infarction reduces the incidence of death, heart failure and adverse LV remodeling at one-year follow-up.
Collapse
|
50
|
Abstract
Reperfusion is mandatory to salvage ischemic myocardium from infarction, but reperfusion per se contributes to injury and ultimate infarct size. Therefore, cardioprotection beyond that by timely reperfusion is needed to reduce infarct size and improve the prognosis of patients with acute myocardial infarction. The conditioning phenomena provide such cardioprotection, insofar as brief episodes of coronary occlusion/reperfusion preceding (ischemic preconditioning) or following (ischemic postconditioning) sustained myocardial ischemia with reperfusion reduce infarct size. Even ischemia/reperfusion in organs remote from the heart provides cardioprotection (remote ischemic conditioning). The present review characterizes the signal transduction underlying the conditioning phenomena, including their physical and chemical triggers, intracellular signal transduction, and effector mechanisms, notably in the mitochondria. Cardioprotective signal transduction appears as a highly concerted spatiotemporal program. Although the translation of ischemic postconditioning and remote ischemic conditioning protocols to patients with acute myocardial infarction has been fairly successful, the pharmacological recruitment of cardioprotective signaling has been largely disappointing to date.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany.
| |
Collapse
|