1
|
Wang YX, Deng ZH, Li YY, Bai K, Ma J, Liu Y, Chen Q. Function of hematopoiesis and bone marrow niche in inflammation and non-hematopoietic diseases. LIFE MEDICINE 2025; 4:lnaf015. [PMID: 40376111 PMCID: PMC12076419 DOI: 10.1093/lifemedi/lnaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/24/2025] [Indexed: 05/18/2025]
Abstract
Hematopoiesis and the behavior of hematopoietic stem and progenitor cells (HSPCs) are regulated by the bone marrow niche. Here, we introduce the major niche cell types in bone marrow and their response to stress condition. We highlight the hematopoietic response and bone marrow niche adaptation to inflammatory condition and non-hematopoietic diseases, which are not systematically summarized. These emerging data suggest targeting hematopoiesis and bone marrow niche may provide novel therapeutic target to precisely control the progression of the diseases.
Collapse
Affiliation(s)
- Yu-xiang Wang
- Center for Cell Lineage Atlas, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 101408, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou 510530, China
| | - Zhao-hua Deng
- Center for Cell Lineage Atlas, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 101408, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou 510530, China
| | - Yu-yan Li
- Center for Cell Lineage Atlas, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 101408, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou 510530, China
| | - Ke Bai
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Jinjin Ma
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
- The Institute of Future Health, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yang Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Qi Chen
- Center for Cell Lineage Atlas, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou 510530, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory for Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou 510530, China
| |
Collapse
|
2
|
Mäntylä T, Wang C, Hänninen M, Immonen K, Jäntti T, Lassus J, Tikkanen I, Pulkki K, Devaux Y, Harjola VP, Lakkisto P, CardShock Study Investigators 1. Circulating levels of miR-20b-5p are associated with survival in cardiogenic shock. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100284. [PMID: 39927096 PMCID: PMC11804825 DOI: 10.1016/j.jmccpl.2025.100284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 02/11/2025]
Abstract
Cardiogenic shock (CS) is a medical emergency with high in-hospital mortality. New biomarkers are needed to identify patients at a greater risk of adverse outcomes. This study aimed to investigate the prognostic potential of microRNAs (miRNAs) in assessment of the outcome of cardiogenic shock. Circulating miRNA levels were measured by quantitative PCR in plasma samples collected at baseline from 165 patients of the multicenter, prospective, observational CardShock study and compared between in-hospital and 90-day survivors and non-survivors. Of the 10 studied miRNAs, median levels of miR-20b-5p at baseline were significantly higher in in-hospital and 90-day survivors compared to non-survivors [median 0.014 arbitrary units (AU) (interquartile range (IQR) 0.003-0.024) vs. 0.008 AU (IQR 0.001-0.015), p = 0.013] and [0.015 AU (IQR 0.003-0.025) vs. 0.010 AU (IQR 0.001-0.015), p = 0.012], respectively. In Cox regression analysis, miR-20b-5p levels in the highest quartile were significantly associated with 90-day survival (adjusted hazard ratio 2.47 (95 % confidence interval 1.16-5.28), p = 0.019) when adjusted for CardShock Risk Score variables (age, confusion at presentation, previous myocardial infarction or coronary artery bypass grafting, acute coronary syndrome (ACS) etiology, left ventricular ejection fraction, lactate, and estimated glomerular filtration rate). A similar association of highest quartile miR-20b-5p levels with 90-day survival was also confirmed in ACS patient subcohort (79 % of CS patients). The results of this study indicate that circulating levels of miR-20b-5p at baseline could help in assessing in-hospital and 90-day survival in CS patients.
Collapse
Affiliation(s)
- Tuomas Mäntylä
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki and Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Chunguang Wang
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Mikko Hänninen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Katariina Immonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Toni Jäntti
- Department of Cardiology, University of Helsinki and Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Johan Lassus
- Department of Cardiology, University of Helsinki and Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Ilkka Tikkanen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kari Pulkki
- Department of Clinical Chemistry and Hematology, University of Helsinki and Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Veli-Pekka Harjola
- Division of Emergency Medicine, University of Helsinki, Department of Emergency Services and Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Päivi Lakkisto
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki and Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - CardShock Study Investigators1
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki and Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Department of Cardiology, University of Helsinki and Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
- Division of Emergency Medicine, University of Helsinki, Department of Emergency Services and Medicine, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
3
|
Kreutz J, Harbaum L, Barutcu CB, Rehman AS, Patsalis N, Mihali K, Chatzis G, Choukeir M, Syntila S, Schieffer B, Markus B. CytoSorb ® Hemadsorption in Cardiogenic Shock: A Real-World Analysis of Hemodynamics, Organ Function, and Clinical Outcomes During Mechanical Circulatory Support. Biomedicines 2025; 13:324. [PMID: 40002736 PMCID: PMC11853450 DOI: 10.3390/biomedicines13020324] [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: 12/26/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Cardiogenic shock (CS), characterized by inadequate tissue perfusion due to cardiac dysfunction, has a high mortality rate despite advances in treatment. Systemic inflammation and organ failure exacerbate the severity of CS. Extracorporeal hemadsorption techniques such as CytoSorb® have been introduced to control inflammation. However, evidence of their efficacy, particularly in patients on various mechanical circulatory support (MCS) systems, remains limited. Methods: This retrospective study analyzed data from 129 CS patients treated with CytoSorb® at the University Hospital of Marburg between August 2019 and December 2023. Those patients receiving MCS were grouped according to MCS type: (1) Impella, (2) VA-ECMO, and (3) ECMELLA. The hemodynamic parameters of circulatory support (e.g., MCS flow rates and vasoactive inotropic score, VIS) and laboratory and ventilation parameters were assessed 24 h before start of CytoSorb® therapy (T1) and 24 h after completion of CytoSorb® therapy (T2). Results: Of 129 CS patients (mean age: 64.7 ± 13.1 years), 103 (79.8%) received MCS. Comparing T1 and T2, there was a significant reduction in VIS in the entire cohort (T1: 38.0, T2: 16.3; p = 0.002), with a concomitant significant reduction in the level of MCS support in all subgroups, indicating successful weaning. Analysis of laboratory parameters showed significant reductions in lactate (T1: 2.1, T2: 1.3 mmol/L; p = 0.014), myoglobin (T1: 1549.0, T2: 618.0 µg/L; p < 0.01), lactate dehydrogenase (T1: 872.0, T2: 632.0 U/L; p = 0.048), and procalcitonin (T1: 2.9, T2: 1.6 µg/L; p < 0.001). However, a significant decrease in platelets (T1: 140.0, T2: 54.0 tsd/µL; p < 0.001) and albumin (T1: 25.0, T2: 22.0 g/dL; p < 0.001) was also documented. The median SOFA score of the entire cohort was 15.0 (IQR 12.0-16.0), predicting a mortality rate of >80%, which could be reduced to 60.5% in the present study. Conclusions: During CytoSorb® therapy in CS, a significant reduction in VIS was demonstrated, resulting in improved organ perfusion. Therefore, the results of this study underline that CytoSorb® therapy can be considered a useful "component" in the complex management of CS, especially when combined with MCS. To refine and optimize treatment strategies in CS, prospective studies are needed to better define the role of hemadsorption.
Collapse
Affiliation(s)
- Julian Kreutz
- Department of Cardiology, Angiology, and Intensive Care Medicine, University Hospital, Philipps University of Marburg, 35043 Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Nunez JI, Uehara M, Patel SR, Forest SJ, Rochlani Y, Madan S, Sims DB, Mellas N, Ashley JE, Rahmanian M, Carlese A, Goldstein DJ, Jorde UP, Saeed O. Changes in Neutrophil-to-Lymphocyte Ratio During Venoarterial Extracorporeal Membrane Oxygenation. ASAIO J 2025:00002480-990000000-00622. [PMID: 39817568 DOI: 10.1097/mat.0000000000002376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025] Open
Abstract
Mortality remains elevated during venoarterial extracorporeal membrane oxygenation support (VA-ECMO) for cardiogenic shock and the role of inflammation is uncertain. By using the neutrophil-to-lymphocyte ratio (NLR), we investigated inflammatory dynamics during VA-ECMO and their relation to clinical outcomes. A single-center, retrospective cohort study was conducted. Patients receiving steroids or on-device support for less than 48 hours were excluded. Patients were grouped as those who did and did not have a persistent rise in NLR during the 24-48 hour interval after VA-ECMO placement. Overall, 253 patients comprised the study cohort. In-hospital mortality was 56%. Neutrophil-to-lymphocyte ratio was higher at 24 hours after VA-ECMO placement compared to pre-ECMO (Δ4.36, interquartile range [IQR]: -0.23 to 8.61, p < 0.001). Persistent increase in NLR during the 24-48 hour interval after VA-ECMO placement was associated with higher in-hospital mortality (adjusted hazard ratio [aHR]: 1.51, 95% confidence interval [CI]: 1.02-2.25, p = 0.04). The magnitude of this rise in NLR was incrementally related to greater in-hospital mortality (Δ0-5: 72%, aHR: 1.61, 95% CI: 1.03-2.54, p = 0.039; Δ>5: 79%, aHR: 1.64, 95% CI: 1.03-2.63, p = 0.037) in comparison 52%, for those with a drop in NLR. Venoarterial extracorporeal membrane oxygenation exacerbates inflammation, as evident by a rise in NLR, which is progressively higher in nonsurvivors.
Collapse
Affiliation(s)
- Jose I Nunez
- From the Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Mayuko Uehara
- Departments of Cardiothoracic and Vascular Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Snehal R Patel
- From the Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Stephen J Forest
- Departments of Cardiothoracic and Vascular Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Yogita Rochlani
- From the Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Shivank Madan
- From the Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Daniel B Sims
- From the Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Nicholas Mellas
- Departments of Cardiothoracic and Vascular Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Justin E Ashley
- Departments of Cardiothoracic and Vascular Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Marjan Rahmanian
- Division of Critical Care Medicine and Pulmonology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Anthony Carlese
- Division of Critical Care Medicine and Pulmonology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Daniel J Goldstein
- Departments of Cardiothoracic and Vascular Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Ulrich P Jorde
- From the Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Omar Saeed
- From the Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
5
|
Jentzer JC, Berg DD, Chonde MD, Dahiya G, Elliott A, Rampersad P, Sinha SS, Truesdell AG, Yohannes S, Vallabhajosyula S. Mixed Cardiogenic-Vasodilatory Shock: Current Insights and Future Directions. JACC. ADVANCES 2025; 4:101432. [PMID: 39720581 PMCID: PMC11666941 DOI: 10.1016/j.jacadv.2024.101432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/05/2024] [Indexed: 12/26/2024]
Abstract
This state-of-the-art review describes the potential etiologies, pathophysiology, and management of mixed shock in the context of a proposed novel classification system. Cardiogenic-vasodilatory shock occurs when cardiogenic shock is complicated by inappropriate vasodilation, impairing compensatory mechanisms, and contributing to worsening shock. Vasodilatory-cardiogenic shock occurs when vasodilatory shock is complicated by myocardial dysfunction, resulting in low cardiac output. Primary mixed shock occurs when a systemic insult triggers both myocardial dysfunction and vasoplegia. Regardless of the etiology of mixed shock, the hemodynamic profile can be similar, and outcomes tend to be poor. Identification and treatment of both the initial and complicating disease processes is essential along with invasive hemodynamic monitoring given the evolving nature of mixed shock states. Hemodynamic support typically involves a combination of inotropes and vasopressors, with few data available to guide the use of mechanical circulatory support. Consensus definitions and novel treatment strategies are needed for this dangerous condition.
Collapse
Affiliation(s)
- Jacob C. Jentzer
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - David D. Berg
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meshe D. Chonde
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai, Los Angeles, California, USA
| | - Garima Dahiya
- Division of Pulmonary and Critical Care Medicine, Duke University, Durham, North Carolina, USA
| | - Andrea Elliott
- Department of Cardiology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Shashank S. Sinha
- Inova Schar Heart and Vascular, Inova Fairfax Medical Campus, Falls Church, Virginia, USA
| | - Alexander G. Truesdell
- Inova Schar Heart and Vascular, Inova Fairfax Medical Campus, Falls Church, Virginia, USA
| | - Seife Yohannes
- Department of Critical Care Medicine, Medstar Washington Hospital Center, Washington DC, USA
| | - Saraschandra Vallabhajosyula
- Division of Cardiology, Department of Medicine, Warren Alpert Medical School of Brown University, and Lifespan Cardiovascular Institute, Providence, Rhode Island, USA
| |
Collapse
|
6
|
Beaini H, Chunawala Z, Cheeran D, Araj F, Wrobel C, Truby L, Saha A, Thibodeau JT, Farr M. Cardiogenic Shock: Focus on Non-Cardiac Biomarkers. Curr Heart Fail Rep 2024; 21:604-614. [PMID: 39078556 DOI: 10.1007/s11897-024-00676-8] [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] [Accepted: 07/08/2024] [Indexed: 07/31/2024]
Abstract
PURPOSE OF REVIEW To examine the evolving multifaceted nature of cardiogenic shock (CS) in the context of non-cardiac biomarkers that may improve CS management and risk stratification. RECENT FINDINGS There are increasing data highlighting the role of lactate, glucose, and other markers of inflammation and end-organ dysfunction in CS. These biomarkers provide a more comprehensive understanding of the concurrent hemo-metabolic and cellular disturbances observed in CS and offer insights beyond standard structural and functional cardiac assessments. Non-cardiac biomarkers both refine the diagnostic accuracy and improve the prognostic assessments in CS. Further studies revolving around novel biomarkers are warranted to support more targeted and effective therapeutic and management interventions in these high-risk patients.
Collapse
Affiliation(s)
- Hadi Beaini
- Department of Medicine, The University of Texas Southwestern Medical Center, 5959 Harry Hines Blvd, Dallas, TX, 75235, USA
| | - Zainali Chunawala
- Department of Medicine, The University of Texas Southwestern Medical Center, 5959 Harry Hines Blvd, Dallas, TX, 75235, USA
- Parkland Memorial Hospital, Dallas, TX, USA
| | - Daniel Cheeran
- Department of Medicine, The University of Texas Southwestern Medical Center, 5959 Harry Hines Blvd, Dallas, TX, 75235, USA
- Dallas Veteran's Administration Hospital, Dallas, TX, USA
| | - Faris Araj
- Department of Medicine, The University of Texas Southwestern Medical Center, 5959 Harry Hines Blvd, Dallas, TX, 75235, USA
- Parkland Memorial Hospital, Dallas, TX, USA
| | - Christopher Wrobel
- Department of Medicine, The University of Texas Southwestern Medical Center, 5959 Harry Hines Blvd, Dallas, TX, 75235, USA
- Parkland Memorial Hospital, Dallas, TX, USA
| | - Lauren Truby
- Department of Medicine, The University of Texas Southwestern Medical Center, 5959 Harry Hines Blvd, Dallas, TX, 75235, USA
- Parkland Memorial Hospital, Dallas, TX, USA
| | - Amit Saha
- Department of Medicine, The University of Texas Southwestern Medical Center, 5959 Harry Hines Blvd, Dallas, TX, 75235, USA
- Parkland Memorial Hospital, Dallas, TX, USA
| | - Jennifer T Thibodeau
- Department of Medicine, The University of Texas Southwestern Medical Center, 5959 Harry Hines Blvd, Dallas, TX, 75235, USA
- Parkland Memorial Hospital, Dallas, TX, USA
| | - Maryjane Farr
- Department of Medicine, The University of Texas Southwestern Medical Center, 5959 Harry Hines Blvd, Dallas, TX, 75235, USA.
- Parkland Memorial Hospital, Dallas, TX, USA.
| |
Collapse
|
7
|
Lucinian YA, Martineau P, Abikhzer G, Harel F, Pelletier-Galarneau M. Novel tracers to assess myocardial inflammation with radionuclide imaging. J Nucl Cardiol 2024; 42:102012. [PMID: 39069249 DOI: 10.1016/j.nuclcard.2024.102012] [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: 02/16/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Myocardial inflammation plays a central role in the pathophysiology of various cardiac diseases. While FDG-PET is currently the primary method for molecular imaging of myocardial inflammation, its effectiveness is hindered by physiological myocardial uptake as well as its propensity for uptake by multiple disease-specific mechanisms. Novel radiotracers targeting diverse inflammatory immune cells and molecular pathways may provide unique insight through the visualization of underlying mechanisms central to the pathogenesis of inflammatory cardiac diseases, offering opportunities for increased understanding of immunocardiology. Moreover, the potentially enhanced specificity may lead to better quantification of disease activity, aiding in the guidance and monitoring of immunomodulatory therapy. This review aims to provide an update on advancements in non-FDG radiotracers for imaging myocardial inflammatory diseases, with a focus on cardiac sarcoidosis, myocarditis, and acute myocardial infarction.
Collapse
Affiliation(s)
| | | | - Gad Abikhzer
- Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
8
|
Wang Y, Zeller M, Auffret V, Georgiopoulos G, Räber L, Roffi M, Templin C, Muller O, Liberale L, Ministrini S, Stamatelopoulos K, Stellos K, Camici GG, Montecucco F, Rickli H, Maza M, Radovanovic D, Cottin Y, Chague F, Niederseer D, Lüscher TF, Kraler S. Sex-specific prediction of cardiogenic shock after acute coronary syndromes: the SEX-SHOCK score. Eur Heart J 2024; 45:4564-4578. [PMID: 39217456 PMCID: PMC11560280 DOI: 10.1093/eurheartj/ehae593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/05/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS Cardiogenic shock (CS) remains the primary cause of in-hospital death after acute coronary syndromes (ACS), with its plateauing mortality rates approaching 50%. To test novel interventions, personalized risk prediction is essential. The ORBI (Observatoire Régional Breton sur l'Infarctus) score represents the first-of-its-kind risk score to predict in-hospital CS in ACS patients undergoing percutaneous coronary intervention (PCI). However, its sex-specific performance remains unknown, and refined risk prediction strategies are warranted. METHODS This multinational study included a total of 53 537 ACS patients without CS on admission undergoing PCI. Following sex-specific evaluation of ORBI, regression and machine-learning models were used for variable selection and risk prediction. By combining best-performing models with highest-ranked predictors, SEX-SHOCK was developed, and internally and externally validated. RESULTS The ORBI score showed lower discriminative performance for the prediction of CS in females than males in Swiss (area under the receiver operating characteristic curve [95% confidence interval]: 0.78 [0.76-0.81] vs. 0.81 [0.79-0.83]; P =.048) and French ACS patients (0.77 [0.74-0.81] vs. 0.84 [0.81-0.86]; P = .002). The newly developed SEX-SHOCK score, now incorporating ST-segment elevation, creatinine, C-reactive protein, and left ventricular ejection fraction, outperformed ORBI in both sexes (females: 0.81 [0.78-0.83]; males: 0.83 [0.82-0.85]; P < .001), which prevailed following internal and external validation in RICO (females: 0.82 [0.79-0.85]; males: 0.88 [0.86-0.89]; P < .001) and SPUM-ACS (females: 0.83 [0.77-0.90], P = .004; males: 0.83 [0.80-0.87], P = .001). CONCLUSIONS The ORBI score showed modest sex-specific performance. The novel SEX-SHOCK score provides superior performance in females and males across the entire spectrum of ACS, thus providing a basis for future interventional trials and contemporary ACS management.
Collapse
Affiliation(s)
- Yifan Wang
- Center for Molecular Cardiology, University of Zurich, Wagistreet 12, 8952 Schlieren, Switzerland
| | - Marianne Zeller
- Department of Cardiology, CHU Dijon Bourgogne, Dijon, France
- Physiolopathologie et Epidémiologie Cérébro-Cardiovasculaire (PEC2), EA 7460, Univ Bourgogne, Dijon, France
| | - Vincent Auffret
- Inserm LTSI U1099, Université de Rennes 1, CHU Rennes Service de Cardiologie, Rennes, France
| | - Georgios Georgiopoulos
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Lorenz Räber
- Department of Cardiology, Swiss Heart Center, Inselspital Bern, Bern, Switzerland
| | - Marco Roffi
- Department of Cardiology, Geneva University Hospitals, Geneva, Switzerland
| | - Christian Templin
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Olivier Muller
- Department of Cardiology, Lausanne University Hospital-CHUV, Lausanne, Switzerland
| | - Luca Liberale
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, Genoa, Italy
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Wagistreet 12, 8952 Schlieren, Switzerland
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Konstantinos Stellos
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Mannheim, Germany
- Helmholtz Institute for Translational AngioCardioScience (HI-TAC), MDC, Heidelberg University, Heidelberg, Germany
- Faculty of Medical Sciences, Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Wagistreet 12, 8952 Schlieren, Switzerland
| | - Fabrizio Montecucco
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, Genoa, Italy
| | - Hans Rickli
- Cardiology Department, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Maud Maza
- Department of Cardiology, CHU Dijon Bourgogne, Dijon, France
| | - Dragana Radovanovic
- AMIS Plus Data Centre, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Yves Cottin
- Department of Cardiology, CHU Dijon Bourgogne, Dijon, France
| | - Frédéric Chague
- Department of Cardiology, CHU Dijon Bourgogne, Dijon, France
| | - David Niederseer
- Hochgebirgsklinik, Medicine Campus Davos, Herman-Burchard-Strasse 1, Davos 7270, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Medicine Campus Davos, Davos, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Wagistreet 12, 8952 Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, Heart Division and Cardiovascular Academic Group, King’s College, London, UK
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Wagistreet 12, 8952 Schlieren, Switzerland
- Department of Cardiology and Internal Medicine, Cantonal Hospital Baden, Im Ergel 1, 5404 Baden, Switzerland
| |
Collapse
|
9
|
Siopi SA, Antonitsis P, Karapanagiotidis GT, Tagarakis G, Voucharas C, Anastasiadis K. Cardiac Failure and Cardiogenic Shock: Insights Into Pathophysiology, Classification, and Hemodynamic Assessment. Cureus 2024; 16:e72106. [PMID: 39575019 PMCID: PMC11581444 DOI: 10.7759/cureus.72106] [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] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Heart failure is defined as increased intracardiac pressures, either alone or combined with reduced cardiac output. Clinically, it is presented with signs and symptoms of congestion and compensated perfusion. Cardiogenic shock, on the other hand, is the spectrum of hemodynamic disturbances that lead to hypoperfusion or need for circulatory support, due to cardiac disease. Both entities affect millions of people worldwide, have a dismal prognosis, and constitute a severe socioeconomic burden. Heart failure can be the aftermath of ischemic heart disease, hypertension, arrhythmias, or cardiomyopathies. It undergoes multiple classifications, facilitating its investigation and treatment. The pathogenetic mechanisms differ in various types of heart failure, regarding the affected ventricles, the duration of symptoms, and their primary/secondary onset. These mechanisms reflect the complex interactions between cardiopulmonary, vascular, and hepatorenal systems. Acute deterioration of cardiac function can lead to cardiogenic shock. Myocardial infarction accounts for 81% of such cases. Healthy lifestyle and timely management of coronary artery disease are paramount, as they can prevent this life-threatening situation and reduce mortality and the economic burden for healthcare systems. Irrespective of the etiology, cardiogenic shock is interpreted using the pressure-volume loop. This can be modified for each ventricle, the underlying pathophysiology, and the time since symptoms' onset. It therefore provides valuable information about the native circulation and the expected alterations under mechanical or pharmacological support, facilitating the decision-making progress. In 2019, given the phenotypical heterogeneity of cardiogenic shock, the Society for Cardiovascular Angiography and Interventions introduced a classification system. According to this, patients are stratified in five stages proportionally to the severity of their condition. Aside from this classification, various biochemical, imaging, and hemodynamic monitoring indices are used to assess coagulation pathway and cardiac, hepatorenal, and pulmonary function, enabling the heart team to tailor therapy. Additionally, the prognostication progress is facilitated by scores, such as the Observatoire Regional Breton sur l'Infarctus (ORBI) score, the intra-aortic balloon pump (IABP) SHOCK-II score, and the CardShock score, indicating suitable escalation or de-escalation strategies. Despite the current progress, there are several areas of advancement regarding the role of vasoactive drugs in cardiogenic shock, revascularization options, mechanical ventilation patterns, hypothermia treatment, and mechanical circulatory support protocols.
Collapse
Affiliation(s)
- Stavroula A Siopi
- Cardiovascular Medicine, Aristotle University of Thessaloniki, Thessaloniki, GRC
| | | | | | - Georgios Tagarakis
- Cardiothoracic Surgery, Aristotle University of Thessaloniki, Thessaloniki, GRC
| | - Christos Voucharas
- Cardiothoracic Surgery, Aristotle University of Thessaloniki, Thessaloniki, GRC
| | | |
Collapse
|
10
|
Del Buono MG, La Vecchia G, D'Aiello A, Pedicino D, Pinnacchio G, Genuardi L, Montone RA, Saponara G, Di Renzo A, Conte C, Cribari F, Moroni F, Filomia S, Brecciaroli M, Aurigemma C, Liuzzo G, Trani C, Burzotta F, Sanna T. Clinical Characteristics, Management, and Outcomes in Cardiogenic Shock: Insights From a High-Volume Italian Cardiac Intensive Care Unit. J Cardiovasc Pharmacol 2024; 84:210-219. [PMID: 39115720 PMCID: PMC11309343 DOI: 10.1097/fjc.0000000000001584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/12/2024] [Indexed: 08/10/2024]
Abstract
ABSTRACT Cardiogenic shock (CS) is a life-threatening condition. The aim of this study is to evaluate the clinical characteristics, management, and complication rate of patients with CS admitted to a high-volume hospital in Italy. We retrospectively reviewed the clinical, echocardiographic, and laboratory data, therapeutic management, and outcomes of patients with CS admitted to the Policlinico Gemelli (Rome) between January 1, 2020, and January 1, 2023. We included 96 patients [median age 71 years, interquartile range 60-79; 65 (68%) males], of whom 49 patients (51%) presented CS secondary to acute myocardial infarction and 60 (63%) with a de novo presentation of CS. Dobutamine was the most frequently used inotrope and noradrenaline the most frequently used vasopressor (adopted in 56% and 82% of cases, respectively). Forty-five (47%) patients died during the hospitalization. Nonsurvivors were older and had a higher inflammatory burden at admission, elevated lactate levels, a greater increase in lactate levels, higher left ventricular filling pressures, and worse right ventricular function. C-reactive protein levels [odds ratio (OR) 1.03, 95% confidence interval (CI) (1.00-1.04), P = 0.027], lactate levels at admission (OR 3.49, 95% CI, 1.59-7.63, P = 0.02), and increase in lactate levels (OR 2.8, 95% CI, 1.37-5.75, P = 0.005) were independent predictors of in-hospital all-cause death. Our data contribute to the assessment of the regional variations in the management and outcomes of patients with CS. We observed a high mortality and complication rate. Lactate acidosis and C-reactive protein measured at admission may help in identifying patients at higher risk of adverse in-hospital outcomes.
Collapse
Affiliation(s)
- Marco Giuseppe Del Buono
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulia La Vecchia
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Operative Unit of Diagnostic Interventional Cardiology, Isola Tiberina-Gemelli Isola, Rome, Italy; and
| | - Alessia D'Aiello
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Daniela Pedicino
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gaetano Pinnacchio
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lorenzo Genuardi
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rocco Antonio Montone
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gianluigi Saponara
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Di Renzo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cristina Conte
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesco Cribari
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesco Moroni
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, Division of Cardiovascular Medicine, Heart and Vascular Center, University of Virginia, Charlottesville, VA
| | - Simone Filomia
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mattia Brecciaroli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cristina Aurigemma
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanna Liuzzo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Trani
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesco Burzotta
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Tommaso Sanna
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
11
|
Isailă OM, Ion OM, Luta R, Catinas R, Ionita A, Haisan D, Hostiuc S. Postmortem Immunohistochemical Findings in Early Acute Myocardial Infarction: A Systematic Review. Int J Mol Sci 2024; 25:7625. [PMID: 39062865 PMCID: PMC11277133 DOI: 10.3390/ijms25147625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The diagnosis of early acute myocardial infarction is of particular importance in forensic practice considering the frequency of sudden cardiac death and the difficulty of positively identifying it through classical histological methods if survival is less than 6 h. This article aims to analyze potential immunohistochemical markers that could be useful in diagnosing acute myocardial infarction within the first 6 h of its onset. We conducted an extensive evaluation of the literature according to the PRISMA guidelines for reporting systematic literature reviews. We searched the Web of Science and PubMed databases from their inception to 2023 using the following keywords: "myocardial infarction" and "immunohistochemistry". Fifteen studies met the inclusion criteria. Immunohistochemical markers as complement factors and CD59, myoglobin, fibrinogen, desmin, tumor necrosis factor alpha (TNF-α), P-38, JNK (Jun N Terminal Kinase), transforming growth factor β1 (TGF-β1), cardiac troponins, fibronectin, H-FABP (heart fatty acid binding protein), dityrosine, fibronectin, CD15, IL-1β, IL-6, IL-15, IL-8, MCP-1, ICAM-1, CD18, and tryptase can be used to identify the first six hours of acute myocardial infarction. These markers are mostly studied in experimental animal models. It is necessary to conduct extensive studies on human myocardial tissue fragments, which will involve the analysis of several immunohistochemical markers and careful analysis of the available data on perimortem events, resuscitation, and postmortem intervals in the context of a uniform laboratory methodology.
Collapse
Affiliation(s)
- Oana-Maria Isailă
- Faculty of Dental Medicine, Department of Legal Medicine and Bioethics, University of Medicine and Pharmacy ”Carol Davila” Bucharest, 050474 Bucharest, Romania; (O.-M.I.); (O.M.I.)
- National Institute of Legal Medicine “Mina Minovici”, 042122 Bucharest, Romania; (R.L.); (R.C.); (A.I.); (D.H.)
| | - Oana Mihaela Ion
- Faculty of Dental Medicine, Department of Legal Medicine and Bioethics, University of Medicine and Pharmacy ”Carol Davila” Bucharest, 050474 Bucharest, Romania; (O.-M.I.); (O.M.I.)
- National Institute of Legal Medicine “Mina Minovici”, 042122 Bucharest, Romania; (R.L.); (R.C.); (A.I.); (D.H.)
| | - Robert Luta
- National Institute of Legal Medicine “Mina Minovici”, 042122 Bucharest, Romania; (R.L.); (R.C.); (A.I.); (D.H.)
| | - Raluca Catinas
- National Institute of Legal Medicine “Mina Minovici”, 042122 Bucharest, Romania; (R.L.); (R.C.); (A.I.); (D.H.)
| | - Ana Ionita
- National Institute of Legal Medicine “Mina Minovici”, 042122 Bucharest, Romania; (R.L.); (R.C.); (A.I.); (D.H.)
| | - Diana Haisan
- National Institute of Legal Medicine “Mina Minovici”, 042122 Bucharest, Romania; (R.L.); (R.C.); (A.I.); (D.H.)
| | - Sorin Hostiuc
- Faculty of Dental Medicine, Department of Legal Medicine and Bioethics, University of Medicine and Pharmacy ”Carol Davila” Bucharest, 050474 Bucharest, Romania; (O.-M.I.); (O.M.I.)
- National Institute of Legal Medicine “Mina Minovici”, 042122 Bucharest, Romania; (R.L.); (R.C.); (A.I.); (D.H.)
| |
Collapse
|
12
|
Baldetti L, Gallone G, Filiberti G, Pescarmona L, Cesari A, Rizza V, Roagna E, Gurrieri D, Peveri B, Nocera L, Cianfanelli L, Marcelli G, De Lio G, Boretto P, Angelini F, Gramegna M, Pazzanese V, Sacchi S, Calvo F, Ajello S, De Ferrari GM, Frea S, Scandroglio AM. Mixed Shock Complicating Cardiogenic Shock: Frequency, Predictors, and Clinical Outcomes. Circ Heart Fail 2024; 17:e011404. [PMID: 38979611 DOI: 10.1161/circheartfailure.123.011404] [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: 11/10/2023] [Accepted: 03/11/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Patients presenting with cardiogenic shock (CS) are at risk of developing mixed shock (MS), characterized by distributive-inflammatory phenotype. However, no objective definition exists for this clinical entity. METHODS We assessed the frequency, predictors, and prognostic relevance of MS complicating CS, based on a newly proposed objective definition. MS complicating CS was defined as an objective shock state secondary to both an ongoing cardiogenic cause and a distributive-inflammatory phenotype arising at least 12 hours after the initial CS diagnosis, as substantiated by predefined longitudinal changes in hemodynamics, clinical, and laboratory parameters. RESULTS Among 213 consecutive patients admitted at 2 cardiac intensive care units with CS, 13 with inflammatory-distributive features at initial presentation were excluded, leading to a cohort of 200 patients hospitalized with pure CS (67±13 years, 96% Society of Cardiovascular Angiography and Interventions CS stage class C or higher). MS complicating CS occurred in 24.5% after 120 (29-216) hours from CS diagnosis. Lower systolic arterial pressure (P=0.043), hepatic injury (P=0.049), and suspected/definite infection (P=0.013) at CS diagnosis were independent predictors of MS development. In-hospital mortality (53.1% versus 27.8%; P=0.002) and hospital stay (21 [13-48] versus 17 [9-27] days; P=0.018) were higher in the MS cohort. At logistic multivariable analysis, MS diagnosis (odds ratio [OR], 3.00 [95% CI, 1.39-6.63]; Padj=0.006), age (OR, 1.06 [95% CI, 1.03-1.10] years; Padj<0.001), admission systolic arterial pressure <100 mm Hg (OR, 2.41 [95% CI, 1.19-4.98]; Padj=0.016), and admission serum creatinine (OR, 1.61 [95% CI, 1.19-2.26]; Padj=0.003) conferred higher odds of in-hospital death, while early temporary mechanical circulatory support was associated with lower in-hospital death (OR, 0.36 [95% CI, 0.17-0.75]; Padj=0.008). CONCLUSIONS MS complicating CS, objectively defined leveraging on longitudinal changes in distributive and inflammatory features, occurs in one-fourth of patients with CS, is predicted by markers of CS severity and inflammation at CS diagnosis, and portends higher hospital mortality.
Collapse
Affiliation(s)
- Luca Baldetti
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Guglielmo Gallone
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Gaia Filiberti
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Luca Pescarmona
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Andrea Cesari
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Vincenzo Rizza
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Edoardo Roagna
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Davide Gurrieri
- Mathematics Department, Polytechnic University of Milan, Italy (D.G.)
| | - Beatrice Peveri
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Lorenzo Nocera
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Lorenzo Cianfanelli
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Gianluca Marcelli
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Giulia De Lio
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
| | - Paolo Boretto
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
| | - Filippo Angelini
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
| | | | - Vittorio Pazzanese
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Stefania Sacchi
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Francesco Calvo
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Silvia Ajello
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Gaetano Maria De Ferrari
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Simone Frea
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
| | - Anna Mara Scandroglio
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| |
Collapse
|
13
|
Levy B, Girerd N, Duarte K, Antoine ML, Monzo L, Ouattara A, Delmas C, Brodie D, Combes A, Kimmoun A, Baudry G. Hypothermia in patients with cardiac arrest prior to ECMO-VA: Insight from the HYPO-ECMO trial. Resuscitation 2024; 200:110235. [PMID: 38762081 DOI: 10.1016/j.resuscitation.2024.110235] [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: 03/19/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 05/20/2024]
Abstract
AIM Venoarterial extracorporeal membrane oxygenation (VA-ECMO) has become a common intervention for patients with cardiogenic shock (CS), often complicated by cardiac arrest (CA). Moderate hypothermia (MH) has shown promise in mitigating ischemia-reperfusion injury following CA. The HYPO-ECMO trial aimed to compare the effect of MH versus normothermia in refractory CS rescued by VA-ECMO. The primary aim of this non-predefined post hoc study was to assess the treatment effect of MH in the subgroup of patients with cardiac arrest (CA) within the HYPO-ECMO trial. Additionally, we will evaluate the prognostic significance of CA in these patients. METHODS This post hoc analysis utilized data from the randomized HYPO-ECMO trial conducted across 20 French cardiac shock care centers between October 2016 and July 2019. Participants included intubated patients receiving VA-ECMO for CS for less than 6 h, with 334 patients completing the trial. Patients were randomized to early MH (33-34 °C) or normothermia (36-37 °C) for 24 h. RESULTS Of the 334 patients, 159 (48%) experienced preceding CA. Mortality in the CA group was 50.9% at 30 days and 59.1% at 180 days, compared to 42.3% and 51.4% in the no-CA group, respectively (adjusted risk difference [RD] at 30 days, 8.1% [-0.8 to 17.1%], p = 0.074 and RD at 180 days 7.0% [-3.0 to 16.9%], p = 0.17). MH was associated with a significant reduction in primary (RD -13.3% [-16.3 to -0.3%], p = 0.031) and secondary outcomes in the CA group only (p < 0.025 for all), with a significant interaction between MH and CA status for 180-day mortality [p = 0.03]. CONCLUSIONS This post hoc analysis suggests that MH shows potential for reducing mortality and composite endpoints in patients with cardiac arrest and refractory CS treated with VA-ECMO without an increased risk of severe bleeding or infection. Further research is needed to validate these findings and elucidate underlying mechanisms.
Collapse
Affiliation(s)
- Bruno Levy
- Médecine Intensive et Réanimation, CHRU Nancy, Pôle Cardio-Médico-Chirurgical, Vandoeuvre-les-Nancy, France; INSERM U1116, Faculté de Médecine, Vandoeuvre-les-Nancy, France; Université de Lorraine, Nancy, France.
| | - Nicolas Girerd
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique, INSERM 1433, CHRU de Nancy, Institut Lorrain du Coeur et des Vaisseaux, Nancy, France; INI-CRCT (Cardiovascular and Renal Clinical Trialists), F-CRIN Network, Nancy, France
| | - Kevin Duarte
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique, INSERM 1433, CHRU de Nancy, Institut Lorrain du Coeur et des Vaisseaux, Nancy, France; INI-CRCT (Cardiovascular and Renal Clinical Trialists), F-CRIN Network, Nancy, France
| | - Marie-Lauren Antoine
- Centre Régional de Pharmacovigilance de Nancy, Vigilance des Essais Cliniques - CHRU Nancy-Brabois, France
| | - Luca Monzo
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique, INSERM 1433, CHRU de Nancy, Institut Lorrain du Coeur et des Vaisseaux, Nancy, France; INI-CRCT (Cardiovascular and Renal Clinical Trialists), F-CRIN Network, Nancy, France
| | - Alexandre Ouattara
- CHU Bordeaux, Department of Anaesthesia and Critical Care, Magellan Medico-Surgical Centre, Bordeaux, France; University Bordeaux, INSERM, UMR 1034, Biology of Cardiovascular Diseases, Pessac, France
| | - Clément Delmas
- Intensive Cardiac Care Unit, Rangueil University Hospital, Toulouse, France; REICATRA, Université de Lorraine, Nancy, France
| | - Daniel Brodie
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Alain Combes
- Sorbonne Université, INSERM, UMRS_1166-ICAN, Institute of Cardiometabolism and Nutrition, Paris, France; Service de Médecine Intensive-Réanimation, Institut de Cardiologie, APHP Sorbonne Université Hôpital Pitié-Salpêtrière, Paris, France
| | - Antoine Kimmoun
- Médecine Intensive et Réanimation, CHRU Nancy, Pôle Cardio-Médico-Chirurgical, Vandoeuvre-les-Nancy, France; INSERM U1116, Faculté de Médecine, Vandoeuvre-les-Nancy, France; Université de Lorraine, Nancy, France
| | - Guillaume Baudry
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique, INSERM 1433, CHRU de Nancy, Institut Lorrain du Coeur et des Vaisseaux, Nancy, France; INI-CRCT (Cardiovascular and Renal Clinical Trialists), F-CRIN Network, Nancy, France; REICATRA, Université de Lorraine, Nancy, France
| |
Collapse
|
14
|
Masroor M, Ansari MI, Umair M, Taimoor L, Hassan M, Arif MS, Karim M, Abubaker J. Steroids and myocardial infarction: Investigating safety and short-term mortality in critical post-myocardial infarction patients. Am J Med Sci 2024; 368:40-47. [PMID: 38395147 DOI: 10.1016/j.amjms.2024.02.007] [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: 08/10/2023] [Revised: 01/16/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Conventionally, in the pre-percutaneous intervention era, free wall rupture is reported to be a major concern for using steroids in myocardial infarction (MI) patients. Therefore, the aim of this study was to evaluate the safety of the use of steroids in critically ill post-MI patients in terms of hospital course and short-term (up to 180-day) mortality. METHODS We included patients admitted to CCU diagnosed with MI, undergone revascularization, critically ill, and requiring mechanical ventilator (MV) support. The hospital course and short-term (up to 180-day) mortality were independently compared between steroid and non-steroid cohorts and propensity-matched non-steroid cohorts. RESULTS A total of 312 patients were included, out of which steroids were used in 93 (29.8%) patients during their management. On periodic bedside echocardiography, no free wall rupture was documented in the steroid or non-steroid cohort. When compared steroids with a propensity-matched non-steroid cohort, MV duration >24 h was 66.7% vs. 59.1%; p = 0.288, major bleeding was 6.5% vs. 3.2%; p = 0.305, need for renal replacement therapy was 9.7% vs. 8.6%; p = 0.799, in-hospital mortality was 35.5% vs. 23.7%; p = 0.077, and 180-day mortality was 48.4% vs. 41.9%; p = 0.377, respectively. The hazard ratio was 1.22 [95% CI: 0.80 to 1.88] compared to the propensity-matched non-steroid cohort. The ejection fraction (%) was found to be the independent predictor of 180-day mortality with an adjusted odds ratio of 0.92 [95% CI: 0.86 to 0.98]. CONCLUSIONS In conclusion, using steroids is safe in post-MI patients with no significant increase in short-term mortality risk.
Collapse
Affiliation(s)
- Madiha Masroor
- National Institute of Cardiovascular Diseases, Rafiqui (H.J.) Shaheed Road, Karachi 75510, Pakistan.
| | - Muhammad Imran Ansari
- National Institute of Cardiovascular Diseases, Rafiqui (H.J.) Shaheed Road, Karachi 75510, Pakistan
| | - Madiha Umair
- National Institute of Cardiovascular Diseases, Rafiqui (H.J.) Shaheed Road, Karachi 75510, Pakistan
| | - Lalarukh Taimoor
- National Institute of Cardiovascular Diseases, Rafiqui (H.J.) Shaheed Road, Karachi 75510, Pakistan
| | - Mujtaba Hassan
- National Institute of Cardiovascular Diseases, Rafiqui (H.J.) Shaheed Road, Karachi 75510, Pakistan
| | - Muhammad Sohaib Arif
- National Institute of Cardiovascular Diseases, Rafiqui (H.J.) Shaheed Road, Karachi 75510, Pakistan
| | - Musa Karim
- National Institute of Cardiovascular Diseases, Rafiqui (H.J.) Shaheed Road, Karachi 75510, Pakistan
| | - Jawed Abubaker
- National Institute of Cardiovascular Diseases, Rafiqui (H.J.) Shaheed Road, Karachi 75510, Pakistan
| |
Collapse
|
15
|
Peng C, Lu Y, Li R, Zhang L, Liu Z, Xu X, Wang C, Hu R, Tan W, Zhou L, Wang Y, Yu L, Wang Y, Tang B, Jiang H. Neuroimmune modulation mediated by IL-6: A potential target for the treatment of ischemia-induced ventricular arrhythmias. Heart Rhythm 2024; 21:610-619. [PMID: 38160759 DOI: 10.1016/j.hrthm.2023.12.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/09/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Neural remodeling in the left stellate ganglion (LSG), as mediated by neuroimmune reactions, promotes cardiac sympathetic nerve activity (SNA) and thus increases the incidence of ventricular arrhythmias (VAs). Interleukin-6 (IL-6) is an important factor of the neuroimmune interaction. OBJECTIVE The present study explored the effects of IL-6 on LSG hyperactivity and the incidence of VAs. METHODS Eighteen beagles were randomly allocated to a control group (saline with myocardial infarction [MI], n = 6), adeno-associated virus (AAV) group (AAV with MI, n = 6), and IL-6 group (overexpression of IL-6 via AAV vector with MI, n = 6). Ambulatory electrocardiography was performed before and 30 days after AAV microinjection into the LSG. LSG function and ventricular electrophysiology were assessed at 31 days after surgery, and a canine MI model was established. Samples of the LSG were collected for immunofluorescence staining and molecular biological evaluation. Blood samples and 24-hour Holter data were obtained from 24 patients with acute MI on the day after they underwent percutaneous coronary intervention to assess the correlation between IL-6 levels and SNA. RESULTS IL-6 overexpression increased cardiac SNA and worsened postinfarction VAs. Furthermore, sustained IL-6 overexpression enhanced LSG function, promoted expression of nerve growth factor, c-fos, and fos B in the LSG, and activated the signal transducer and activator of transcription 3/regulator of G protein signalling 4 signaling pathway. Clinical sample analysis revealed a correlation between serum IL-6 levels and heart rate variability frequency domain index as well as T-wave alternans. CONCLUSION IL-6 levels are correlated with cardiac SNA. Chronic overexpression of IL-6 mediates LSG neural remodeling through the signal transducer and activator of transcription 3/regulator of G protein signalling 4 signaling pathway, elevating the risk of VA after MI.
Collapse
Affiliation(s)
- Chen Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Yanmei Lu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Rui Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Ling Zhang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Xiao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Changyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Ruijie Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Yueyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Yuhong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Baopeng Tang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Autonomic Nervous System Modulation, Wuhan, P.R. China; Cardiac Autonomic Nervous System Research Center of Wuhan University, Wuhan, P.R. China; Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan, P.R. China; Hubei Key Laboratory of Cardiology, Wuhan, P.R. China; Cardiovascular Research Institute of Wuhan University, Wuhan, P.R. China; Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urmuqi, P.R. China.
| |
Collapse
|
16
|
Gregers E, Frederiksen PH, Udesen NLJ, Linde L, Banke A, Povlsen AL, Larsen JP, Hassager C, Jensen LO, Lassen JF, Schmidt H, Ravn HB, Heegaard PMH, Møller JE. Immediate inflammatory response to mechanical circulatory support in a porcine model of severe cardiogenic shock. Intensive Care Med Exp 2024; 12:39. [PMID: 38647741 PMCID: PMC11035503 DOI: 10.1186/s40635-024-00625-8] [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: 01/16/2024] [Accepted: 04/14/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND In selected cases of cardiogenic shock, veno-arterial extracorporeal membrane oxygenation (V-A ECMO) is combined with trans valvular micro axial flow pumps (ECMELLA). Observational studies indicate that ECMELLA may reduce mortality but exposing the patient to two advanced mechanical support devices may affect the early inflammatory response. We aimed to explore inflammatory biomarkers in a porcine cardiogenic shock model managed with V-A ECMO or ECMELLA. METHODS Fourteen landrace pigs had acute myocardial infarction-induced cardiogenic shock with minimal arterial pulsatility by microsphere embolization and were afterwards managed 1:1 with either V-A ECMO or ECMELLA for 4 h. Serial blood samples were drawn hourly and analyzed for serum concentrations of interleukin 6 (IL-6), IL-8, tumor necrosis factor alpha, and serum amyloid A (SAA). RESULTS An increase in IL-6, IL-8, and SAA levels was observed during the experiment for both groups. At 2-4 h of support, IL-6 levels were higher in ECMELLA compared to V-A ECMO animals (difference: 1416 pg/ml, 1278 pg/ml, and 1030 pg/ml). SAA levels were higher in ECMELLA animals after 3 and 4 h of support (difference: 401 ng/ml and 524 ng/ml) and a significant treatment-by-time effect of ECMELLA on SAA was identified (p = 0.04). No statistical significant between-group differences were observed in carotid artery blood flow, urine output, and lactate levels. CONCLUSIONS Left ventricular unloading with Impella during V-A ECMO resulted in a more extensive inflammatory reaction despite similar end-organ perfusion.
Collapse
Affiliation(s)
- Emilie Gregers
- Department of Cardiology, The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen O, Denmark.
| | | | - Nanna L J Udesen
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Louise Linde
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Ann Banke
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Amalie L Povlsen
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Jeppe P Larsen
- Department of Cardiothoracic Anesthesiology, Odense University Hospital, Odense, Denmark
| | - Christian Hassager
- Department of Cardiology, The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen O, Denmark
| | - Lisette O Jensen
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Jens F Lassen
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Henrik Schmidt
- Department of Cardiothoracic Anesthesiology, Odense University Hospital, Odense, Denmark
| | - Hanne B Ravn
- Department of Cardiothoracic Anesthesiology, Odense University Hospital, Odense, Denmark
| | - Peter M H Heegaard
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Jacob E Møller
- Department of Cardiology, The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen O, Denmark
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
17
|
Schupp T, Rusnak J, Egner-Walter S, Ruka M, Dudda J, Bertsch T, Müller J, Mashayekhi K, Tajti P, Ayoub M, Akin I, Behnes M. Prognosis of cardiogenic shock with and without acute myocardial infarction: results from a prospective, monocentric registry. Clin Res Cardiol 2024; 113:626-641. [PMID: 37093246 DOI: 10.1007/s00392-023-02196-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023]
Abstract
OBJECTIVE The study investigates the prognostic impact of cardiogenic shock (CS) stratified by the presence or absence of acute myocardial infarction (AMI). BACKGROUND Intensive care unit (ICU) related mortality in CS patients remains unacceptably high despite improvement concerning the treatment of CS patients. METHODS Consecutive patients with CS from 2019 to 2021 were included monocentrically. The prognostic impact of CS related to AMI was compared to patients without AMI-related CS. The primary endpoint was 30-day all-cause mortality. Statistical analyses included Kaplan-Meier analyses, multivariable Cox proportional regression analyses and propensity score matching. RESULTS 273 CS patients were included (AMI-related CS: 49%; non-AMI-related CS: 51%). The risk of 30-day all-cause mortality was increased in patients with AMI-related CS (64% vs. 47%; HR = 1.653; 95% CI 1.199-2.281; p = 0.002), which was still observed after multivariable adjustment (HR = 1.696; 95% CI 1.153-2.494; p = 0.007). Even after propensity score matching (i.e., 87 matched pairs), AMI was still an independent predictor of 30-day mortality (HR = 1.524; 95% CI 1.020-2.276; p = 0.040). In contrast, non-ST-segment AMI (NSTEMI) and STEMI were associated with comparable prognosis (log-rank p = 0.528). CONCLUSION AMI-related CS was associated with increased 30-day all-cause mortality compared to patients with CS not related to AMI. In contrast, the prognosis of STEMI- and NSTEMI-CS patients was comparable.
Collapse
Affiliation(s)
- Tobias Schupp
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, First Department of Medicine, University Medical Center Mannheim (UMM), Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Jonas Rusnak
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, First Department of Medicine, University Medical Center Mannheim (UMM), Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Sascha Egner-Walter
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, First Department of Medicine, University Medical Center Mannheim (UMM), Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Marinela Ruka
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, First Department of Medicine, University Medical Center Mannheim (UMM), Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Jonas Dudda
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, First Department of Medicine, University Medical Center Mannheim (UMM), Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Thomas Bertsch
- Institute of Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Julian Müller
- Clinic for Interventional Electrophysiology, Heart Centre Bad Neustadt, Bad Neustadt a. d. Saale, Germany
- Department of Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany
| | - Kambis Mashayekhi
- Department of Internal Medicine and Cardiology, Mediclin Heart Centre Lahr, Lahr, Germany
| | - Péter Tajti
- Gottsegen György National Cardiovascular Center, Budapest, Hungary
| | - Mohammed Ayoub
- Division of Cardiology and Angiology, Heart Center University of Bochum-Bad, Oeynhausen, Germany
| | - Ibrahim Akin
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, First Department of Medicine, University Medical Center Mannheim (UMM), Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Michael Behnes
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, First Department of Medicine, University Medical Center Mannheim (UMM), Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany.
| |
Collapse
|
18
|
Mhanna M, Al-Abdouh A, Sauer MC, Jabri A, Abusnina W, Safi M, Beran A, Mansour S. Hypothermia as an Adjunctive Therapy in Cardiogenic Shock: A Systematic Review and Meta-Analysis. Ther Hypothermia Temp Manag 2023; 13:160-169. [PMID: 37074118 DOI: 10.1089/ther.2023.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
In the setting of out-of-hospital cardiac arrest, therapeutic hypothermia (TH) has been shown to improve clinical outcomes. However, trials showing the advantage of TH did not include patients with cardiogenic shock (CS). We performed a comprehensive literature search for studies that evaluated the efficacy and safety of adjunctive TH compared with the standard of care (SOC) in patients with CS. The primary outcome was the mortality rate (in-hospital, short-, and mid-term). The secondary outcomes were the TH-related complications, duration of Intensive Care Unit (ICU) stay, duration of mechanical ventilation (MV-days), and improvement in cardiac function. Relative risk (RR) or the standardized mean difference (SMD) and corresponding 95% confidence intervals (CIs) were calculated using the random-effects model. A total of 7 clinical studies (3 RCTs included), and 712 patients (341 in the TH group and 371 in the SOC group) were included. As compared with the SOC, TH was not associated with a statistically significant improvement in the in-hospital (RR: 0.73%, 95% CI: 0.51-1.03; p = 0.08), short-term (RR: 0.90%, 95% CI: 0.75-1.06; p = 0.21), or mid-term (RR: 0.93%, 95% CI: 0.78-1.10; p = 0.38) mortality rates. Despite the improvement in the cardiac function in the TH group (SMD: 1.08, 95% CI: 0.02-2.1; p = 0.04), the TH strategy did not significantly shorten the MV days, or the ICU stay (p-values >0.05). Finally, there was a trend toward higher risks for infection, major bleeding, and the need for blood transfusion in the TH group. According to our meta-analysis of published clinical studies, TH is not beneficial in patients with CS and has a marginal safety profile. Larger-scale RCTs are needed to further clarify our results.
Collapse
Affiliation(s)
- Mohammed Mhanna
- Division of Cardiology, Department of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Ahmad Al-Abdouh
- Department of Internal Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Michael C Sauer
- Department of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Ahmad Jabri
- Department of Cardiology, Case Western Reserve University/MetroHealth Medical Center, Cleveland, Ohio, USA
| | - Waiel Abusnina
- Division of Cardiology, Creighton University, Omaha, Nebraska, USA
| | - Mohammed Safi
- Department of Internal Medicine, The University of Toledo, Toledo, Ohio, USA
| | - Azizullah Beran
- Department of Gastroenterology, Indiana University, Indianapolis, Indiana, USA
| | - Shareef Mansour
- Division of Cardiology, Department of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
19
|
Buckel M, Maclean P, Knight JC, Lawler PR, Proudfoot AG. Extending the 'host response' paradigm from sepsis to cardiogenic shock: evidence, limitations and opportunities. Crit Care 2023; 27:460. [PMID: 38012789 PMCID: PMC10683227 DOI: 10.1186/s13054-023-04752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
Recent clinical and research efforts in cardiogenic shock (CS) have largely focussed on the restoration of the low cardiac output state that is the conditio sine qua non of the clinical syndrome. This approach has failed to translate into improved outcomes, and mortality has remained static at 30-50%. There is an unmet need to better delineate the pathobiology of CS to understand the observed heterogeneity of presentation and treatment effect and to identify novel therapeutic targets. Despite data in other critical illness syndromes, specifically sepsis, the role of dysregulated inflammation and immunity is hitherto poorly described in CS. High-dimensional molecular profiling, particularly through leukocyte transcriptomics, may afford opportunity to better characterise subgroups of patients with shared mechanisms of immune dysregulation. In this state-of-the-art review, we outline the rationale for considering molecular subtypes of CS. We describe how high-dimensional molecular technologies can be used to identify these subtypes, and whether they share biological features with sepsis and other critical illness states. Finally, we propose how the identification of molecular subtypes of patients may enrich future clinical trial design and identification of novel therapies for CS.
Collapse
Affiliation(s)
- Marie Buckel
- Department of Perioperative Medicine, Bart's Heart Centre, St. Bartholomew's Hospital, London, UK
| | - Patrick Maclean
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Julian C Knight
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Patrick R Lawler
- Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, ON, Canada
- McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Alastair G Proudfoot
- Department of Perioperative Medicine, Bart's Heart Centre, St. Bartholomew's Hospital, London, UK.
- Queen Mary University of London, London, UK.
| |
Collapse
|
20
|
Review of Pathophysiology of Cardiogenic Shock and Escalation of Mechanical Circulatory Support Devices. Curr Cardiol Rep 2023; 25:213-227. [PMID: 36847990 DOI: 10.1007/s11886-023-01843-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2023] [Indexed: 03/01/2023]
Abstract
PURPOSE OF REVIEW Cardiogenic shock (CS) is a complex clinical entity that continues to carry a high risk of mortality. The landscape of CS management has changed with the advent of several temporary mechanical circulatory support (MCS) devices designed to provide hemodynamic support. It remains challenging to understand the role of different temporary MCS devices in patients with CS, as many of these patients are critically ill, requiring complex care with multiple MCS device options. Each temporary MCS device can provide different types and levels of hemodynamic support. It is important to understand the risk/benefit profile of each one of them for appropriate device selection in patients with CS. RECENT FINDINGS MCS may be beneficial in CS patients through augmentation of cardiac output with subsequent improvement of systemic perfusion. Selecting the optimal MCS device depends on several variables including the underlying etiology of CS, clinical strategy of MCS use (bridge to recovery, bridge to transplant or durable MCS, or abridge to decision), amount of hemodynamic support needed, associated respiratory failure, and institutional preference. Furthermore, it is even more challenging to determine the appropriate time to escalate from one MCS device to another or combine different MCS devices. In this review, we discuss the current available data published in the literature on the management of CS and propose a standardized approach for escalation of MCS devices in patients with CS. Shock teams can play an important role to help in hemodynamic-guided management and algorithm-based step-by-step approach in early initiation and escalation of temporary MCS devices at different stages of CS. It is important to define the etiology of CS, and stage of shock and recognize univentricular vs biventricular shock for appropriate device selection and escalation of therapy.
Collapse
|
21
|
Management of Cardiogenic Shock Unrelated to Acute Myocardial Infarction. Can J Cardiol 2023; 39:406-419. [PMID: 36731605 DOI: 10.1016/j.cjca.2023.01.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 02/01/2023] Open
Abstract
Cardiogenic shock is an extreme manifestation of acute decompensated heart failure. Cardiogenic shock is often caused by-and has traditionally been studied in the setting of-acute myocardial infarction (AMI CS); however, there is increasing incidence and recognition of cardiogenic shock not associated with acute myocardial infarction (non-AMI CS) as a distinct entity. Despite decades of study and technologic advancements, cardiogenic shock mortality remains as high as 50%, regardless of etiology. New approaches to shock phenotyping and classification have emerged, with a focus on appropriately matching patient physiology to a growing list of available interventions. Further study is needed to determine whether these efforts will lead to more nuanced use of mechanical circulatory support and improved patient outcomes, especially in non-AMI CS. In the meantime, models of care incorporating multidisciplinary decision making, such as shock teams, may improve patient selection and outcomes.
Collapse
|
22
|
Bruno RR, Wolff G, Kelm M, Jung C. Pharmacological treatment of cardiogenic shock - A state of the art review. Pharmacol Ther 2022; 240:108230. [PMID: 35697151 DOI: 10.1016/j.pharmthera.2022.108230] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 12/14/2022]
Abstract
Cardiogenic shock is a clinical syndrome of impaired tissue perfusion caused by primary cardiac dysfunction and inadequate cardiac output. It represents one of the most lethal clinical conditions in intensive care medicine with mortality >40%. Management of different clinical presentations of cardiogenic shock includes guidance of cardiac preload, afterload, heart rate and contractility by differential pharmacological modulation of volume, systemic and pulmonary vascular resistance and cardiac output besides reversing the triggering cause. Data from large registries and randomized controlled trials on optimal diagnostic guidance as well as choice of pharmacological agents has accrued significantly in recent years. This state-of-the-art review summarizes the basic concepts of cardiogenic shock, the diagnostic work-up and currently available evidence and guideline recommendations on pharmacological treatment of cardiogenic shock.
Collapse
Affiliation(s)
- Raphael Romano Bruno
- Heinrich-Heine-University Duesseldorf, Medical Faculty, Department of Cardiology, Pulmonology and Vascular Medicine, Duesseldorf, Germany
| | - Georg Wolff
- Heinrich-Heine-University Duesseldorf, Medical Faculty, Department of Cardiology, Pulmonology and Vascular Medicine, Duesseldorf, Germany
| | - Malte Kelm
- Heinrich-Heine-University Duesseldorf, Medical Faculty, Department of Cardiology, Pulmonology and Vascular Medicine, Duesseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Duesseldorf, Germany
| | - Christian Jung
- Heinrich-Heine-University Duesseldorf, Medical Faculty, Department of Cardiology, Pulmonology and Vascular Medicine, Duesseldorf, Germany.
| |
Collapse
|
23
|
Sevdımbas S, Satar S, Gulen M, Acehan S, Acele A, Koksaldı Sahin G, Aka Satar D. Blood urea nitrogen/albumin ratio on admission predicts mortality in patients with non ST segment elevation myocardial infarction. Scandinavian Journal of Clinical and Laboratory Investigation 2022; 82:454-460. [PMID: 36128642 DOI: 10.1080/00365513.2022.2122075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The aim of this study is to reveal the predictive power of biomarkers and SYNTAX (SX) score for short-term mortality in patients diagnosed with non-ST-segment elevation myocardial infarction (NSTEMI) in the emergency department. This is prospective observational cohort study. Demographic characteristics of the patients, laboratory parameters on admission, left ventricular ejection fraction (LVEF) percentages, affected vessels in angiography (CAG) and the treatment strategy [medical therapy, percutaneous transluminal coronary angioplasty (PTCA), coronary angio by-pass graft] and SX scores were recorded on the data collection form. ROC curve was used to investigate the predictivity of blood urea nitrogen/albumin ratio (BAR), procalcitonin, C-reactive protein (CRP), high sensitivity cardiac troponin I (Hs-cTnI), CRP to serum albumin ratio (CAR), neutrophil to lymphocyte ratio (NLR) and SX scores in mortality. Multivariate analysis of biomarkers and SX score was performed to estimate the patients' 30-day mortality. Of the 415 patients were included in the study. ROC analysis of BAR, CAR, CRP, Procalcitonin, Hs-cTnI, NLR and SX score to predict mortality was statistically significant. BAR (OR: 1.280, 95% CI: 1.113-1.472, p = .001) and SX score (OR: 1.071, 95% CI: 1.018-1.126, p = .007) were found to be independent predictors of 30 days mortality. LVEF reduction, SX score, the number of affected vessels and the frequency of LMCA lesions increase were found to be statistically significant in patients with BAR ≥4.8. BAR, which can be calculated easily and quickly on admission to the emergency department and in clinical practice, may be used to predict mortality in patients with NSTEMI.
Collapse
Affiliation(s)
- Sarper Sevdımbas
- Health Sciences University, Adana City Training and Research Hospital, Emergency Medicine Clinic, Adana, Turkey
| | - Salim Satar
- Health Sciences University, Adana City Training and Research Hospital, Emergency Medicine Clinic, Adana, Turkey
| | - Muge Gulen
- Health Sciences University, Adana City Training and Research Hospital, Emergency Medicine Clinic, Adana, Turkey
| | - Selen Acehan
- Health Sciences University, Adana City Training and Research Hospital, Emergency Medicine Clinic, Adana, Turkey
| | - Armagan Acele
- Health Sciences University, Adana City Training and Research Hospital, Department of Cardiology, Adana, Turkey
| | - Gonca Koksaldı Sahin
- Health Sciences University, Adana City Training and Research Hospital, Emergency Medicine Clinic, Adana, Turkey
| | - Deniz Aka Satar
- Health Sciences University, Adana City Training and Research Hospital, Assisted Reproduction Unit, Andrology Laboratory, Adana, Turkey
| |
Collapse
|
24
|
Luo C, Chen F, Liu L, Ge Z, Feng C, Chen Y. Impact of diabetes on outcomes of cardiogenic shock: A systematic review and meta-analysis. Diab Vasc Dis Res 2022; 19:14791641221132242. [PMID: 36250870 PMCID: PMC9580099 DOI: 10.1177/14791641221132242] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
To provide synthesized evidence on the association of diabetes with clinical outcomes of patients with acute myocardial infarction (AMI) and associated cardiogenic shock (CS). We analyzed observational studies on patients with AMI and CS, identified through a systematic search using PubMed and Scopus databases. The main outcome was mortality and other outcomes of interest were risk of major bleeding, re-infarction, cerebrovascular adverse events, and need for revascularization. We conducted the meta-analysis with data from 15 studies. Compared to patients without diabetes, those with diabetes had an increased risk of in-hospital mortality (OR, 1.34; 95% CI, 1.17-1.54) and cerebrovascular complications (OR, 1.28; 95% CI, 1.11-1.48). We found similar risk of major bleeding (OR, 0.68; 95% CI, 0.43-1.09), re-infarction (OR, 0.98; 95% CI, 0.48-1.98) and need for re-vascularization (OR, 0.96; 95% CI, 0.75-1.22) as well as hospital stay lengths (in days) (WMD 0.00; 95% CI, -0.27-0.28; n = 4; I2 = 99.7%) in the two groups of patients. Patients with diabetes, acute MI and associated cardiogenic shock have increased risks of mortality and adverse cerebrovascular events than those without diabetes.
Collapse
Affiliation(s)
- Chao Luo
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, China
| | - Feng Chen
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, China
| | - Lingpei Liu
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, China
| | - Zuanmin Ge
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, China
| | - Chengzhen Feng
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, China
| | - Yuehua Chen
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, China
- Yuehua Chen, Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, 365 Renming East Road, Jinhua, Zhejiang 321000, China.
| |
Collapse
|
25
|
Krittanawong C, Rivera MR, Shaikh P, Kumar A, May A, Mahtta D, Jentzer J, Civitello A, Katz J, Naidu SS, Cohen MG, Menon V. SKey Concepts Surrounding Cardiogenic Shock. Curr Probl Cardiol 2022; 47:101303. [PMID: 35787427 DOI: 10.1016/j.cpcardiol.2022.101303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 11/03/2022]
Abstract
Cardiogenic shock (CS) is the final common pathway of impaired cardiovascular performance that results in ineffective forward cardiac output producing clinical and biochemical signs of organ hypoperfusion. CS represents the most common cause of shock in the cardiac intensive care unit (CICU) and accounts for a substantial proportion of CICU patient deaths. Despite significant advances in revascularization techniques, pharmacologic therapeutics and mechanical support devices, CS remains associated with a high mortality rate. Indeed, the prevalence of CS within the CICU appears to be increasing. CS can be differentiated as phenotypes reflecting different metabolic, inflammatory, and hemodynamic profiles, depending also on anatomic substrate and congestion profile. Future prospective studies and clinical trials may further characterize these phenotypes and apply targeted intervention for each phenotype and SCAI SHOCK stage rather than a one-size-fits-all approach. Overall, there are 8 key concepts of CS; 1) the mortality associated with CS; 2) Shock attributed to AMI may be declining in both incidence and associated mortality; 3) providers should think about hemodynamic, metabolic, inflammation and cardiac function in totality to assess CS; 4) CS is a dynamic process; 5) no randomized trials evaluating use of the PAC in patients with CS; 6) most data supporting neosynephrine as first line agent in CS; 7) most registries suggest that almost half of CS patients do not have any mechanical support, and the vast majority of the remainder utilize the IABP; and 8) patients with AMI CS should receive emergent PCI of the culprit vessel.
Collapse
Affiliation(s)
- Chayakrit Krittanawong
- Section of Cardiology, Baylor College of Medicine, Baylor St. Luke's Medical Center, Texas Heart Institute, Houston, TX.
| | - Mario Rodriguez Rivera
- John T. Milliken Department of Medicine, Division of Cardiovascular Disease. Barnes-Jewish Hospital/Washington University in St.Louis School of Medicine
| | - Preet Shaikh
- John T. Milliken Department of Medicine, Barnes-Jewish Hospital/Washington University in St.Louis School of Medicine
| | - Anirudh Kumar
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH
| | - Adam May
- John T. Milliken Department of Medicine, Division of Cardiovascular Disease, Section of Critical Care Cardiology. Barnes-Jewish Hospital/Washington University in St.Louis School of Medicine
| | - Dhruv Mahtta
- Section of Cardiology, Baylor College of Medicine, Baylor St. Luke's Medical Center, Texas Heart Institute, Houston, TX
| | - Jacob Jentzer
- Department of Cardiovascular Medicine; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Andrew Civitello
- Section of Cardiology, Baylor College of Medicine, Baylor St. Luke's Medical Center, Texas Heart Institute, Houston, TX
| | - Jason Katz
- Department of Medicine, Division of Cardiology, Duke University, Durham, NC
| | - Srihari S Naidu
- Department of Cardiology, Westchester Medical Centre, New York Medical College, Valhalla, NY
| | - Mauricio G Cohen
- Cardiovascular Division, University of Miami Miller School of Medicine, FL, USA
| | - Venu Menon
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
26
|
TandemHeart-Associated Fever. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2022. [DOI: 10.1097/ipc.0000000000001186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Krychtiuk KA, Vrints C, Wojta J, Huber K, Speidl WS. Basic mechanisms in cardiogenic shock: part 1-definition and pathophysiology. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2022; 11:356-365. [PMID: 35218350 DOI: 10.1093/ehjacc/zuac021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/17/2022] [Accepted: 02/07/2022] [Indexed: 05/23/2023]
Abstract
Cardiogenic shock mortality rates remain high despite significant advances in cardiovascular medicine and the widespread uptake of mechanical circulatory support systems. Except for early invasive angiography and percutaneous coronary intervention of the infarct-related artery, the most widely used therapeutic measures are based on low-quality evidence. The grim prognosis and lack of high-quality data warrant further action. Part 1 of this two-part educational review defines cardiogenic shock and discusses current treatment strategies. In addition, we summarize current knowledge on basic mechanisms in the pathophysiology of cardiogenic shock, focusing on inflammation and microvascular disturbances, which may ultimately be translated into diagnostic or therapeutic approaches to improve the outcome of our patients.
Collapse
Affiliation(s)
- Konstantin A Krychtiuk
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Duke Clinical Research Institute, Durham, NC, USA
| | - Christiaan Vrints
- Research Group Cardiovascular Diseases, Department GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Johann Wojta
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Kurt Huber
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- 3rd Department of Internal Medicine, Cardiology and Intensive Care Unit, Wilhelminenhospital, Vienna, Austria
- Medical School, Sigmund Freud University, Vienna, Austria
| | - Walter S Speidl
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| |
Collapse
|
28
|
Muzafarova T, Motovska Z. Laboratory Predictors of Prognosis in Cardiogenic Shock Complicating Acute Myocardial Infarction. Biomedicines 2022; 10:1328. [PMID: 35740350 PMCID: PMC9220203 DOI: 10.3390/biomedicines10061328] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 05/29/2022] [Accepted: 06/01/2022] [Indexed: 12/02/2022] Open
Abstract
Cardiogenic shock is a state of reduced cardiac output leading to hypotension, pulmonary congestion, and hypoperfusion of tissues and vital organs. Despite the advances in intensive care over the last years, the morbidity and mortality of patients remain high. The available studies of patients with cardiogenic shock suggest a connection between clinical variables, the level of biomarkers, the results of imaging investigations, strategies of management and the outcome of this group of patients. The management of patients with cardiogenic shock initially complicating acute myocardial infarction is challenging, and the number of studies in this area is growing fast. The purpose of this review is to summarize the currently available evidence on cardiogenic shock initially complicating acute myocardial infarction with particular attention to predictors of prognosis, focusing on laboratory variables (established and new), and to discuss the practical implementation. Currently available scoring systems developed during the past few decades predict the clinical outcome of this group of patients using some of the established biomarkers among other variables. With the new laboratory biomarkers that have shown their predictive value in cardiogenic shock outcomes, a new design of scoring systems would be of interest. Identifying high-risk patients offers the opportunity for early decision-making.
Collapse
Affiliation(s)
| | - Zuzana Motovska
- Cardiocenter, Third Faculty of Medicine Charles University and University Hospital Kralovske Vinohrady, 10034 Prague, Czech Republic;
| |
Collapse
|
29
|
Mitsis A, Kadoglou NPE, Lambadiari V, Alexiou S, Theodoropoulos KC, Avraamides P, Kassimis G. Prognostic role of inflammatory cytokines and novel adipokines in acute myocardial infarction: An updated and comprehensive review. Cytokine 2022; 153:155848. [PMID: 35301174 DOI: 10.1016/j.cyto.2022.155848] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 12/19/2022]
Abstract
Acute myocardial infarction (AMI) is one of the major causes of morbidity and mortality worldwide. The inflammation response during and after AMI is common and seems to play a key role in the peri-AMI period, related with ischaemia-reperfusion injury, adverse cardiac remodelling, infarct size and poor prognosis. In this article, we provide an updated and comprehensive overview of the most important cytokines and adipokines involved in the complex pathophysiology mechanisms in AMI, summarizing their prognostic role post-AMI. Data so far support that elevated levels of the major proinflammatory cytokines TNFα, IL-6 and IL-1 and the adipokines adiponectin, visfatin and resistin, are linked to high mortality and morbidity. In contrary, there is evidence that anti-inflammatory cytokines and adipokines as IL-10, omentin-1 and ghrelin can suppress the AMI-induced inflammatory response and are correlated with better prognosis. Mixed data make unclear the role of the novel adipokines leptin and apelin. After all, imbalance of pro-inflammatory and anti-inflammatory cytokines may result in worst AMI prognosis. The incorporation of these inflammation biomarkers in established prognostic models could further improve their prognostic power improving overall the management of AMI patients.
Collapse
Affiliation(s)
- Andreas Mitsis
- Cardiology Department, Nicosia General Hospital, Cyprus.
| | | | - Vaia Lambadiari
- Second Department of Internal Medicine, Research Institute and Diabetes Centre, Athens University Medical School, Attikon University General Hospital, Athens, Greece
| | - Sophia Alexiou
- Second Cardiology Department, "Hippokration" Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - George Kassimis
- Second Cardiology Department, "Hippokration" Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
30
|
Orrego R, Gaete B. Soporte extracorpóreo en shock cardiogénico con ECMO veno-arterial. REVISTA MÉDICA CLÍNICA LAS CONDES 2022. [DOI: 10.1016/j.rmclc.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
31
|
Attinger-Toller A, Bossard M, Cioffi GM, Tersalvi G, Madanchi M, Bloch A, Kobza R, Cuculi F. Ventricular Unloading Using the Impella TM Device in Cardiogenic Shock. Front Cardiovasc Med 2022; 9:856870. [PMID: 35402561 PMCID: PMC8984099 DOI: 10.3389/fcvm.2022.856870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/21/2022] [Indexed: 12/02/2022] Open
Abstract
Cardiogenic shock (CS) remains a leading cause of hospital death. However, the use of mechanical circulatory support has fundamentally changed CS management over the last decade and is rapidly increasing. In contrast to extracorporeal membrane oxygenation as well as counterpulsation with an intraaortic balloon pump, ventricular unloading by the Impella™ device actively reduces ventricular volume as well as pressure and augments systemic blood flow at the same time. By improving myocardial oxygen supply and enhancing systemic circulation, the Impella device potentially protects myocardium, facilitates ventricular recovery and may interrupt the shock spiral. So far, the evidence supporting the use of Impella™ in CS patients derives mostly from observational studies, and there is a need for adequate randomized trials. However, the Impella™ device appears a promising technology for management of CS patients. But a profound understanding of the device, its physiologic impact and clinical application are all important when evaluating CS patients for percutaneous circulatory support. This review provides a comprehensive overview of the percutaneous assist device Impella™. Moreover, it highlights in depth the rationale for ventricular unloading in CS and describes practical aspects to optimize care for patients requiring hemodynamic support.
Collapse
Affiliation(s)
- Adrian Attinger-Toller
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Lucerne, Switzerland,*Correspondence: Adrian Attinger-Toller
| | - Matthias Bossard
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Lucerne, Switzerland
| | | | - Gregorio Tersalvi
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Mehdi Madanchi
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Andreas Bloch
- Department of Intensive Care Medicine, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Richard Kobza
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Florim Cuculi
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Lucerne, Switzerland
| |
Collapse
|
32
|
Beer BN, Jentzer JC, Weimann J, Dabboura S, Yan I, Sundermeyer J, Kirchhof P, Blankenberg S, Schrage B, Westermann D. Early risk stratification in patients with cardiogenic shock irrespective of the underlying cause - The Cardiogenic Shock Score (CSS). Eur J Heart Fail 2022; 24:657-667. [PMID: 35119176 DOI: 10.1002/ejhf.2449] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/25/2022] [Accepted: 01/31/2022] [Indexed: 11/10/2022] Open
Abstract
AIMS Early risk stratification is essential to guide treatment in cardiogenic shock (CS). Existing CS risk scores were derived in selected cohorts, without accounting for the heterogeneity of CS. The aim of this study was to develop a universal risk score (CSS) for all CS patients, irrespective of underlying cause. METHODS AND RESULTS Within a registry of 1,308 CS unselected patients admitted to a tertiary-care hospital between 2009 and 2019, a Cox regression model was fitted to derive the CSS, with 30-day mortality as main outcome. CSS's predictive ability was compared to the IABP-Shock-II score, the CardShock score and SCAI classification by C-indices and validated in an external cohort of 934 CS patients. Based on the Cox regression, 9 predictors were included in the CSS: age, sex, acute myocardial infarction (AMI-CS), systolic blood pressure, heart rate, pH, lactate, glucose and cardiac arrest. CSS had the highest C-index in the overall cohort (0.740 vs. 0.677/0.683 for IABP-Shock-II score/CardShock score), in patients with AMI-CS (0.738 vs. 0.675/0.689 for IABP-Shock-II score/CardShock score) and in patients with non-AMI-CS (0.734 vs. 0.677/0.669 for IABP-Shock-II score/CardShock score). In the external validation cohort, the CSS had a C-index of 0.73, which was higher than all other tested scores. CONCLUSION The CSS provides improved information on the risk of death in unselected patients with CS compared to existing scores, irrespective of its cause. Because it is based on point-of-care variables which can be obtained even in critical situations, the CSS has the potential to guide treatment decisions in CS. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Benedikt N Beer
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Germany
| | - Jacob C Jentzer
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jessica Weimann
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany
| | - Salim Dabboura
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Germany
| | - Isabell Yan
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany
| | - Jonas Sundermeyer
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany
| | - Paulus Kirchhof
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Germany.,Institute of Cardiovascular Sciences, University of Birmingham, UK
| | - Stefan Blankenberg
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Germany
| | - Benedikt Schrage
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Germany
| | - Dirk Westermann
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Germany
| |
Collapse
|
33
|
Levy B, Girerd N, Amour J, Besnier E, Nesseler N, Helms J, Delmas C, Sonneville R, Guidon C, Rozec B, David H, Bougon D, Chaouch O, Walid O, Hervé D, Belin N, Gaide-Chevronnay L, Rossignol P, Kimmoun A, Duarte K, Slutsky AS, Brodie D, Fellahi JL, Ouattara A, Combes A. Effect of Moderate Hypothermia vs Normothermia on 30-Day Mortality in Patients With Cardiogenic Shock Receiving Venoarterial Extracorporeal Membrane Oxygenation: A Randomized Clinical Trial. JAMA 2022; 327:442-453. [PMID: 35103766 PMCID: PMC8808325 DOI: 10.1001/jama.2021.24776] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022]
Abstract
IMPORTANCE The optimal approach to the use of venoarterial extracorporeal membrane oxygenation (ECMO) during cardiogenic shock is uncertain. OBJECTIVE To determine whether early use of moderate hypothermia (33-34 °C) compared with strict normothermia (36-37 °C) improves mortality in patients with cardiogenic shock receiving venoarterial ECMO. DESIGN, SETTING, AND PARTICIPANTS Randomized clinical trial of patients (who were eligible if they had been endotracheally intubated and were receiving venoarterial ECMO for cardiogenic shock for <6 hours) conducted in the intensive care units at 20 French cardiac shock care centers between October 2016 and July 2019. Of 786 eligible patients, 374 were randomized. Final follow-up occurred in November 2019. INTERVENTIONS Early moderate hypothermia (33-34 °C; n = 168) for 24 hours or strict normothermia (36-37 °C; n = 166). MAIN OUTCOMES AND MEASURES The primary outcome was mortality at 30 days. There were 31 secondary outcomes including mortality at days 7, 60, and 180; a composite outcome of death, heart transplant, escalation to left ventricular assist device implantation, or stroke at days 30, 60, and 180; and days without requiring a ventilator or kidney replacement therapy at days 30, 60, and 180. Adverse events included rates of severe bleeding, sepsis, and number of units of packed red blood cells transfused during venoarterial ECMO. RESULTS Among the 374 patients who were randomized, 334 completed the trial (mean age, 58 [SD, 12] years; 24% women) and were included in the primary analysis. At 30 days, 71 patients (42%) in the moderate hypothermia group had died vs 84 patients (51%) in the normothermia group (adjusted odds ratio, 0.71 [95% CI, 0.45 to 1.13], P = .15; risk difference, -8.3% [95% CI, -16.3% to -0.3%]). For the composite outcome of death, heart transplant, escalation to left ventricular assist device implantation, or stroke at day 30, the adjusted odds ratio was 0.61 (95% CI, 0.39 to 0.96; P = .03) for the moderate hypothermia group compared with the normothermia group and the risk difference was -11.5% (95% CI, -23.2% to 0.2%). Of the 31 secondary outcomes, 30 were inconclusive. The incidence of moderate or severe bleeding was 41% in the moderate hypothermia group vs 42% in the normothermia group. The incidence of infections was 52% in both groups. The incidence of bacteremia was 20% in the moderate hypothermia group vs 30% in the normothermia group. CONCLUSIONS AND RELEVANCE In this randomized clinical trial involving patients with refractory cardiogenic shock treated with venoarterial ECMO, early application of moderate hypothermia for 24 hours did not significantly increase survival compared with normothermia. However, because the 95% CI was wide and included a potentially important effect size, these findings should be considered inconclusive. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02754193.
Collapse
Affiliation(s)
- Bruno Levy
- Médecine Intensive et Réanimation, CHRU Nancy, Pôle Cardio-Médico-Chirurgical, Vandoeuvre-les-Nancy, France
- INSERM U1116, Faculté de Médecine, Vandoeuvre-les-Nancy, France
- Université de Lorraine, Nancy, France
| | - Nicolas Girerd
- Université de Lorraine, INSERM, Centre d’Investigations Cliniques Plurithématique, INSERM 1433, CHRU de Nancy, Institut Lorrain du Coeur et des Vaisseaux, Nancy, Frances
- INI-CRCT (Cardiovascular and Renal Clinical Trialists) F-CRIN Network, Nancy, France
| | - Julien Amour
- Institut de Perfusion, de Réanimation et d’Anesthésie de Chirurgie Cardiaque Paris Sud, Hôpital Privé Jacques Cartier, Massy, France
| | - Emmanuel Besnier
- Department of Anaesthesiology and Critical Care, Rouen University Hospital, Rouen, France
- Normandie University, UNIROUEN, INSERM U1096, EnVi, Rouen, France
| | - Nicolas Nesseler
- Department of Anesthesia and Critical Care, Pontchaillou, University Hospital of Rennes, Rennes, France
- University Rennes, CHU de Rennes, Inra, INSERM, Institut NUMECAN – UMR_A 1341, UMR_S 1241, CIC 1414 (Centre d’Investigation Clinique de Rennes), Rennes, France
| | - Julie Helms
- Université de Strasbourg, Faculté de Médecine, Hôpitaux Universitaires de Strasbourg, Service de Médecine Intensive-Réanimation, Nouvel Hôpital Civil, Strasbourg, France
| | - Clément Delmas
- Intensive Cardiac Care Unit, Rangueil University Hospital, Toulouse, France
| | - Romain Sonneville
- AP-HP, Bichat Hospital, Medical and infectious diseases ICU, Paris, France
| | | | - Bertrand Rozec
- Service d’Anesthésie-Réanimation, Hôpital G&R Laennec CHU de Nantes, Nantes, France
- L’institut du Thorax INSERM, CNRS, CHU Nantes, UNIV Nantes, Nantes, France
| | - Helène David
- Department of Anesthesiology and Critical Care Medicine, Arnaud de Villeneuve Hospital, CHU Montpellier, Montpellier, France
- Montpellier University, INSERM, CNRS, PhyMedExp, Montpellier, France
| | - David Bougon
- Service de Réanimation, Centre Hospitalier Annecy, Genevois, France
| | - Oussama Chaouch
- Hôpital Européen Georges Pompidou, AP-HP, Department of Anesthesiology and Critical Care Medicine, Université Paris Descartes, Paris, France
| | - Oulehri Walid
- Service d’Anesthésie-Réanimation et Médecine péri-Opératoire, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Dupont Hervé
- Réanimation Médico-Chirurgicale Cardio-Thoracique, Vasculaire et Respiratoire, CHU Amiens Picardie, Amiens, France
| | - Nicolas Belin
- Service de Réanimation Médicale, CHU Besançon, Besançon, France
| | - Lucie Gaide-Chevronnay
- Unité de Réanimation Cardiovasculaire et Thoracique, Pôle Anesthésie Réanimation, CHU de Grenoble Alpes, Grenoble, France
| | | | - Antoine Kimmoun
- Médecine Intensive et Réanimation, CHRU Nancy, Pôle Cardio-Médico-Chirurgical, Vandoeuvre-les-Nancy, France
| | - Kevin Duarte
- Université de Lorraine, Centre d’Investigations Cliniques Plurithématique, INSERM 1433, CHRU de Nancy, Institut Lorrain du Coeur et des Vaisseaux, Nancy, France
| | - Arthur S. Slutsky
- Keenan Research Center, Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Medicine, Surgery, and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Brodie
- Department of Medicine, College of Physicians and Surgeons, Columbia University, and the Center for Acute Respiratory Failure, New York-Presbyterian Hospital/Columbia University Medical Center, New York, New York
| | - Jean-Luc Fellahi
- Service d’Anesthésie-Réanimation, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
- Laboratoire CarMeN, INSERM 1060, Université Lyon 1 Claude Bernard, Lyon, France
| | - Alexandre Ouattara
- CHU Bordeaux, Department of Anaesthesia and Critical Care, Magellan Medico-Surgical Centre, Bordeaux, France
- University Bordeaux, INSERM, UMR 1034, Biology of Cardiovascular Diseases, Pessac, France
| | - Alain Combes
- Sorbonne Université, INSERM, UMRS_1166-ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
- Service de Médecine Intensive-Réanimation, Institut de Cardiologie, APHP Sorbonne Université Hôpital Pitié–Salpêtrière, Paris, France
| |
Collapse
|
34
|
Lassus J, Tarvasmäki T, Tolppanen H. Biomarkers in cardiogenic shock. Adv Clin Chem 2022; 109:31-73. [DOI: 10.1016/bs.acc.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
35
|
Complicating Acute Myocardial Infarction. Current Status and Unresolved Targets for Subsequent Research. J Clin Med 2021; 10:jcm10245904. [PMID: 34945202 PMCID: PMC8705405 DOI: 10.3390/jcm10245904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022] Open
Abstract
Mechanical reperfusion with primary angioplasty, as the treatment of choice in acute myocardial infarction (MI), is associated not only with a high percentage of full epicardial and tissue reperfusion but also with a very good immediate and long-term clinical outcome. However, the Achilles heel of MI treatment is its ensemble of complications, such as cardiogenic shock due to severe systolic and/or diastolic dysfunction or MI mechanical complications, including perforation of the left ventricular free wall, papillary muscle rupture with acute mitral regurgitation and ventricular septal rupture. They are associated with an increased or, sometimes, with an extremely high mortality rate, determining the overall mortality in an MI patient population. In this review we summarize the mechanisms of MI complications, current therapeutic management and alternative directions for overcoming their devastating consequences. Moreover, we have sought to indicate gaps in the evidence on current treatments as the potential targets for further clinical research. From the perspective of mortality trends that are not improving, the forthcoming therapeutic management of complicated MI will require an individualized and novel approach based on their thorough pathobiology.
Collapse
|
36
|
Reina-Couto M, Pereira-Terra P, Quelhas-Santos J, Silva-Pereira C, Albino-Teixeira A, Sousa T. Inflammation in Human Heart Failure: Major Mediators and Therapeutic Targets. Front Physiol 2021; 12:746494. [PMID: 34707513 PMCID: PMC8543018 DOI: 10.3389/fphys.2021.746494] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/20/2021] [Indexed: 12/28/2022] Open
Abstract
Inflammation has been recognized as a major pathophysiological contributor to the entire spectrum of human heart failure (HF), including HF with reduced ejection fraction, HF with preserved ejection fraction, acute HF and cardiogenic shock. Nevertheless, the results of several trials attempting anti-inflammatory strategies in HF patients have not been consistent or motivating and the clinical implementation of anti-inflammatory treatments for HF still requires larger and longer trials, as well as novel and/or more specific drugs. The present work reviews the different inflammatory mechanisms contributing to each type of HF, the major inflammatory mediators involved, namely tumor necrosis factor alpha, the interleukins 1, 6, 8, 10, 18, and 33, C-reactive protein and the enzymes myeloperoxidase and inducible nitric oxide synthase, and their effects on heart function. Furthermore, several trials targeting these mediators or involving other anti-inflammatory treatments in human HF are also described and analyzed. Future therapeutic advances will likely involve tailored anti-inflammatory treatments according to the patient's inflammatory profile, as well as the development of resolution pharmacology aimed at stimulating resolution of inflammation pathways in HF.
Collapse
Affiliation(s)
- Marta Reina-Couto
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
- Departamento de Medicina Intensiva, Centro Hospitalar e Universitário São João, Porto, Portugal
| | - Patrícia Pereira-Terra
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Janete Quelhas-Santos
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Carolina Silva-Pereira
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
| | - António Albino-Teixeira
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
| | - Teresa Sousa
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
| |
Collapse
|
37
|
Sasmita BR, Zhu Y, Gan H, Hu X, Xue Y, Xiang Z, Huang B, Luo S. Prognostic value of neutrophil-lymphocyte ratio in cardiogenic shock complicating acute myocardial infarction: A cohort study. Int J Clin Pract 2021; 75:e14655. [PMID: 34320267 DOI: 10.1111/ijcp.14655] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/26/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUNDS Cardiogenic shock (CS) is the most severe complication after acute myocardial infarction (AMI) with mortality above 50%. Inflammatory response is involved in the pathology of CS and AMI. In this study, we aimed to evaluate the prognostic value of admission neutrophil-lymphocyte ratio (NLR) in patients with CS complicating AMI. METHODS Two hundred and seventeen consecutive patients with CS after AMI were divided into two groups according to the admission NLR cut-off value ≤7.3 and >7.3. The primary outcome was 30-day all-cause mortality and the secondary end-point was the composite events of major adverse cardiovascular events (MACE), including all-cause mortality, ventricular tachycardia/ventricular fibrillation, atrioventricular block, gastrointestinal haemorrhage and non-fatal stroke. Cox proportional hazard models were performed to analyse the association of NLR with the outcome. NLR cut-off value was determined by Youden index. RESULTS Patients with NLR > 7.3 were older and presented with lower lymphocyte count, higher admission heart rate, B-type natriuretic peptide, leucocyte, neutrophil and creatinine (all P < .05). During a period of 30-day follow-up after admission, mortality in patients with NLR > 7.3 was significantly higher than in patients with NLR ≤ 7.3 (73.7% vs. 26.3%, P < .001). The incidence of MACE was also remarkably higher in patients with NLR > 7.3 (87.9% vs. 53.4%, P < .001). After multivariable adjustment, NLR > 7.3 remained an independent predictor for higher risk of 30-day mortality (HR 2.806; 95%CI 1.784, 4.415, P < .001) and MACE (HR 2.545; 95%CI 1.791, 3.617, P < .001). CONCLUSIONS Admission NLR could be used as an important tool for short-term prognostic evaluation in patients with CS complicating AMI and higher NLR is an independent predictor for increased 30-day all-cause mortality and MACE.
Collapse
Affiliation(s)
- Bryan Richard Sasmita
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuansong Zhu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongbo Gan
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiankang Hu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuzhou Xue
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenxian Xiang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bi Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
38
|
Zhang Z, Hu Q, Hu T. Association of Lymphocyte to Monocyte Ratio and Risk of in-Hospital Mortality in Patients with Cardiogenic Shock: A Propensity Score Matching Study. Int J Gen Med 2021; 14:4459-4468. [PMID: 34408483 PMCID: PMC8367081 DOI: 10.2147/ijgm.s325907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/21/2021] [Indexed: 11/23/2022] Open
Abstract
Background Lymphocyte to monocyte ratio (LMR) has been long implicated in the prediction of many inflammatory-related diseases. However, the possible value as prognostic marker of LMR have not been evaluated in cardiogenic shock (CS) patients. The aim of the study was to assess the relationship between LMR on admission and in-hospital mortality in CS patients. Methods Data on patients diagnosed with CS were extracted from the Medical Information Mart for Intensive Care-IV (MIMIC-IV) database. We performed a single-institution, retrospective study of 1487 CS patients and determined the optimal cut-off for LMR by X-tile software. Propensity score matching (PSM) and inverse probabilities of treatment weighting (IPTW) were conducted to control confounders. Cox proportional hazards model was performed to evaluate the relationship between LMR and in-hospital mortality. Kaplan-Meier curves and receiver operating characteristics (ROC) analysis were applied to assess the prognostic value of LMR. Results The optimal cut-off value for LMR was 0.9. Cox proportional hazards model demonstrated that lower LMR (< 0.9) was independently associated with in-hospital mortality with hazard ratio (HR) of 1.40 (1.12-1.74, P = 0.003). The results were consistent with survival analyses (P < 0.001, Log rank test). Adding LMR< 0.9 to the sequential organ failure assessment (SOFA) score improved discrimination and risk stratification for in-hospital mortality. Conclusion Lower level of LMR is related to higher risk of in-hospital mortality of patients with CS. As an easily available biomarker, LMR can independently predict the in-hospital mortality in CS patients.
Collapse
Affiliation(s)
- Zhengwei Zhang
- Department of Critical Care Medicine, Chengdu Second People's Hospital, Chengdu, People's Republic of China
| | - Qionghua Hu
- Department of Critical Care Medicine, Chengdu Second People's Hospital, Chengdu, People's Republic of China
| | - Tianyang Hu
- Department of Cardiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW We describe the pathophysiology of cardiogenic shock (CS), from the main pathways to the inflammatory mechanisms and the proteomic features. RECENT FINDINGS Although the classical pathophysiological pathways underlying CS, namely reduced organ perfusion due to inadequate cardiac output and peripheral vasoconstriction, have been well-established for a long time, the role of macro-and micro-hemodynamics in the magnitude of the disease and its prognosis has been investigated extensively only over the last few years. Moreover, to complete the complex picture of CS pathophysiology, the study of cytokine cascade, inflammation, and proteomic analysis has been addressed recently. SUMMARY Understanding the pathophysiology of CS is important to treat it optimally.
Collapse
|
40
|
Sun J, Zhang Q, Liu X, Shang X. Downregulation of IFIT3 relieves the inflammatory response and myocardial fibrosis of mice with myocardial infarction and improves their cardiac function. Exp Anim 2021; 70:522-531. [PMID: 34234081 PMCID: PMC8614010 DOI: 10.1538/expanim.21-0060] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Myocardial infarction (MI) is a common cardiovascular disease with high morbidity and mortality. In this study, we explored the role of interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) in MI. MI was induced by ligation of the left anterior descending coronary artery. Lentivirus-mediated RNA interference of IFIT3 expression was performed by tail vein injection 72 h before MI modeling. Cardiac injury indexes and inflammatory response were examined 3 days after MI. Cardiac function indexes, infarct size, and cardiac fibrosis were assessed 4 weeks after MI. IFIT3 expression was upregulated in myocardial tissues at both 3 days and 4 weeks after MI. Knockdown of IFIT3 significantly relieved the myocardial injury, as evidenced by the decrease in serum levels of cTnI and CK-MB. In addition, IFIT3 knockdown significantly reduced the number of CD68+ macrophages and the levels of interleukin-1β, interleukin-6, and tumor necrosis factor-α, indicating that the inflammatory response was relieved. Moreover, IFIT3 silencing also significantly improved cardiac function and reduced infarct size, myocardial fibrosis, and collagen content in mice with MI. Mechanically, the present study showed that the activation of the mitogen-activated protein kinase (MAPK) pathway was observed in myocardial tissues of MI mice, which was blocked by IFIT3 knockdown, as indicated by the decreased phosphorylation of JNK, p-38, and ERK. Collectively, our results revealed the role of IFIT3 in the inflammatory response and myocardial fibrosis after MI, indicating that IFIT3 might be a potential target for MI treatment.
Collapse
Affiliation(s)
- Jianhua Sun
- Department of Internal Medicine, Hebei Medical University.,Department of Cardiology, Tangshan Gongren Hospital Affiliated to Hebei Medical University
| | - Qi Zhang
- Department of Internal Medicine, Hebei Medical University
| | - Xiaokun Liu
- Department of Internal Medicine, Hebei Medical University
| | - Xiaoming Shang
- Department of Internal Medicine, Hebei Medical University
| |
Collapse
|
41
|
Jin J, Shi Z, Pang X. Association between low-density lipoprotein cholesterol level and mortality in patients with cardiogenic shock: a retrospective cohort study. BMJ Open 2021; 11:e044668. [PMID: 34215599 PMCID: PMC8256757 DOI: 10.1136/bmjopen-2020-044668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
AIMS Inflammation plays a key role in the pathophysiology of cardiogenic shock (CS). Low-density lipoprotein cholesterol (LDL-C) is a biomarker of inflammation and is used to predict prognostic outcomes of several diseases. The primary purpose of this study was to evaluate if LDL-C can be used as a biomarker to predict the mortality of CS. METHODS AND RESULTS Records of critically ill patients with CS were identified from the Medical Information Mart for Intensive Care III database. A multivariate Cox regression model was employed to adjust for imbalances by incorporating parameters and potential confounders.A total of 551 critically ill patients with CS were enrolled for this analysis, including 207 with LDL-C <1.8 mmol/L and 344 with LDL-C ≥1.8 mmol/L. Results of multivariate Cox regression models found that higher concentration of LDL-C (LDL-C ≥1.8mmol/L) was associated with a reduced risk of in-hospital mortality (HR 0.66, 95% CI 0.50 to 0.87; p=0.003) and 28-day mortality (HR 0.61, 95% CI 0.46 to 0.80; p=0.002) LDL-C in patients with CS. Patients with LDL-C ≥1.8 mmol/L were independently associated with improved in-hospital survival (HR 0.32, 95% CI 0.20 to 0.52, p<0.001) and 28-day survival (HR 0.51, 95% CI 0.33 to 0.73, p=0.002) compared with patients with LDL-C <1.8 mmol/L. The impact of LDL-C on in-hospital mortality and 28-day mortality persisted in patients with acute coronary syndrome (ACS) and was not statistically significant in the non-ACS subgroup. CONCLUSIONS Our study observed that increased LDL-C level was related with improved survival in patients with CS, but not with improved outcomes in patients with uncomplicated ACS. The results need to be verified in randomised controlled trials.
Collapse
Affiliation(s)
- Jiali Jin
- Department of Cardiology, Zhuji People's Hospital of Zhejiang Province, Zhuji, China
| | - Zhewei Shi
- Department of Cardiology, Zhuji People's Hospital of Zhejiang Province, Zhuji, China
| | - Xiaomin Pang
- Department of Emergency, People's Hospital of Tiantai County, Taizhou, Zhejiang, China
| |
Collapse
|
42
|
Tamara A, Coulson DJ, Latief JS, Bakhashab S, Weaver JU. Upregulated anti-angiogenic miR-424-5p in type 1 diabetes (model of subclinical cardiovascular disease) correlates with endothelial progenitor cells, CXCR1/2 and other parameters of vascular health. Stem Cell Res Ther 2021; 12:249. [PMID: 33985567 PMCID: PMC8120744 DOI: 10.1186/s13287-021-02332-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/07/2021] [Indexed: 01/14/2023] Open
Abstract
Background In spite of clinical progress, cardiovascular disease (CVD) remains the predominant cause of mortality worldwide. Overexpression studies in animals have proven miR-424-5p to have anti-angiogenic properties. As type 1 diabetes mellitus (T1DM) without CVD displays endothelial dysfunction and reduced circulating endothelial progenitor cells (cEPCs), it offers a model of subclinical CVD. Therefore, we explored miR-424-5p, cytokines and vascular health in T1DM. Methods Twenty-nine well-controlled T1DM patients with no CVD and 20-matched controls were studied. Cytokines IL8, TNF-α, IL7, VEGF-C, cEPCs/CD45dimCD34+CD133+ cells and ex-vivo proangiogenic cells (PACs)/fibronectin adhesion assay (FAA) were measured. MiR-424-5p in plasma and peripheral blood mononuclear cells (PBMC) along with mRNAs in PBMC was evaluated. Results We found an elevation of IL7 (p = 0.008), IL8 (p = 0.003), TNF-α (p = 0.041), VEGF-C (p = 0.013), upregulation of mRNA CXCR1 (p = 0.009), CXCR2 (p < 0.001) and reduction of cEPCs (p < 0.001), PACs (p < 0.001) and FAA (p = 0.017) in T1DM. MiR-424-5p was upregulated in T1DM in PBMC (p < 0.001). MiR-424-5p was negatively correlated with cEPCs (p = 0.006), PACs (p = 0.005) and FAA (p < 0.001) and positively with HbA1c (p < 0.001), IL7 (p = 0.008), IL8 (p = 0.017), VEGF-C (p = 0.007), CXCR1 (p = 0.02) and CXCR2 (p = 0.001). ROC curve analyses showed (1) miR-424-5p to be a biomarker for T1DM (p < 0.001) and (2) significant upregulation of miR-424-5p, defining subclinical CVD, occurred at HbA1c of 46.5 mmol/mol (p = 0.002). Conclusion We validated animal research on anti-angiogenic properties of miR-424-5p in T1DM. MiR-424-5p may be a biomarker for onset of subclinical CVD at HbA1c of 46.5 mmol/mol (pre-diabetes). Thus, miR-424-5p has potential use for CVD monitoring whilst anti-miR-424-5p-based therapies may be used to reduce CVD morbidity/mortality in T1DM.
Collapse
Affiliation(s)
- Alice Tamara
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Faculty of Medicine, Universitas Indonesia, Jakarta, 10430, Indonesia
| | - David J Coulson
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Jevi Septyani Latief
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Faculty of Medicine, Universitas Indonesia, Jakarta, 10430, Indonesia
| | - Sherin Bakhashab
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 80218, Saudi Arabia
| | - Jolanta U Weaver
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK. .,Department of Diabetes, Queen Elizabeth Hospital, Gateshead, Newcastle upon Tyne, NE9 6SH, UK. .,Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
43
|
Kimmoun A, Duarte K, Harjola VP, Tarvasmäki T, Levy B, Mebazaa A, Gibot S. Soluble triggering receptor expressed on myeloid cells-1 is a marker of organ injuries in cardiogenic shock: results from the CardShock Study. Clin Res Cardiol 2021; 111:604-613. [PMID: 33677708 DOI: 10.1007/s00392-021-01823-0] [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] [Received: 12/03/2020] [Accepted: 02/16/2021] [Indexed: 12/01/2022]
Abstract
AIMS Optimal outcome after cardiogenic shock (CS) depends on a coordinated healing response in which both debris removal and extracellular matrix tissue repair play a crucial role. Excessive inflammation can perpetuate a vicious circle, positioning leucocytes as central protagonists and potential therapeutic targets. High levels of circulating Triggering Receptor Expressed on Myeloid cells-1 (TREM-1), were associated with death in acute myocardial infarction confirming excessive inflammation as determinant of bad outcome. The present study aims to describe the association of soluble TREM-1 with 90-day mortality and with various organ injuries in patients with CS. METHODS AND RESULTS This is a post-hoc study of CardShock, a prospective, multicenter study assessing the clinical presentation and management in patients with CS. At the time of this study, 87 patients had available plasma samples at either baseline, and/or 48 h and/or 96-120 h for soluble TREM-1 (sTREM-1) measurements. Plasma concentration of sTREM-1 was higher in 90-day non-survivors than survivors at baseline [median: 1392 IQR: (724-2128) vs. 621 (525-1233) pg/mL, p = 0.008), 48 h (p = 0.019) and 96-120 h (p = 0.029). The highest tertile of sTREM-1 at baseline (threshold: 1347 pg/mL) was associated with 90-day mortality with an unadjusted HR 3.08 CI 95% (1.48-6.42). sTREM-1 at baseline was not associated to hemodynamic parameters (heart rate, blood pressure, use of vasopressors or inotropes) but rather with organ injury markers: renal (estimated glomerular filtration rate, p = 0.0002), endothelial (bio-adrenomedullin, p = 0.018), myocardial (Suppression of Tumourigenicity 2, p = 0.002) or hepatic (bilirubin, p = 0.008). CONCLUSION In CS patients TREM-1 pathway is highly activated and gives an early prediction of vital organ injuries and outcome.
Collapse
Affiliation(s)
- Antoine Kimmoun
- Université de Lorraine, CHRU de Nancy, Médecine Intensive et Réanimation Brabois, INSERM U942 and U1116, F-CRIN-INIC RCT, Vandœuvre-lès-Nancy, France
| | - Kevin Duarte
- Université de Lorraine, CHRU de Nancy, INSERM CIC-P 1433, INSERM, F-CRIN-INI CRCT, Vandœuvre-lès-Nancy, France
| | - Veli-Pekka Harjola
- Emergency Medicine, Department of Emergency Medicine and Services, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Tuukka Tarvasmäki
- Cardiology, Heart and Lung Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Bruno Levy
- Université de Lorraine, CHRU de Nancy, Médecine Intensive et Réanimation Brabois, INSERM U942 and U1116, F-CRIN-INIC RCT, Vandœuvre-lès-Nancy, France
| | - Alexandre Mebazaa
- Département d'Anesthésie et Réanimation, Université de Paris, AP-HP, CHU Lariboisière, INSERM U942, F-CRIN-INI CRCT, Paris, France
| | - Sebastien Gibot
- Université de Lorraine, CHRU de Nancy, Médecine Intensive et Réanimation Central, INSERM U1116, Nancy, France.
| | | |
Collapse
|
44
|
Shiraishi Y, Kawana M, Nakata J, Sato N, Fukuda K, Kohsaka S. Time-sensitive approach in the management of acute heart failure. ESC Heart Fail 2020; 8:204-221. [PMID: 33295126 PMCID: PMC7835610 DOI: 10.1002/ehf2.13139] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/23/2020] [Accepted: 11/11/2020] [Indexed: 12/25/2022] Open
Abstract
Acute heart failure (AHF) has become a global public health burden largely because of the associated high morbidity, mortality, and cost. The treatment options for AHF have remained relatively unchanged over the past decades. Historically, clinical congestion alone has been considered the main target for treatment of acute decompensation in patients with AHF; however, this is an oversimplification of the complex pathophysiology. Within the similar clinical presentation of congestion, significant differences in pathophysiological mechanisms exist between the fluid accumulation and redistribution. Tissue hypoperfusion is another vital characteristic of AHF and should be promptly treated with appropriate interventions. In addition, recent clinical trials of novel therapeutic strategies have shown that heart failure management is ‘time sensitive’ and suggested that treatment selection based on individual aetiologies, triggers, and risk factor profiles could lead to better outcomes. In this review, we aim to describe the specifics of the ‘time‐sensitive’ approach by the clinical phenotypes, for example, pulmonary/systemic congestion and tissue hypoperfusion, wherein patients are classified based on pathophysiological conditions. This mechanistic classification, in parallel with the comprehensive risk assessment, has become a cornerstone in the management of patients with AHF and thus supports effective decision making by clinicians. We will also highlight how therapeutic modalities should be individualized according to each clinical phenotype.
Collapse
Affiliation(s)
- Yasuyuki Shiraishi
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masataka Kawana
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Jun Nakata
- Division of Intensive and Cardiovascular Care Unit, Department of Cardiology, Nippon Medical School Hospital, Tokyo, Japan
| | - Naoki Sato
- Department of Cardiovascular Medicine, Kawaguchi Cardiovascular and Respiratory Hospital, Saitama, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shun Kohsaka
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
45
|
Kristono GA, Holley AS, Hally KE, Brunton-O'Sullivan MM, Shi B, Harding SA, Larsen PD. An IL-6-IL-8 score derived from principal component analysis is predictive of adverse outcome in acute myocardial infarction. Cytokine X 2020; 2:100037. [PMID: 33604561 PMCID: PMC7885891 DOI: 10.1016/j.cytox.2020.100037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/01/2020] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Many studies have shown that elevated biomarkers of inflammation following acute myocardial infarction (AMI) are associated with major adverse cardiovascular events (MACE). However, the optimal way of measuring the complex inflammatory response following AMI has not been determined. In this study we explore the use of principal component analysis (PCA) utilising multiple inflammatory cytokines to generate a combined cytokine score that may be predictive of MACE post-AMI. METHODS Thirteen inflammatory cytokines were measured in plasma of 317 AMI patients, drawn 48-72 h following symptom onset. Patients were followed-up for one year to determine the incidence of MACE. PCA was used to generate a combined score using six cytokines that were detectable in the majority of patients (IL-1β, -6, -8, and -10; MCP-1; and RANTES), and using a subset of cytokines that were associated with MACE on univariate analysis. Multivariate models using baseline characteristics, elevated individual cytokines and PCA-derived scores determined independent predictors of MACE. RESULTS IL-6 and IL-8 were significantly associated with MACE on univariate analysis and were combined using PCA into an IL-6-IL-8 score. The combined cytokine score and IL-6-IL-8 PCA-derived score were both significantly associated with MACE on univariate analysis. In multivariate models IL-6-IL-8 scores (OR = 2.77, p = 0.007) and IL-6 levels (OR = 2.18, p = 0.035) were found to be independent predictors of MACE. CONCLUSION An IL-6-IL-8 score derived from PCA was found to independently predict MACE at one year and was a stronger predictor than any individual cytokine, which suggests this may be an appropriate strategy to quantify inflammation post-AMI. Further investigation is required to determine the optimal set of cytokines to measure in this context.
Collapse
Key Words
- ACS, Acute coronary syndrome
- AF, Atrial fibrillation
- AMI, Acute myocardial infarction
- AUC, Area under the curve
- Acute myocardial infarction
- BMI, Body mass index
- CAD, Coronary artery disease
- CBA, Cytometric bead array
- CHF, Chronic heart failure
- CI, Confidence interval
- CVD, Cardiovascular disease
- Cytokine score
- EFA, Exploratory factor analysis
- ELISA, Enzyme-linked immunosorbent assay
- GDF-15, Growth differentiation factor-15
- GM-CSF, Granulocyte-macrophage colony-stimulating factor
- HTN, Hypertension
- IFNγ, Interferon gamma
- IL-(number), Interleukin-(number)
- IQR, Interquartile range
- Interleukin-6
- Interleukin-8
- MACE, Major adverse cardiovascular events
- MCP-1, Monocyte chemoattractant protein-1
- MFI, Mean fluorescence intensity
- MI, Myocardial infarction
- Major adverse cardiovascular events
- NSTEMI, Non-ST elevation myocardial infarction
- OR, Odds ratio
- PCA, Principal component analysis
- PCI, Percutaneous coronary intervention
- Principal component analysis
- RANTES, Regulated upon activation normal T-cell expressed and secreted
- ROC, Receiver operator characteristic
- STEMI, ST-elevation myocardial infarction
- TGF-β1, Tumour growth factor-beta 1
- TIA, Transient ischaemic attack
- TNF-α, Tumour necrosis factor alpha
- TRAIL-R2, Tumour necrosis factor-related apoptosis-inducing ligand receptor 2
- TnT, Troponin T
- VEGF, Vascular endothelial growth factor
- h, Hours
- p, P-value
Collapse
Affiliation(s)
- Gisela A. Kristono
- Department of Surgery and Anaesthesia, University of Otago Wellington, New Zealand
- Wellington Cardiovascular Research Group, New Zealand
| | - Ana S. Holley
- Department of Surgery and Anaesthesia, University of Otago Wellington, New Zealand
- Wellington Cardiovascular Research Group, New Zealand
| | - Kathryn E. Hally
- Department of Surgery and Anaesthesia, University of Otago Wellington, New Zealand
- Wellington Cardiovascular Research Group, New Zealand
- School of Biological Sciences, Victoria University of Wellington, New Zealand
| | - Morgane M. Brunton-O'Sullivan
- Department of Surgery and Anaesthesia, University of Otago Wellington, New Zealand
- Wellington Cardiovascular Research Group, New Zealand
| | - Bijia Shi
- Wellington Cardiovascular Research Group, New Zealand
- Cardiology Department, Capital and Coast District Health Board, New Zealand
| | - Scott A. Harding
- Wellington Cardiovascular Research Group, New Zealand
- Cardiology Department, Capital and Coast District Health Board, New Zealand
| | - Peter D. Larsen
- Department of Surgery and Anaesthesia, University of Otago Wellington, New Zealand
- Wellington Cardiovascular Research Group, New Zealand
| |
Collapse
|
46
|
Cormican DS, Sonny A, Crowley J, Sheu R, Sun T, Gibson CM, Núñez-Gil IJ, Ramakrishna H. Acute Myocardial Infarction Complicated by Cardiogenic Shock: Analysis of the Position Statement From the European Society of Cardiology Acute Cardiovascular Care Association, With Perioperative Implications. J Cardiothorac Vasc Anesth 2020; 35:3098-3104. [PMID: 33234469 DOI: 10.1053/j.jvca.2020.10.062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/28/2022]
Abstract
Effective management of cardiogenic shock (CS) is hampered by a lack of evidence-based information. This is a high-mortality condition, without clear, evidence-based guidelines for perioperative management, specifically-a lack of target endpoints for treatment (e.g.: mean arterial pressure or oxygenation), utility of regional care systems or the benefits of palliative care. The Acute Cardiovascular Care Association (ACCA) of the European Society of Cardiology (ESC) recently published a position statement that aimed to offer contemporary guidance on the diagnosis and treatment of acute myocardial infarction (AMI) complicated by CS. Herein, we review this complex clinical topic and review the ACCA statement on AMI associated with CS, with a focus on relevance to perioperative management.
Collapse
Affiliation(s)
- Daniel S Cormican
- Department of Anesthesiology, Divisions of Cardiothoracic Anesthesiology and Critical Care Medicine, Allegheny Health Network, Pittsburgh, PA
| | - Abraham Sonny
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA
| | - Jerome Crowley
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA
| | - Richard Sheu
- Department of Anesthesiology and Pain Medicine, University of Washington Medical Center, Seattle, WA
| | - Terri Sun
- Department of Anesthesiology and Pain Medicine, University of Washington Medical Center, Seattle, WA
| | | | - Iván J Núñez-Gil
- Interventional Cardiology. Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain
| | - Harish Ramakrishna
- Division of Cardiovascular and Thoracic Anesthesiology, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
47
|
Gaubert M, Laine M, Resseguier N, Aissaoui N, Puymirat E, Lemesle G, Michelet P, Hraiech S, Lévy B, Delmas C, Bonello L. Hemodynamic Profiles of Cardiogenic Shock Depending on Their Etiology. J Clin Med 2020; 9:jcm9113384. [PMID: 33105580 PMCID: PMC7690259 DOI: 10.3390/jcm9113384] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/28/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022] Open
Abstract
The pathophysiology of cardiogenic shock (CS) varies depending on its etiology, which may lead to different hemodynamic profiles (HP) and may help tailor therapy. We aimed to assess the HP of CS patients according to their etiologies of acute myocardial infarction (AMI) and acute decompensated chronic heart failure (ADCHF). We included patients admitted for CS secondary to ADCHF and AMI. HP were measured before the administration of any inotrope or vasopressor. Systemic Vascular Resistances index (SVRi), Cardiac Index (CI), and Cardiac Power Index (CPI) were measured by trans-thoracic Doppler echocardiography on admission. Among 37 CS patients, 28 had CS secondary to ADCHF or AMI and were prospectively included. The two groups were similar in terms of demographic data and shock severity criteria. AMI CS was associated with lower SVRi compared to CS related to ADCHF: 2010 (interquartile range (IQR): 1895-2277) vs. 2622 (2264-2993) dynes-s·cm-5·m-2 (p = 0.002). A trend toward a higher CI was observed: respectively 2.13 (1.88-2.18) vs. 1.78 (1.65-1.96) L·min-1·m-2 (p = 0.067) in AMICS compared to ADCHF. CS patients had different HP according to their etiologies. AMICS had lower SVR and tended to have a higher CI compared to ADHF CS. These differences should be taken into account for patient selection in future research.
Collapse
Affiliation(s)
- Mélanie Gaubert
- Cardiology Department, APHM, Mediterranean Association for Research and Studies in Cardiology (MARS Cardio), Centre for CardioVascular and Nutrition Research (C2VN), Aix-Marseille Univ, INSERM 1263, INRA 1260, Hopital Nord, 13015 Marseille, France; (M.G.); (M.L.)
| | - Marc Laine
- Cardiology Department, APHM, Mediterranean Association for Research and Studies in Cardiology (MARS Cardio), Centre for CardioVascular and Nutrition Research (C2VN), Aix-Marseille Univ, INSERM 1263, INRA 1260, Hopital Nord, 13015 Marseille, France; (M.G.); (M.L.)
| | - Noémie Resseguier
- Support Unit for Clinical Research and Economic Evaluation, APHM, 13385 Marseille, France;
| | - Nadia Aissaoui
- Department of Critical Care Unit, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou (HEGP), Université Paris-Descartes, 15015 Paris, France;
| | - Etienne Puymirat
- Département de Cardiologie, Hôpital Européen Georges Pompidou, Assistance Publique des Hôpitaux de Paris, 75015 Paris, France;
| | - Gilles Lemesle
- USIC et Centre Hémodynamique, Institut Coeur Poumon, Centre Hospitalier Régional et Universitaire de Lille, Faculté de Médecine de l’Université de Lille, Institut Pasteur de Lille, Unité INSERM UMR 1011, and FACT (French Alliance for Cardiovascular Trials), F-59000 Lille, France;
| | - Pierre Michelet
- Service d’accueil des Urgences, Hopital Timone, 13005 Marseille, France;
| | - Sami Hraiech
- Resuscitation Department, Aix-Marseille Univ, APHM, Hôpital Nord, 13005 Marseille, France;
| | - Bruno Lévy
- CHRU Nancy, Service de Réanimation Médicale Brabois, Pôle Cardiovasculaire et Réanimation Médicale, Hôpital Brabois, 54511 Vandoeuvre les Nancy, France;
| | - Clément Delmas
- INSERM UMR-1048, Intensive Cardiac Care Unit, Rangueil University Hospital, 31400 Toulouse, France;
| | - Laurent Bonello
- Cardiology Department, APHM, Mediterranean Association for Research and Studies in Cardiology (MARS Cardio), Centre for CardioVascular and Nutrition Research (C2VN), Aix-Marseille Univ, INSERM 1263, INRA 1260, Hopital Nord, 13015 Marseille, France; (M.G.); (M.L.)
- Correspondence:
| |
Collapse
|
48
|
Peng Y, Xue Y, Wang J, Xiang H, Ji K, Wang J, Lin C. Association between neutrophil-to-albumin ratio and mortality in patients with cardiogenic shock: a retrospective cohort study. BMJ Open 2020; 10:e039860. [PMID: 33077569 PMCID: PMC7574943 DOI: 10.1136/bmjopen-2020-039860] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES To investigate the prognostic value of neutrophil-to-albumin ratio (NAR) in critically ill patients with cardiogenic shock (CS). DESIGN A retrospective cohort study. SETTING A single centre in Boston, USA. PARTICIPANTS 475 patients with CS were included, among which 272 (57.3%) were men and 328 (69.1%) were white. PRIMARY AND SECONDARY OUTCOME MEASURES The primary outcome was 90-day mortality and the secondary outcomes were 30-day and 365-day mortality. RESULTS A significant positive correlation between NAR levels and 90-day, 30-day or 365-day mortality was observed. For 90-day mortality, the adjusted HR (95% CI) values given NAR levels 23.54-27.86 and >27.86 were 1.71 (1.14 to 2.55) and 1.93 (1.27 to 2.93) compared with the reference (NAR<23.47). Receiver operator characteristic curve analysis showed that NAR had a certain prognostic value in predicting 90-day mortality of CS, which was more sensitive than the neutrophil percentage or the serum albumin level alone (0.651 vs 0.509, 0.584). For the secondary outcomes, the upward trend remained statistically significant. CONCLUSIONS NAR level was associated with the mortality of CS patients. The prognostic value of NAR was more sensitive than the neutrophil percentage or the serum albumin level alone, but not as good as Sequential Organ Failure Assessment or Simplified Acute Physiology Score.
Collapse
Affiliation(s)
- Yangpei Peng
- Department of Nephrology, Wenzhou Medical University Second Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Yangjing Xue
- Department of Cardiology, Wenzhou Medical University Second Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Jinsheng Wang
- Department of Cardiology, Wenzhou Medical University Second Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Huaqiang Xiang
- Department of Cardiology, Wenzhou Medical University Second Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Kangting Ji
- Department of Cardiology, Wenzhou Medical University Second Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Jie Wang
- Department of Endocrinology, Yanbian University Hospital, Yanji, Jilin, China
| | - Cong Lin
- Department of Cardiology, Wenzhou Medical University Second Affiliated Hospital, Wenzhou, Zhejiang, China
| |
Collapse
|
49
|
Positive association between systemic immune-inflammatory index and mortality of cardiogenic shock. Clin Chim Acta 2020; 511:97-103. [PMID: 33045194 DOI: 10.1016/j.cca.2020.09.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Cardiogenic shock (CGS) is not only a state of hypoperfusion, but also related to inflammation. The prognostic value of systemic immune-inflammatory index (SII), an innovate biomarker of inflammation, in CGS patients has not been assessed. This study aims to explore the associations between SII and mortality in patients with CGS. METHODS Data on patients diagnosed with CGS were extracted from MIMIC-III database version 1.4. The follow-up started on the patients' first admission to ICU. The primary outcome was 30-day mortality. 90-day and 365-day mortality were the secondary outcomes. Cox proportional hazards models were used to investigate the associations between SII and mortality of CGS patients. RESULTS 707 patients with CGS were included in our study (59.8% male, 67.5% the white, 70.27 ± 14.56 years). For 30-day mortality, the HR (95% CI) value of high-SII group was 2.17 (1.60, 2.93) compared with the reference of low-SII group (P < 0.0001). The HR value of mid-SII group, however, showed none statistical significance (HR: 1.03, 95% CI: 0.74-1.43, P = 0.8516). When adjusted for age, gender and ethnicity in Model I, the adjusted HR (95% CI) value of high-SII group was 2.28 (1.69, 3.09). When further adjusted for heart rate, SBP, serum potassium, PTT, INR and ECI in Model II, the adjusted HR value of high-SII group was still statistically significant (HR: 2.08, 95% CI: 1.52-2.86, P < 0.0001). Similar results were also shown in the secondary outcomes of 90-day and 365-day mortality. CONCLUSIONS High level of SII is associated with increased short- and long-term mortality of patients with CGS. SII, a readily available biomarker, can independently predict the prognosis of CGS patients.
Collapse
|
50
|
Desai R, Hanna B, Singh S, Gupta S, Deshmukh A, Kumar G, Sachdeva R, Berman AE. Percutaneous Ventricular Assist Device vs. Intra-Aortic Balloon Pump for Hemodynamic Support in Acute Myocardial Infarction-Related Cardiogenic Shock and Coexistent Atrial Fibrillation: A Nationwide Propensity-Matched Analysis'. Am J Med Sci 2020; 361:55-62. [PMID: 33008567 DOI: 10.1016/j.amjms.2020.08.018] [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: 01/13/2020] [Revised: 08/04/2020] [Accepted: 08/10/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND Patients suffering an acute myocardial infarction complicated by cardiogenic shock (AMICS) may experience clinical deterioration with concomitant atrial fibrillation (AF). Recent data suggest that percutaneous ventricular assist devices (pVADs) provide superior hemodynamic support over intra-aortic balloon pump (IABP) in AMICS. In patients with AF+AMICS, however, outcomes data comparing these two devices remain limited. METHODS Using the National Inpatient Sample datasets (2008-2014) and a propensity-score matched analysis, we compared the outcomes of AMICS+AF hospitalized patients undergoing PCI with pVAD vs. IABP support. RESULTS A total of 12,842 AMICS+AF patients were identified (pVAD=468, IABP=12,374). The matched groups (pVAD=443, IABP=443) were comparable in terms of mean age (70.3 ± 12.0 vs. 70.4 ± 11.0yrs, p = 0.92). The utilization of pVAD was higher in whites but lower in Medicare/Medicaid beneficiaries as compared to IABP. The pVAD group demonstrated higher rates of obesity (13.6% vs. 7.8%, p = 0.006) and dyslipidemia (48.4% vs. 41.8%, p = 0.05). There was no difference in the in-hospital mortality (40.5% vs. 36.8%, p = 0.25); however, pVAD group had a lower incidence of post-procedural MI and higher incidences of stroke (7.8% vs. 4.4%, p = 0.03), hemorrhage (5.6% vs. 2.3%, p = 0.01), discharges to home health care (13.5% vs. 10.1%, p<0.001) and to other facilities (29.1% vs. 24.9%, p<0.001) as compared to IABP group. There was no difference between the groups in terms of mean length of stay or hospital charges. CONCLUSIONS All-cause inpatient mortality was similar in AMICS+AF patients undergoing PCI who were treated with either pVAD or IABP. The pVAD group, however, experienced more complications while consuming greater healthcare resources.
Collapse
Affiliation(s)
- Rupak Desai
- Division of Cardiology, Atlanta VA Medical Center, Decatur, GA, United States
| | - Bishoy Hanna
- Division of Cardiology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Sandeep Singh
- Division of Clinical Epidemiology, Biostatistics, and Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sonu Gupta
- Division of Cardiology, Morehouse School of Medicine, Atlanta, GA, United States
| | | | - Gautam Kumar
- Division of Cardiology, Atlanta VA Medical Center, Decatur, GA, United States; Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Rajesh Sachdeva
- Division of Cardiology, Atlanta VA Medical Center, Decatur, GA, United States; Division of Cardiology, Morehouse School of Medicine, Atlanta, GA, United States; Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States; Division of Cardiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Adam E Berman
- Division of Cardiology, Medical College of Georgia, Augusta University, Augusta, GA, United States; Division of Health Economics and Modeling, Medical College of Georgia, Augusta University, Augusta, GA, United States.
| |
Collapse
|