1
|
Punnanithinont N, Kambalapalli S, Iskander B, Ichikawa K, Krishnan S, Lakshmanan S, Roy SK, Budoff M. "Anti-inflammatory Therapies in Atherosclerosis - Where are we going?". Curr Atheroscler Rep 2024; 27:19. [PMID: 39699771 DOI: 10.1007/s11883-024-01267-7] [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] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
PURPOSE OF REVIEW Inflammation has been commonly known for the past decade as a part of the pathophysiology of atherosclerosis, along with lipid accumulation. However, some patients with optimized lipid-lowering therapy still have elevated inflammatory biomarkers. Anti-inflammation therapies were developed to eradicate this residual risk. We summarized the primary inflammatory pathway and recent clinical trials in anti-inflammation therapies. RECENT FINDINGS Colchicine Cardiovascular Outcomes Trial (COLCOT) and LoDoCo2 (Colchicine Reduces Risk of Major Cardiovascular Events in Chronic Coronary Disease) found that low-dose colchicine significantly reduced cardiovascular death, myocardial infarction (MI), ischemic stroke and coronary revascularization in patients with recent MI within 30 days and chronic coronary disease respectively. The US Food and Drug Administration approved low-dose colchicine in 2023 for patients with established atherosclerotic cardiovascular disease (ASCVD). However, its use was limited for chronic kidney disease (CKD) patients. Reduction in Inflammation in Patients with Advanced Chronic Renal Disease Utilizing Antibody Mediated Interleukin-6 Inhibition (RESCUE) was conducted using Ziltivekimab, an IL-6 ligand monoclonal antibody and found that it significantly reduced high-sensitivity C-reactive protein, an inflammatory surrogate marker. There is an ongoing phase-3 clinical trial, Ziltivekimab Versus Placebo Cardiovascular Outcomes in Participants with Atherosclerotic Cardiovascular Disease, Chronic Kidney Disease, and Systemic Inflammation trial (ZEUS), which will be essential for further anti-inflammation therapy for patients with CKD. Numerous clinical trials have investigated anti-inflammation therapies. Colchicine is by far the only one that has the potential to be widely used due to its cost-effectiveness. Further research is needed on other novel anti-inflammation therapies and their real-world implementation.
Collapse
Affiliation(s)
- Natdanai Punnanithinont
- The Lundquist Institute, UCLA Medical Center Harbor, 1124 W Carson St, CA 90502, Torrance, US.
| | - Soumya Kambalapalli
- The Lundquist Institute, UCLA Medical Center Harbor, 1124 W Carson St, CA 90502, Torrance, US
| | - Beshoy Iskander
- The Lundquist Institute, UCLA Medical Center Harbor, 1124 W Carson St, CA 90502, Torrance, US
| | - Keishi Ichikawa
- The Lundquist Institute, UCLA Medical Center Harbor, 1124 W Carson St, CA 90502, Torrance, US
| | - Srikanth Krishnan
- The Lundquist Institute, UCLA Medical Center Harbor, 1124 W Carson St, CA 90502, Torrance, US
| | - Suvasini Lakshmanan
- The Lundquist Institute, UCLA Medical Center Harbor, 1124 W Carson St, CA 90502, Torrance, US
| | - Sion K Roy
- The Lundquist Institute, UCLA Medical Center Harbor, 1124 W Carson St, CA 90502, Torrance, US
| | - Matthew Budoff
- The Lundquist Institute, UCLA Medical Center Harbor, 1124 W Carson St, CA 90502, Torrance, US
| |
Collapse
|
2
|
Zuriaga MA, Yu Z, Matesanz N, Truong B, Ramos-Neble BL, Asensio-López MC, Uddin MM, Nakao T, Niroula A, Zorita V, Amorós-Pérez M, Moro R, Ebert BL, Honigberg MC, Pascual-Figal D, Natarajan P, Fuster JJ. Colchicine prevents accelerated atherosclerosis in TET2-mutant clonal haematopoiesis. Eur Heart J 2024; 45:4601-4615. [PMID: 39212933 PMCID: PMC11560281 DOI: 10.1093/eurheartj/ehae546] [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: 03/17/2023] [Revised: 05/14/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS Somatic mutations in the TET2 gene that lead to clonal haematopoiesis (CH) are associated with accelerated atherosclerosis development in mice and a higher risk of atherosclerotic disease in humans. Mechanistically, these observations have been linked to exacerbated vascular inflammation. This study aimed to evaluate whether colchicine, a widely available and inexpensive anti-inflammatory drug, prevents the accelerated atherosclerosis associated with TET2-mutant CH. METHODS In mice, TET2-mutant CH was modelled using bone marrow transplantations in atherosclerosis-prone Ldlr-/- mice. Haematopoietic chimeras carrying initially 10% Tet2-/- haematopoietic cells were fed a high-cholesterol diet and treated with colchicine or placebo. In humans, whole-exome sequencing data and clinical data from 37 181 participants in the Mass General Brigham Biobank and 437 236 participants in the UK Biobank were analysed to examine the potential modifying effect of colchicine prescription on the relationship between CH and myocardial infarction. RESULTS Colchicine prevented accelerated atherosclerosis development in the mouse model of TET2-mutant CH, in parallel with suppression of interleukin-1β overproduction in conditions of TET2 loss of function. In humans, patients who were prescribed colchicine had attenuated associations between TET2 mutations and myocardial infarction. This interaction was not observed for other mutated genes. CONCLUSIONS These results highlight the potential value of colchicine to mitigate the higher cardiovascular risk of carriers of somatic TET2 mutations in blood cells. These observations set the basis for the development of clinical trials that evaluate the efficacy of precision medicine approaches tailored to the effects of specific mutations linked to CH.
Collapse
Affiliation(s)
- María A Zuriaga
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Zhi Yu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
| | - Nuria Matesanz
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Buu Truong
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
| | - Beatriz L Ramos-Neble
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Mari C Asensio-López
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Cardiology Department, Hospital Virgen de la Arrixaca, IMIB-Arrixaca and University of Murcia, Murcia, Spain
| | - Md Mesbah Uddin
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
| | - Tetsushi Nakao
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Abhishek Niroula
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Virginia Zorita
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Marta Amorós-Pérez
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Rosa Moro
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael C Honigberg
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Domingo Pascual-Figal
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Cardiology Department, Hospital Virgen de la Arrixaca, IMIB-Arrixaca and University of Murcia, Murcia, Spain
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA
| | - José J Fuster
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| |
Collapse
|
3
|
Klimek K, Groener D, Chen X, Rowe SP, Speer T, Higuchi T, Werner RA. Molecular imaging along the heart-kidney axis. Theranostics 2024; 14:7111-7121. [PMID: 39629123 PMCID: PMC11610144 DOI: 10.7150/thno.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/22/2024] [Indexed: 12/06/2024] Open
Abstract
Cardiorenal syndrome (CRS) involves bidirectional crosstalk between the failing heart and the kidneys. Depending on the primum movens (primary cardiac or renal injury), systems-based interactions in the secondary affected organ may include pro-fibrotic signaling, overzealous inflammation, impaired nerve integrity or overactivity of specific renal transporters mediating glucose absorption. Those pathophysiological pillars can be investigated by molecular imaging using SPECT or PET agents. Targeted whole-body molecular imaging may allow for a) systems-based analysis along the heart-kidney axis, b) may provide prognostic information on longitudinal organ-based functional decline or c) may be used for guidance of reparative intervention based on peak activation identified on PET (paradigm of cardiorenal theranostics). We will discuss the current state of translational molecular imaging for CRS, along with future clinical aspects in the field.
Collapse
Affiliation(s)
- Konrad Klimek
- Goethe University Frankfurt, University Hospital, Department of Nuclear Medicine, Clinic for Radiology and Nuclear Medicine, Frankfurt, Germany
| | - Daniel Groener
- Goethe University Frankfurt, University Hospital, Department of Nuclear Medicine, Clinic for Radiology and Nuclear Medicine, Frankfurt, Germany
| | - Xinyu Chen
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Bavaria 86156, Germany
| | - Steven P. Rowe
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Thimoteus Speer
- Department of Internal Medicine 4 - Nephrology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Else Kroener-Fresenius-Zentrum for Nephrological Research, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine and Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Rudolf A. Werner
- Goethe University Frankfurt, University Hospital, Department of Nuclear Medicine, Clinic for Radiology and Nuclear Medicine, Frankfurt, Germany
- Division of Nuclear Medicine and Molecular Imaging, The Russell H Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- DZHK (German Centre for Cardiovascular Research), Partner Site Frankfurt Rhine-Main, Frankfurt, Germany
| |
Collapse
|
4
|
Kumar V, Vellapandian C. Unraveling the Nexus Between Ambient Air Pollutants and Cardiovascular Morbidity: Mechanistic Insights and Therapeutic Horizons. Cureus 2024; 16:e68650. [PMID: 39371734 PMCID: PMC11452354 DOI: 10.7759/cureus.68650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Air pollution poses a significant threat to cardiovascular health, contributing to the development and progression of various heart diseases. This review delves into the intricate relationship between ambient air pollutants and cardiovascular morbidity, elucidating the underlying mechanisms and exploring potential therapeutic approaches. We discuss the major types of air pollutants, including particulate matter (PM), nitrogen dioxide (NO2), sulfur dioxide (SO2), ozone (O3), and carbon monoxide (CO), and their respective roles in exacerbating cardiovascular conditions. The review highlights the key mechanisms by which air pollutants adversely impact the cardiovascular system, including systemic inflammation, oxidative stress, endothelial dysfunction, autonomic nervous system imbalance, and dysregulation of blood coagulation and thrombosis. Vulnerable populations, including children, the elderly, and those with pre-existing health conditions, are disproportionately affected. Air quality regulations aim to mitigate these effects by reducing pollutant levels, with the overall goal of lowering cardiovascular morbidity and improving public health outcomes. Specifically, stringent regulations focus on curbing vehicular emissions and industrial pollutants and promoting cleaner energy sources. Recent data underscore the importance of addressing environmental and behavioral risk factors to prevent the growing global burden of cardiovascular disease. This review synthesizes the mechanistic pathways through which pollutants contribute to cardiovascular damage and highlights the urgent need for early detection strategies and targeted therapies. Improving public health through stricter air quality control measures and raising awareness of the health risks associated with pollution is crucial for mitigating the long-term cardiovascular impacts of air pollution.
Collapse
Affiliation(s)
- Vishal Kumar
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, IND
| | - Chitra Vellapandian
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, IND
| |
Collapse
|
5
|
Bay B, Arnold N, Waldeyer C. C-reactive protein, pharmacological treatments and diet: how to target your inflammatory burden. Curr Opin Lipidol 2024; 35:141-148. [PMID: 38277208 DOI: 10.1097/mol.0000000000000922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
PURPOSE OF REVIEW This article focuses on pharmacological agents as well as dietary changes aimed at the reduction of the inflammatory burden measured by circulating C-reactive protein concentrations. RECENT FINDINGS Over the last years, repurposed as well as new anti-inflammatory agents have been investigated in outcome trials in the cardiovascular field. Currently, a specific inhibition of the inflammatory cascade via the interleukin-6 ligand antibody ziltivekimab is being explored in large-scale outcome trials, after the efficacy of this agent with regard to the reduction of inflammatory biomarkers was proven recently. Next to the investigated pharmacological agents, specific dietary patterns possess the ability to improve the inflammatory burden. This enables patients themselves to unlock a potential health benefit ahead of the initiation of a specific medication targeting the inflammatory pathway. SUMMARY Both pharmacological agents as well as diet provide the opportunity to improve the inflammatory profile and thereby lower C-reactive protein concentrations. Whilst advances in the field of specific anti-inflammatory treatments have been made over the last years, their broad implementation is currently limited. Therefore, optimization of diet (and other lifestyle factors) could provide a cost effective and side-effect free intervention to target low-grade vascular inflammation.
Collapse
Affiliation(s)
- Benjamin Bay
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natalie Arnold
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck
- Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Waldeyer
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck
| |
Collapse
|
6
|
Li JN, Wang MY, Tan YR, Wang LL. Multidirectional Intervention of Chinese Herbal Medicine in the Prevention and Treatment of Atherosclerosis: From Endothelial Protection to Immunomodulation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:925-947. [PMID: 38798151 DOI: 10.1142/s0192415x24500381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Atherosclerosis is a significant risk factor for developing cardiovascular disease and a leading cause of death worldwide. The occurrence of atherosclerosis is closely related to factors such as endothelial injury, lipid deposition, immunity, and inflammation. Conventional statins, currently used in atherosclerosis treatment, have numerous adverse side effects that limit their clinical utility, prompting the urgent need to identify safer and more effective therapeutic alternatives. Growing evidence indicates the significant potential of Chinese herbs in atherosclerosis treatment. Herbal monomer components, such as natural flavonoid compounds extracted from herbs like Coptis chinensis and Panax notoginseng, have been utilized for their lipid-lowering and inflammation-inhibiting effects in atherosclerosis treatment. These herbs can be used as single components in treating diseases and with other Chinese medicines to form herbal combinations. This approach targets the disease mechanism in multiple ways, enhancing the therapeutic effects. Thus, this review examines the roles of Chinese herbal medicine monomers and Chinese herbal compounds in inhibiting atherosclerosis, including regulating lipids, improving endothelial function, reducing oxidative stress, regulating inflammation and the immune response, and apoptosis. By highlighting these roles, our study offers new perspectives on atherosclerosis treatment with Chinese herbs and is anticipated to contribute to advancements in related research fields.
Collapse
Affiliation(s)
- Jia-Ni Li
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| | - Meng-Yu Wang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| | - Yu-Rong Tan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| | - Li-Li Wang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| |
Collapse
|
7
|
Mohebbi D, Ogurtsova K, Dyczmons J, Dintsios M, Kairies-Schwarz N, Jung C, Icks A. Cost-effectiveness of incentives for physical activity in coronary heart disease in Germany: pre-trial health economic model of a complex intervention following the new MRC framework. BMJ Open Sport Exerc Med 2024; 10:e001896. [PMID: 38808264 PMCID: PMC11131112 DOI: 10.1136/bmjsem-2024-001896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
Objectives The German Incentives for Physical Activity in Cardiac Patients trial is a three-arm, randomised controlled trial for secondary prevention of coronary heart disease (CHD). Guidance for developing complex interventions recommends pre-trial health economic modelling. The aim of this study is to model the long-term cost-effectiveness of the incentive-based physical activity interventions in a population with CHD. Methods A decision-analytical Markov model was developed from a health services provider perspective, following a cohort aged 65 years with a previous myocardial infarction for 25 years. Monetary and social incentives were compared relative to no incentive. Intervention effects associated with physical activity were used to determine the costs, quality-adjusted life-years (QALYs) gained, incremental cost-effectiveness and cost-utility ratios. The probability of cost-effectiveness was calculated through sensitivity analyses. Results The incremental QALYs gained from the monetary and social incentives, relative to control, were respectively estimated at 0.01 (95% CI 0.00 to 0.01) and 0.04 (95% CI 0.02 to 0.05). Implementation of the monetary and social incentive interventions increased the costs by €874 (95% CI €744 to €1047) and €909 (95% CI €537 to €1625). Incremental cost-utility ratios were €25 912 (95% CI €15 056 to €50 210) and €118 958 (95% CI €82 930 to €196 121) per QALY gained for the social and monetary incentive intervention, respectively. With a willingness-to-pay threshold set at €43 000/QALY, equivalent to the per-capita gross domestic product in Germany, the probability that the social and monetary incentive intervention would be seen as cost-effective was 95% and 0%, respectively. Conclusions Exercise-based secondary prevention using inventive schemes may offer a cost-effective strategy to reduce the burden of CHD.
Collapse
Affiliation(s)
- Damon Mohebbi
- Institute for Health Services Research and Health Economics, Centre for Health and Society, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Health Services Research and Health Economics, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katherine Ogurtsova
- Institute of Occupational, Social and Environmental Medicine, Centre for Health and Society, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jan Dyczmons
- Institute for Health Services Research and Health Economics, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Markos Dintsios
- Institute for Health Services Research and Health Economics, Centre for Health and Society, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Nadja Kairies-Schwarz
- Institute for Health Services Research and Health Economics, Centre for Health and Society, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christian Jung
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID (Cardiovascular Research Institute Düsseldorf), Düsseldorf, Germany
| | - Andrea Icks
- Institute for Health Services Research and Health Economics, Centre for Health and Society, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Health Services Research and Health Economics, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
8
|
Maidana D, Arroyo-Álvarez A, Arenas-Loriente A, Barreres-Martín G, Muñoz-Alfonso C, Bompart Berroteran D, Esteve Claramunt F, Blanco del Burgo R, Cepas-Guillén P, Garcia-Blas S, Bonanad C. Inflammation as a New Therapeutic Target among Older Patients with Ischemic Heart Disease. J Clin Med 2024; 13:363. [PMID: 38256497 PMCID: PMC10816645 DOI: 10.3390/jcm13020363] [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: 12/17/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular (CV) diseases remain a global health challenge, with ischemic heart disease (IHD) being the primary cause of both morbidity and mortality. Despite optimal pharmacological therapy, older patients with IHD exhibit an increased susceptibility to recurrent ischemic events, significantly impacting their prognosis. Inflammation is intricately linked with the aging process and plays a pivotal role in the evolution of atherosclerosis. Emerging anti-inflammatory therapies have shown promise in reducing ischemic events among high-risk populations. This review aims to explore the potential of targeted anti-inflammatory interventions in improving clinical outcomes and the quality of life for older patients with IHD.
Collapse
Affiliation(s)
- Daniela Maidana
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Andrea Arroyo-Álvarez
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | | | - Guillermo Barreres-Martín
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Carles Muñoz-Alfonso
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Daznia Bompart Berroteran
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Francisca Esteve Claramunt
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | - Regina Blanco del Burgo
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
| | | | - Sergio Garcia-Blas
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
- Cardiology Department, Clinic University Hospital of Valencia, 46026 Valencia, Spain
| | - Clara Bonanad
- INCLIVA—Instituto de Investigación Sanitaria, Biomedical Research Institute, 46010 Valencia, Spain (F.E.C.)
- Cardiology Department, Clinic University Hospital of Valencia, 46026 Valencia, Spain
| |
Collapse
|
9
|
Luan Y, Luan Y, Jiao Y, Liu H, Huang Z, Feng Q, Pei J, Yang Y, Ren K. Broadening Horizons: Exploring mtDAMPs as a Mechanism and Potential Intervention Target in Cardiovascular Diseases. Aging Dis 2023; 15:2395-2416. [PMID: 38270118 PMCID: PMC11567272 DOI: 10.14336/ad.2023.1130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/30/2023] [Indexed: 01/26/2024] Open
Abstract
Cardiovascular diseases (CVDs) have been recognized as the leading cause of premature mortality and morbidity worldwide despite significant advances in therapeutics. Inflammation is a key factor in CVD progression. Once stress stimulates cells, they release cellular compartments known as damage-associated molecular patterns (DAMPs). Mitochondria can release mitochondrial DAMPs (mtDAMPs) to initiate an immune response when stimulated with cellular stress. Investigating the molecular mechanisms underlying the DAMPs that regulate CVD progression is crucial for improving CVDs. Herein, we discuss the composition and mechanism of DAMPs, the significance of mtDAMPs in cellular inflammation, the presence of mtDAMPs in different types of cells, and the main signaling pathways associated with mtDAMPs. Based on this, we determined the role of DAMPs in CVDs and the effects of mtDAMP intervention on CVD progression. By offering a fresh perspective and comprehensive insights into the molecular mechanisms of DAMPs, this review seeks to provide important theoretical foundations for developing drugs targeting CVDs.
Collapse
Affiliation(s)
- Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Ying Luan
- State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China.
| | - Yuxue Jiao
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Hui Liu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.
| | - Zhen Huang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.
| | - Qi Feng
- Department ofIntegrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Jinyan Pei
- Quality Management Department, The Third People’s Hospital of Henan Provine, Zhengzhou, China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
10
|
Tang J, Li T, Xiong X, Yang Q, Su Z, Zheng M, Chen Q. Colchicine delivered by a novel nanoparticle platform alleviates atherosclerosis by targeted inhibition of NF-κB/NLRP3 pathways in inflammatory endothelial cells. J Nanobiotechnology 2023; 21:460. [PMID: 38037046 PMCID: PMC10690998 DOI: 10.1186/s12951-023-02228-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by arterial plaque formation, is one of the most prominent causes of cardiovascular diseases. However, the current treatments often do not adequately compromise the chronic inflammation-mediated plaque accumulation and the disease progression. Therefore, a new and effective strategy that blocks atherosclerosis-associated inflammation is urgently needed to further reduce the risk. Colchicine, a potent anti-inflammatory medication, has shown great potential in the treatment of atherosclerosis, but its adverse effects have hampered its clinical application. Herein, we developed a novel delivery nanosystem encapsulated with colchicine (VHPK-PLGA@COL), which exhibited improved biosafety and sustained drug release along with the gradual degradation of PLGA and PEG as confirmed both in vitro and in vivo. Surface modification of the nanoparticles with the VHPK peptide ensured its capability to specifically target inflammatory endothelial cells and alleviate atherosclerotic plaque accumulation. In the ApoE - / - atherosclerotic mouse model, both colchicine and VHPK-PLGA@COL treatment significantly decreased the plaque area and enhanced plaque stability by blocking the NF-κB/NLRP3 pathways, while VHPK-PLGA@COL exhibited enhanced therapeutic effects due to its unique ability to target inflammatory endothelial cells without obvious long-term safety concerns. In summary, VHPK-PLGA@COL has the potential to overcome the key translational barriers of colchicine and open new avenues to repurpose this drug for anti-atherosclerotic therapy.
Collapse
Affiliation(s)
- Juan Tang
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Endocrinology, The First People's Hospital of Ziyang, Sichuan, 641300, China
| | - Tao Li
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Ophthalmology, The First People's Hospital of Ziyang, Sichuan, 641300, China
| | - Xiaojing Xiong
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qiaoyun Yang
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zedazhong Su
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Minming Zheng
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Qingwei Chen
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
11
|
Boczar KE, Beanlands R, Wells G, Coyle D. Cost-effectiveness of colchicine for recurrent cardiovascular events. CJC Open 2023. [DOI: 10.1016/j.cjco.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
12
|
May L, Bartolo B, Harrison D, Guzik T, Drummond G, Figtree G, Ritchie R, Rye KA, de Haan J. Translating atherosclerosis research from bench to bedside: navigating the barriers for effective preclinical drug discovery. Clin Sci (Lond) 2022; 136:1731-1758. [PMID: 36459456 PMCID: PMC9727216 DOI: 10.1042/cs20210862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/21/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. An ongoing challenge remains the development of novel pharmacotherapies to treat CVD, particularly atherosclerosis. Effective mechanism-informed development and translation of new drugs requires a deep understanding of the known and currently unknown biological mechanisms underpinning atherosclerosis, accompanied by optimization of traditional drug discovery approaches. Current animal models do not precisely recapitulate the pathobiology underpinning human CVD. Accordingly, a fundamental limitation in early-stage drug discovery has been the lack of consensus regarding an appropriate experimental in vivo model that can mimic human atherosclerosis. However, when coupled with a clear understanding of the specific advantages and limitations of the model employed, preclinical animal models remain a crucial component for evaluating pharmacological interventions. Within this perspective, we will provide an overview of the mechanisms and modalities of atherosclerotic drugs, including those in the preclinical and early clinical development stage. Additionally, we highlight recent preclinical models that have improved our understanding of atherosclerosis and associated clinical consequences and propose model adaptations to facilitate the development of new and effective treatments.
Collapse
Affiliation(s)
- Lauren T. May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | | | - David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN, U.S.A
| | - Tomasz Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Gemma A. Figtree
- Kolling Research Institute, University of Sydney, Sydney, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Rebecca H. Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney 2052, Australia
| | - Judy B. de Haan
- Cardiovascular Inflammation and Redox Biology Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- Department Cardiometabolic Health, University of Melbourne, Parkville, Victoria 3010, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria 3086, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| |
Collapse
|
13
|
Cost-effectiveness of folic acid therapy for primary prevention of stroke in patients with hypertension. BMC Med 2022; 20:407. [PMID: 36280851 PMCID: PMC9594871 DOI: 10.1186/s12916-022-02601-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 10/11/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND For hypertensive patients without a history of stroke or myocardial infarction (MI), the China Stroke Primary Prevention Trial (CSPPT) demonstrated that treatment with enalapril-folic acid reduced the risk of primary stroke compared with enalapril alone. Whether folic acid therapy is an affordable and beneficial treatment strategy for the primary prevention of stroke in hypertensive patients from the Chinese healthcare sector perspective has not been thoroughly explored. METHODS We performed a cost-effectiveness analysis alongside the CSPPT, which randomized 20,702 hypertensive patients. A patient-level microsimulation model based on the 4.5-year period of in-trial data was used to estimate costs, life years, quality-adjusted life years (QALYs), and incremental cost-effectiveness ratios (ICERs) for enalapril-folic acid vs. enalapril over a lifetime horizon from the payer perspective. RESULTS During the in-trial follow-up period, patients receiving enalapril-folic acid gained an average of 0.016 QALYs related primarily to reductions in stroke, and the incremental cost was $706.03 (4553.92 RMB). Over a lifetime horizon, enalapril-folic acid treatment was projected to increase quality-adjusted life years by 0.06 QALYs or 0.03 life-year relative to enalapril alone at an incremental cost of $1633.84 (10,538.27 RMB), resulting in an ICER for enalapril-folic acid compared with enalapril alone of $26,066.13 (168,126.54 RMB) per QALY gained and $61,770.73 (398,421.21 RMB) per life-year gained, respectively. A probabilistic sensitivity analysis demonstrated that enalapril-folic acid compared with enalapril would be economically attractive in 74.5% of simulations at a threshold of $37,663 (242,9281 RMB) per QALY (3x current Chinese per capita GDP). Several high-risk subgroups had highly favorable ICERs < $12,554 (80,976 RMB) per QALY (1x GDP). CONCLUSIONS For both in-trial and over a lifetime, it appears that enalapril-folic acid is a clinically and economically attractive medication compared with enalapril alone. Adding folic acid to enalapril may be a cost-effective strategy for the prevention of primary stroke in hypertensive patients from the Chinese health system perspective.
Collapse
|
14
|
Chaffey L, Roberti A, Greaves DR. Drug repurposing in cardiovascular inflammation: Successes, failures, and future opportunities. Front Pharmacol 2022; 13:1046406. [PMID: 36339576 PMCID: PMC9634418 DOI: 10.3389/fphar.2022.1046406] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/10/2022] [Indexed: 12/15/2022] Open
Abstract
Drug repurposing is an attractive, pragmatic approach to drug discovery that has yielded success across medical fields over the years. The use of existing medicines for novel indications enables dramatically reduced development costs and timescales compared with de novo drug discovery and is therefore a promising strategy in cardiovascular disease, where new drug approvals lag significantly behind that of other fields. Extensive evidence from pre-clinical and clinical studies show that chronic inflammation is a driver of pathology in cardiovascular disease, and many efforts have been made to target cardiovascular inflammation therapeutically. This approach has been met with significant challenges however, namely off-target effects associated with broad-spectrum immunosuppression, particularly in long-term conditions such as cardiovascular disease. Nevertheless, multiple anti-inflammatory medicines have been assessed for efficacy in cardiovascular clinical trials, with most of these being repurposed from their original indications in autoimmune conditions like rheumatoid arthritis. In this review, we discuss the mixed successes of clinical trials investigating anti-inflammatory drugs in cardiovascular disease, with examples such as anti-cytokine monoclonal antibodies, colchicine, and methotrexate. Looking to the future, we highlight potential new directions for drug repurposing in cardiovascular inflammation, including the emerging concepts of drug re-engineering and chrono-pharmacology.
Collapse
Affiliation(s)
| | | | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
Cowie MR, Bozkurt B, Butler J, Briggs A, Kubin M, Jonas A, Adler AI, Patrick-Lake B, Zannad F. How can we optimise health technology assessment and reimbursement decisions to accelerate access to new cardiovascular medicines? Int J Cardiol 2022; 365:61-68. [PMID: 35905826 DOI: 10.1016/j.ijcard.2022.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/26/2022] [Accepted: 07/12/2022] [Indexed: 11/18/2022]
Abstract
Regulatory approvals of, and subsequent access to, innovative cardiovascular medications have declined. How much of this decline relates to the final step of gaining reimbursement for new treatments is unknown. Payers and health technology assessment (HTA) bodies look beyond efficacy and safety to assess whether a new drug improves patient outcomes, quality of life, or satisfaction at a cost that is affordable compared to existing treatments. HTA bodies work within a limited healthcare budget, and this is one of the reasons why only half of newly approved drugs are accepted for reimbursement, or receive restricted or "optimised" recommendations from HTA bodies. All stakeholders have the common goal of facilitating access to safe, effective, and affordable treatments to appropriate patients. An important strategy to expedite this is providing optimal data. This is demonstrably facilitated by early (and ongoing) discussions between all stakeholders. Many countries have formal programmes to provide collaborative regulatory and HTA advice to developers. Other strategies include aligning regulatory and HTA processes, increasing use of real-world evidence, formally defining the decision-making process, and educating stakeholders on the criteria for positive decision making. Industry should focus on developing treatments for unmet medical needs, seek early engagement with HTA and regulatory bodies, improve methodologies for optimal price setting, develop internal systems to collaborate with national and international stakeholders, and conduct post-approval studies. Patient involvement in all stages of development, including HTA, is critical to capture the lived experience and priorities of those whose lives will be impacted by new treatment approvals.
Collapse
Affiliation(s)
- Martin R Cowie
- Royal Brompton Hospital & School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK.
| | - Biykem Bozkurt
- Winters Center for Heart Failure, Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Javed Butler
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Andrew Briggs
- Department of Health Services Research & Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Maria Kubin
- Department of Integrated Evidence Generation, Bayer AG, Wuppertal, Germany
| | - Adrian Jonas
- National Institute for Health and Care Excellence (NICE), London, UK
| | - Amanda I Adler
- Diabetes Trial Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism (OCDEM), Oxford, UK
| | - Bray Patrick-Lake
- Department of Strategic Partnerships, Evidation Health, San Mateo, CA, USA
| | - Faiez Zannad
- Université de Lorraine, Inserm Clinical Investigation Center at Institut Lorrain du Coeur et des Vaisseaux, University Hospital of Nancy, Nancy, France
| |
Collapse
|
16
|
Maierean S, Webb R, Banach M, Mazidi M. The role of inflammation and the possibilities of inflammation reduction to prevent cardiovascular events. EUROPEAN HEART JOURNAL OPEN 2022; 2:oeac039. [PMID: 35919577 PMCID: PMC9271640 DOI: 10.1093/ehjopen/oeac039] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 05/27/2022] [Indexed: 11/16/2022]
Abstract
Chronic systemic inflammation is a risk factor for cardiovascular (CV) disease (CVD). Whether this relationship extends to subclinical inflammation, quantified by values of circulating markers associated with inflammation in the high range of the normal interval, remains debatable. This narrative review evaluates evidence exploring this relationship. A review of pharmacological and non-pharmacological interventions, including diet and lifestyle strategies, supplements, nutraceuticals, and other natural substances aimed at reducing inflammation was also conducted, since few reviews have synthesized this literature. PubMed and EMBASE were used to search the literature and several well-studied triggers of inflammation [oxidized LDL, Lp(a), as well as C-reactive protein (CRP)/high-sensitivity CRP (hs-CRP)] were included to increase sensitivity and address the lack of existing reviews summarizing their influence in the context of inflammation. All resulting references were assessed. Overall, there is good data supporting associations between circulating hs-CRP and CV outcomes. However, the same was not seen in studies evaluating triggers of inflammation, such as oxidized LDL or Lp(a). There is also insufficient evidence showing treatments to target inflammation and lead to reductions in hs-CRP result in improvements in CV outcomes, particularly in those with normal baseline levels of hs-CRP. Regarding pharmacological interventions, statins, bempedoic acid, and apabetalone significantly reduce circulating hs-CRP, unlike PCSK-9 inhibitors. A variety of natural substances and vitamins were also evaluated and none reduced hs-CRP. Regarding non-pharmacological interventions, weight loss was strongly associated with reductions in circulating hs-CRP, whereas various dietary interventions and exercise regimens were not, unless accompanied by weight loss.
Collapse
Affiliation(s)
- Serban Maierean
- Department of Medicine, University of Toronto , Toronto, ON , Canada
| | - Richard Webb
- Faculty of Science, Liverpool Hope University , Taggart Avenue, Liverpool , UK
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz , Rzgowska 281/289, Lodz 93-338 , Poland
- Department of Cardiology and Adult Congenital Heart Diseases, Polish Mother’s Memorial Hospital Research Institute (PMMHRI) , Rzgowska 281/289, Lodz 93-338 , Poland
- Cardiovascular Research Centre, University of Zielona Gora , Zyty 28, 65-046 Zielona Gora , Poland
| | - Mohsen Mazidi
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health , University of Oxford, Oxford , UK
- Department of Twin Research & Genetic Epidemiology, King’s College London , South Wing St Thomas’, London , UK
| |
Collapse
|
17
|
Ng C, Lee KL, Muthiah MD, Wu KX, Chioh FWJ, Tan K, Soon GST, Shabbir A, Loo WM, Low ZS, Chen Q, Tan NS, Ng HH, Dan YY, Cheung C. Endothelial‐immune crosstalk contributes to vasculopathy in nonalcoholic fatty liver disease. EMBO Rep 2022; 23:e54271. [PMID: 35403791 PMCID: PMC9171677 DOI: 10.15252/embr.202154271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022] Open
Abstract
The top cause of mortality in patients with nonalcoholic fatty liver disease (NAFLD) is cardiovascular complications. However, mechanisms of NAFLD‐associated vasculopathy remain understudied. Here, we show that blood outgrowth endothelial cells (BOECs) from NAFLD subjects exhibit global transcriptional upregulation of chemokines and human leukocyte antigens. In mouse models of diet‐induced NAFLD, we confirm heightened endothelial expressions of CXCL12 in the aortas and the liver vasculatures, and increased retention of infiltrated leukocytes within the vessel walls. To elucidate endothelial‐immune crosstalk, we performed immunoprofiling by single‐cell analysis, uncovering T cell intensification in NAFLD patients. Functionally, treatment with a CXCL12‐neutralizing antibody is effective at moderating the enhanced chemotactic effect of NAFLD BOECs in recruiting CD8+ T lymphocytes. Interference with the CXCL12‐CXCR4 axis using a CXCR4 antagonist also averts the impact of immune cell transendothelial migration and restores endothelial barrier integrity. Clinically, we detect threefold more circulating damaged endothelial cells in NAFLD patients than in healthy controls. Our work provides insight into the modulation of interactions with effector immune cells to mitigate endothelial injury in NAFLD.
Collapse
Affiliation(s)
- Chun‐Yi Ng
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Khang Leng Lee
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Mark Dhinesh Muthiah
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Medicine National University Health System Singapore Singapore
| | - Kan Xing Wu
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | | | - Konstanze Tan
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | | | - Asim Shabbir
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Surgery University Surgical Cluster National University Health System Singapore Singapore
| | - Wai Mun Loo
- Department of Medicine National University Health System Singapore Singapore
| | - Zun Siong Low
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology Agency for Science Technology and Research (A*STAR) Singapore Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- School of Biological Sciences Nanyang Technological University Singapore Singapore
| | - Huck Hui Ng
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Institute of Molecular and Cell Biology Agency for Science Technology and Research (A*STAR) Singapore Singapore
- School of Biological Sciences Nanyang Technological University Singapore Singapore
- Genome Institute of Singapore Agency for Science Technology and Research (A*STAR) Singapore Singapore
| | - Yock Young Dan
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Medicine National University Health System Singapore Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- Institute of Molecular and Cell Biology Agency for Science Technology and Research (A*STAR) Singapore Singapore
| |
Collapse
|
18
|
Boczar KE, Beanlands R, Wells G, Coyle D. Cost-Effectiveness of Canakinumab From a Canadian Perspective for Recurrent Cardiovascular Events. CJC Open 2022; 4:441-448. [PMID: 35607490 PMCID: PMC9123368 DOI: 10.1016/j.cjco.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/07/2022] [Indexed: 11/22/2022] Open
Abstract
Background Cardiovascular (CV) disease is a condition with high levels of morbidity and mortality. Canakinumab is a novel monoclonal antibody therapy that has been shown to reduce CV events but is associated with side effects and high cost. The main objective for this analysis is to determine whether canakinumab use is cost-effective for the prevention of recurrent CV events. Methods A decision model was developed to estimate the direct costs and outcomes among patients who have suffered a myocardial infarction and are treated with canakinumab. A lifetime study horizon was used to analyze the base-case costs and utilities from the perspective of the Canadian publicly funded healthcare system. Markov modeling was used in combination with Monte Carlo simulation to derive expected values for costs and quality-adjusted life years (QALYs), permitting the calculation of incremental cost-effectiveness ratios. Results Canakinumab was associated with higher average lifetime costs per patient ($457,982 vs $82,565) and higher average QALYs per patient (14.90 vs 14.20), compared with standard of care. Thus, the incremental cost per QALY gained for canakinumab treatment vs standard-of-care therapy was $535,365. The probability that canakinumab treatment is cost-effective was 0%. Results were consistent over a range of scenario analyses. Conclusions Treatment of patients post-myocardial infarction with canakinumab is not cost-effective, compared with standard-of-care therapy at the current price. Based on currently accepted willingness-to-pay thresholds in Canada, a reduction in price of 91% is required to yield a cost per patient that would be considered appropriate.
Collapse
Affiliation(s)
- Kevin E. Boczar
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Rob Beanlands
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - George Wells
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Doug Coyle
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
19
|
Abstract
Atherosclerosis is considered a chronic, inflammatory disease responsible for more than 15% of all global deaths, secondary to its complications of myocardial infarction, vascular disease, and stroke. Current treatment regimens consist of lipid-lowering pharmaceuticals, control of risk factors, and prevention of plaque rupture and thrombosis with antiplatelet agents. However, a significant burden on society remains due to the morbidity and mortality of coronary artery disease despite our best practices. In addition to dyslipidemia and hemostasis, inflammation has now moved to the proverbial forefront as the remaining obstacle to appropriate management of atherosclerosis. A complex dance of endothelial dysfunction, complement activation, and immune cell-mediated cytokine release underlie the pathogenesis of atherosclerotic plaque development, destabilization, and rupture. Cholesterol-induced sterile inflammation is thought to be central to this process via activation of a protein complex called the nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3 (NLRP3) inflammasome. The focus of this review article will be to examine the NLRP3 inflammasome, which directs the release of interleukin-1, leading to downstream pro-inflammatory effects, and its potential for therapeutic targeting using currently available and future tools in our pharmacologic arsenal. In particular, we focus on the results of several large, recently concluded clinical trials including the Canakinumab Antiinflammatory Thrombosis Outcome Study, Colchicine Cardiovascular Outcomes Trial, and the Low-Dose Colchicine Study, examining the efficacy of direct inhibition of interleukin-1 with canakinumab or a multimodal approach to inhibiting the NLRP3 inflammasome using colchicine, as well as an overview of novel small molecule inhibitors that are still in development.
Collapse
Affiliation(s)
- Gregory Hemenway
- From the Department of Medicine, Temple University Hospital, Philadelphia, PA
| | - William H Frishman
- Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| |
Collapse
|
20
|
Inflammation as a mechanism and therapeutic target in peripheral artery disease. Can J Cardiol 2022; 38:588-600. [PMID: 35114347 DOI: 10.1016/j.cjca.2022.01.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/24/2022] Open
Abstract
Peripheral artery disease is one of three major clinical manifestations of atherosclerosis, the other two being coronary artery and cerebrovascular disease. Despite progress in surgery, antithrombotic therapy and therapies that modify conventional risk factors (lipid-, blood pressure-, and glucose-lowering interventions), patients with peripheral artery disease have unacceptably high risk of vascular complications. Additional strategies to reduce this residual risk are needed. The accumulated evidence that inflammation plays an important role in the pathogenesis of atherosclerosis has spurred recent efforts to evaluate anti-inflammatory agents as an additional therapeutic approach for atherothrombosis prevention and treatment. In this review, we examine the evidence supporting the role of inflammation in atherosclerosis, review recent trials evaluating anti-inflammatory approaches to reduce cardiovascular complications, and offer insights into the opportunities for novel anti-inflammatory strategies to reduce the burden of cardiovascular and limb complications in patients with peripheral artery disease.
Collapse
|
21
|
Olsen MB, Gregersen I, Sandanger Ø, Yang K, Sokolova M, Halvorsen BE, Gullestad L, Broch K, Aukrust P, Louwe MC. Targeting the Inflammasome in Cardiovascular Disease. JACC Basic Transl Sci 2022; 7:84-98. [PMID: 35128212 PMCID: PMC8807732 DOI: 10.1016/j.jacbts.2021.08.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/24/2021] [Accepted: 08/28/2021] [Indexed: 01/10/2023]
Abstract
The pathogenesis of cardiovascular disease (CVD) is complex and multifactorial, and inflammation plays a central role. Inflammasomes are multimeric protein complexes that are activated in a 2-step manner in response to infection or tissue damage. Upon activation the proinflammatory cytokines, interleukins-1β and -18 are released. In the last decade, the evidence that inflammasome activation plays an important role in CVD development became stronger. We discuss the role of different inflammasomes in the pathogenesis of CVD, focusing on atherosclerosis and heart failure. This review also provides an overview of existing experimental studies and clinical trials on inflammasome inhibition as a therapeutic target in these disorders.
Collapse
Key Words
- ACS, acute coronary syndrome
- AIM2, absent in melanoma 2
- ASC, apoptosis associated speck-like protein
- ATP, adenosine triphosphate
- CAD, coronary artery disease
- CRP, C-reactive protein
- CVD, cardiovascular disease
- DAMP, damage associated molecular pattern
- GSDMD, gasdermin-D
- GSDMD-NT, gasdermin-D N-terminal
- HF, heart failure
- HFpEF, HF with preserved ejection fraction
- HFrEF, HF with reduced ejection fraction
- IL, interleukin
- IL-1
- LDL, low-density lipoprotein
- LV, left ventricular
- LVEF, left ventricular ejection fraction
- MI, myocardial infarction
- NF-κB, nuclear factor κB
- NLR, NOD-like receptor
- NLRP3
- NLRP3, NOD-like receptor family pyrin domain containing 3
- NOD, nucleotide-binding oligomerization domain
- PRR, pattern recognition receptor
- STEMI, ST-elevation myocardial infarction
- TLR, toll-like receptor
- atherosclerosis
- cardiovascular disease
- heart failure
- inflammasome
Collapse
Affiliation(s)
- Maria Belland Olsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Øystein Sandanger
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Section of Dermatology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kuan Yang
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Marina Sokolova
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Bente E. Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Lars Gullestad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Cardiac Research Center, Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Cardiac Research Center, Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Mieke C. Louwe
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
22
|
Abstract
INTRODUCTION Gout is the most common form of inflammatory arthritis affecting millions of people around the world. Painful flares and tophaceous deposits can be debilitating, reducing quality of life among those affected and putting strain on health care systems. AREAS COVERED This review provides an overview of the treatment of gout for flare pain management and lowering serum urate. Firstline agents are discussed with emphasis on emerging evidence. Novel therapies are also covered. EXPERT OPINION Lifestyle modifications form a part of gout prevention. Regarding gout flare pharmacotherapy NSAIDs, colchicine and glucocorticoids are first line agents. The IL-1β antagonists also are highly effective for arresting flares but their cost-effectiveness render them as salvage therapies. Allopurinol is an agent of first choice for urate lowering therapy (ULT). In South East Asian and Black populations screening for HLA*B58:01 mutation is a cost-effective approach to decrease the occurrence of the rare but potentially very serious allopurinol hypersensitivity syndrome (AHS.). Febuxostat is another efficacious urate lowering therapy but it has received U.S. FDA black box warning for cardiovascular safety and careful consideration is warranted before its initiation in patients with high cardiovascular risk. Novel uricosurics are a class for continued drug development; verinurad and arhalofenate are agents with future promise. For patients with recalcitrant gout, pegloticase is another effective option in the rheumatologist's armamentarium. Its immunogenicity significantly threatens the achievement of sustained urate lowering responses. Abrogating pegloticase's immunogenicity with immunomodulatory co-therapy may lend to sustained efficacy.
Collapse
Affiliation(s)
- K E G Blake
- Clinical Fellow, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, 1720 2nd Ave South, FOT 839. Birmingham, AL 35294-3408
| | - Jordan L Saag
- Medical Student, University of Central Florida College of Medicine, 6850 Lake Nona Blvd. Orlando, FL 32827
| | - Kenneth G Saag
- Director, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, 1720 2nd Ave South, FOT 839. Birmingham, AL 35294-3408
| |
Collapse
|
23
|
Vilela EM, Fontes-Carvalho R. Inflammation and ischemic heart disease: The next therapeutic target? Rev Port Cardiol 2021; 40:785-796. [PMID: 34857118 DOI: 10.1016/j.repce.2021.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammation plays an important role in several stages of the cardiovascular continuum. In recent decades a plethora of studies have provided new data highlighting the role of inflammation in atherogenesis and atherothrombosis in two-way interactions with various cardiovascular risk factors and further influencing these dynamic processes. The concept of targeting residual inflammatory risk among individuals with ischemic heart disease (IHD) is therefore gaining increasing attention. Recently, several landmark randomized controlled trials have assessed different pharmacological approaches that may mitigate this residual risk. The results of some of these studies, such as CANTOS with canakinumab and COLCOT and LoDoCo2 with colchicine, are promising and have provided data to support this concept. Moreover, though several aspects remain to be clarified, these trials have shown the potential of modulating inflammation as a new target to reduce the risk of cardiovascular events in secondary prevention patients. In the present review, we aim to present a pragmatic overview of the complex interplay between inflammation and IHD, and to critically appraise the current evidence on this issue while presenting future perspectives on this topic of pivotal contemporary interest.
Collapse
Affiliation(s)
- Eduardo M Vilela
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal.
| | - Ricardo Fontes-Carvalho
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal; Cardiovascular Research Center (UniC), Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
24
|
D'Amario D, Cappetta D, Cappannoli L, Princi G, Migliaro S, Diana G, Chouchane K, Borovac JA, Restivo A, Arcudi A, De Angelis A, Vergallo R, Montone RA, Galli M, Liuzzo G, Crea F. Colchicine in ischemic heart disease: the good, the bad and the ugly. Clin Res Cardiol 2021; 110:1531-1542. [PMID: 33713178 PMCID: PMC8484100 DOI: 10.1007/s00392-021-01828-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 02/22/2021] [Indexed: 11/25/2022]
Abstract
Inflammation is the main pathophysiological process involved in atherosclerotic plaque formation, progression, instability, and healing during the evolution of coronary artery disease (CAD). The use of colchicine, a drug used for decades in non-ischemic cardiovascular (CV) diseases and/or systemic inflammatory conditions, stimulated new perspectives on its potential application in patients with CAD. Previous mechanistic and preclinical studies revealed anti-inflammatory and immunomodulatory effects of colchicine exerted through its principal mechanism of microtubule polymerization inhibition, however, other pleiotropic effects beneficial to the CV system were observed such as inhibition of platelet aggregation and suppression of endothelial proliferation. In randomized double-blinded clinical trials informing our clinical practice, low doses of colchicine were associated with the significant reduction of cardiovascular events in patients with stable CAD and chronic coronary syndrome (CCS) while in patients with a recent acute coronary syndrome (ACS), early initiation of colchicine treatment significantly reduced major adverse CV events (MACE). On the other hand, the safety profile of colchicine and its potential causal relationship to the observed increase in non-CV deaths warrants further investigation. For these reasons, postulates of precision medicine and patient-tailored approach with regards to benefits and harms of colchicine treatment should be employed at all times due to potential toxicity of colchicine as well as the currently unresolved signal of harm concerning non-CV mortality. The main goal of this review is to provide a balanced, critical, and comprehensive evaluation of currently available evidence with respect to colchicine use in the setting of CAD.
Collapse
Affiliation(s)
- Domenico D'Amario
- Dipartimento Di Scienze Cardiovascolari E Toraciche, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A.Gemelli 8, Rome, 00168, Italy
- Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, University of Campania L. Vanvitelli, Naples, 80138, Italy
| | | | | | | | - Giovanni Diana
- Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | | | - Josip A Borovac
- Department of Pathophysiology, School of Medicine, University of Split, Split, 21000, Croatia
| | | | | | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania L. Vanvitelli, Naples, 80138, Italy
| | - Rocco Vergallo
- Dipartimento Di Scienze Cardiovascolari E Toraciche, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A.Gemelli 8, Rome, 00168, Italy
- Università Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Rocco A Montone
- Dipartimento Di Scienze Cardiovascolari E Toraciche, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A.Gemelli 8, Rome, 00168, Italy
| | - Mattia Galli
- Università Cattolica del Sacro Cuore, Rome, 00168, Italy
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Giovanna Liuzzo
- Dipartimento Di Scienze Cardiovascolari E Toraciche, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A.Gemelli 8, Rome, 00168, Italy.
- Università Cattolica del Sacro Cuore, Rome, 00168, Italy.
| | - Filippo Crea
- Dipartimento Di Scienze Cardiovascolari E Toraciche, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A.Gemelli 8, Rome, 00168, Italy.
- Università Cattolica del Sacro Cuore, Rome, 00168, Italy.
| |
Collapse
|
25
|
Tennis MA, Smith AJ, Dwyer-Nield LD, Keith RL. Intranasal iloprost prevents tumors in a murine lung carcinogenesis model. Cancer Prev Res (Phila) 2021; 15:11-16. [PMID: 34556494 DOI: 10.1158/1940-6207.capr-21-0086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/12/2021] [Accepted: 09/09/2021] [Indexed: 11/16/2022]
Abstract
Lung cancer chemoprevention with the prostacyclin analogue iloprost is the most promising approach to date for intercepting progression of premalignant lung lesions in former smokers. Previous pre-clinical studies of iloprost used oral delivery, but a study modeling delivery directly to the target organ was needed. In vivo and in vitro studies have identified gene expression changes following iloprost treatment, including increased E-cadherin and PPAR𝛄 and decreased COX2 and Vimentin. We used tumor counts and gene expression to demonstrate the effectiveness of intranasal delivery of iloprost in a murine model of premalignant adenomas. Intranasal delivery of iloprost reduced adenoma multiplicity14 weeks after urethane exposure in FVB mice compared to untreated urethane controls. Intranasal iloprost reversed urethane-induced gene expression changes in tumors and whole lung. These results correspond to previous studies of oral iloprost and in vitro treatment of human bronchial epithelial cells. This study demonstrates that intranasal delivery of iloprost in a mouse model of lung premalignant lesions is effective chemoprevention. This will be an essential tool for exploring mechanisms and outcomes of iloprost chemoprevention, along with supporting ongoing clinical trials of inhaled iloprost chemoprevention.
Collapse
Affiliation(s)
- Meredith A Tennis
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus
| | - Alex J Smith
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus
| | | | - Robert L Keith
- Medicine/Division of Pulmonary Sciences, Eastern Colorado Veterans Affairs Medical Center
| |
Collapse
|
26
|
Vilela EM, Fontes-Carvalho R. Inflammation and ischemic heart disease: The next therapeutic target? Rev Port Cardiol 2021; 40:S0870-2551(21)00321-8. [PMID: 34456098 DOI: 10.1016/j.repc.2021.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/07/2021] [Accepted: 02/16/2021] [Indexed: 10/20/2022] Open
Abstract
Inflammation plays an important role in several stages of the cardiovascular continuum. In recent decades a plethora of studies have provided new data highlighting the role of inflammation in atherogenesis and atherothrombosis in two-way interactions with various cardiovascular risk factors and further influencing these dynamic processes. The concept of targeting residual inflammatory risk among individuals with ischemic heart disease (IHD) is therefore gaining increasing attention. Recently, several landmark randomized controlled trials have assessed different pharmacological approaches that may mitigate this residual risk. The results of some of these studies, such as CANTOS with canakinumab and COLCOT and LoDoCo2 with colchicine, are promising and have provided data to support this concept. Moreover, though several aspects remain to be clarified, these trials have shown the potential of modulating inflammation as a new target to reduce the risk of cardiovascular events in secondary prevention patients. In the present review, we aim to present a pragmatic overview of the complex interplay between inflammation and IHD, and to critically appraise the current evidence on this issue while presenting future perspectives on this topic of pivotal contemporary interest.
Collapse
Affiliation(s)
- Eduardo M Vilela
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal.
| | - Ricardo Fontes-Carvalho
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal; Cardiovascular Research Center (UniC), Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
27
|
Tabares-Guevara JH, Jaramillo JC, Ospina-Quintero L, Piedrahíta-Ochoa CA, García-Valencia N, Bautista-Erazo DE, Caro-Gómez E, Covián C, Retamal-Díaz A, Duarte LF, González PA, Bueno SM, Riedel CA, Kalergis AM, Ramírez-Pineda JR. IL-10-Dependent Amelioration of Chronic Inflammatory Disease by Microdose Subcutaneous Delivery of a Prototypic Immunoregulatory Small Molecule. Front Immunol 2021; 12:708955. [PMID: 34305950 PMCID: PMC8297659 DOI: 10.3389/fimmu.2021.708955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
One of the interventional strategies to reestablish the immune effector/regulatory balance, that is typically altered in chronic inflammatory diseases (CID), is the reinforcement of endogenous immunomodulatory pathways as the one triggered by interleukin (IL)-10. In a recent work, we demonstrated that the subcutaneous (sc) administration of an IL-10/Treg-inducing small molecule-based formulation, using a repetitive microdose (REMID) treatment strategy to preferentially direct the effects to the regional immune system, delays the progression of atherosclerosis. Here we investigated whether the same approach using other IL-10-inducing small molecule, such as the safe, inexpensive, and widely available polyphenol curcumin, could induce a similar protective effect in two different CID models. We found that, in apolipoprotein E deficient mice, sc treatment with curcumin following the REMID strategy induced atheroprotection that was not consequence of its direct systemic lipid-modifying or antioxidant activity, but instead paralleled immunomodulatory effects, such as reduced proatherogenic IFNγ/TNFα-producing cells and increased atheroprotective FOXP3+ Tregs and IL-10-producing dendritic and B cells. Remarkably, when a similar strategy was used in the neuroinflammatory model of experimental autoimmune encephalomyelitis (EAE), significant clinical and histopathological protective effects were evidenced, and these were related to an improved effector/regulatory cytokine balance in restimulated splenocytes. The essential role of curcumin-induced IL-10 for neuroprotection was confirmed by the complete abrogation of the clinical effects in IL-10-deficient mice. Finally, the translational therapeutic prospection of this strategy was evidenced by the neuroprotection observed in mice starting the treatment one week after disease triggering. Collectively, results demonstrate the power of a simple natural IL-10-inducing small molecule to tackle chronic inflammation, when its classical systemic and direct pharmacological view is shifted towards the targeting of regional immune cells, in order to rationally harness its immunopharmacological potential. This shift implies that many well-known IL-10-inducing small molecules could be easily reformulated and repurposed to develop safe, innovative, and accessible immune-based interventions for CID.
Collapse
Affiliation(s)
- Jorge H Tabares-Guevara
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Julio C Jaramillo
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Laura Ospina-Quintero
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Christian A Piedrahíta-Ochoa
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Natalia García-Valencia
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - David E Bautista-Erazo
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Erika Caro-Gómez
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Camila Covián
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angello Retamal-Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José R Ramírez-Pineda
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
28
|
Bosmans LA, Shami A, Atzler D, Weber C, Gonçalves I, Lutgens E. Glucocorticoid induced TNF receptor family-related protein (GITR) - A novel driver of atherosclerosis. Vascul Pharmacol 2021; 139:106884. [PMID: 34102305 DOI: 10.1016/j.vph.2021.106884] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is a lipid-driven, chronic inflammatory disease. In spite of efficient lipid lowering treatments, such as statins and PCSK9 inhibitors, patients, especially those with elevated inflammatory biomarkers, still have a significant residual cardiovascular disease risk. Novel drugs targeting inflammatory mediators are needed to further reduce this residual risk. Agonistic immune checkpoint proteins, including CD86, CD40L and CD40, have been shown to be drivers of atherosclerosis. Recently, glucocorticoid-induced tumour necrosis factor receptor family-related protein (GITR), a co-stimulatory immune checkpoint protein, was identified to be pivotal in cardiovascular disease. Cardiovascular patients have elevated soluble GITR plasma levels compared to healthy controls. Furthermore, in human carotid endarterectomy plaques, GITR expression was higher in plaques from symptomatic compared to asymptomatic patients and correlated with features of plaque vulnerability. Moreover, depleting GITR reduced atherosclerotic plaque development in mice. GITR-deficient monocytes and macrophages exhibited less inflammatory potential and reduced migratory capacity. In this review, we discuss GITR's effects on various immune cells, mechanisms, signalling pathways and finally GITR's potential as a novel drug target in atherosclerosis.
Collapse
Affiliation(s)
- Laura A Bosmans
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, the Netherlands
| | - Annelie Shami
- Department of Clinical Sciences Malmö, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Dorothee Atzler
- Institute of Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität (LMU Munich), Munich, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany; Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians Universität (LMU Munich), Munich, Germany
| | - Christian Weber
- Institute of Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität (LMU Munich), Munich, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, Maastricht, the Netherlands
| | - Isabel Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Clinical Research Centre, Malmö, Sweden; Department of Cardiology, Skåne University Hospital, Sweden
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, the Netherlands; Institute of Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität (LMU Munich), Munich, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
29
|
Reynolds MR, Gong T, Li S, Herzog CA, Charytan DM. Cost-Effectiveness of Coronary Artery Bypass Grafting and Percutaneous Coronary Intervention in Patients With Chronic Kidney Disease and Acute Coronary Syndromes in the US Medicare Program. J Am Heart Assoc 2021; 10:e019391. [PMID: 33787323 PMCID: PMC8174359 DOI: 10.1161/jaha.120.019391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/24/2021] [Indexed: 01/24/2023]
Abstract
Background Coronary revascularization provides important long-term clinical benefits to patients with high-risk presentations of coronary artery disease, including those with chronic kidney disease. The cost-effectiveness of coronary interventions in this setting is not known. Methods and Results We developed a Markov cohort simulation model to assess the cost-effectiveness of percutaneous coronary intervention (PCI) and coronary artery bypass grafting (CABG) in patients with chronic kidney disease who were hospitalized with acute myocardial infarction or unstable angina. Model inputs were primarily drawn from a sample of 14 300 patients identified using the Medicare 20% sample. Survival, quality-adjusted life-years, costs, and cost-effectiveness were projected over a 20-year time horizon. Multivariable models indicated higher 30-day mortality and end-stage renal disease with both PCI and CABG, and higher stroke with CABG, relative to medical therapy. However, the model projected long-term gains of 0.72 quality-adjusted life-years (0.97 life-years) for PCI compared with medical therapy, and 0.93 quality-adjusted life-years (1.32 life-years) for CABG compared with PCI. Incorporation of long-term costs resulted in incremental cost-effectiveness ratios of $65 326 per quality-adjusted life-year gained for PCI versus medical therapy, and $101 565 for CABG versus PCI. Results were robust to changes in input parameters but strongly influenced by the background costs of the population, and the time horizon. Conclusions For patients with chronic kidney disease and high-risk coronary artery disease presentations, PCI and CABG were both associated with markedly increased costs as well as gains in quality-adjusted life expectancy, with incremental cost-effectiveness ratios indicating intermediate value in health economic terms.
Collapse
Affiliation(s)
- Matthew R. Reynolds
- Lahey Hospital & Medical CenterBurlingtonMA
- Baim Institute for Clinical ResearchBostonMA
| | - Tingting Gong
- Chronic Disease Research GroupHennepin Healthcare Research InstituteMinneapolisMN
| | - Shuling Li
- Chronic Disease Research GroupHennepin Healthcare Research InstituteMinneapolisMN
| | - Charles A. Herzog
- Chronic Disease Research GroupHennepin Healthcare Research InstituteMinneapolisMN
- Department of MedicineHennepin Healthcare and University of MinnesotaMinneapolisMN
| | | |
Collapse
|
30
|
Thanigaimani S, Phie J, Krishna SM, Moxon J, Golledge J. Effect of disease modifying anti-rheumatic drugs on major cardiovascular events: a meta-analysis of randomized controlled trials. Sci Rep 2021; 11:6627. [PMID: 33758292 PMCID: PMC7987985 DOI: 10.1038/s41598-021-86128-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 03/05/2021] [Indexed: 02/08/2023] Open
Abstract
Disease modifying anti-rheumatic drugs (DMARDs) were developed to treat joint inflammation. There is growing evidence that anti-inflammatory drugs prevent major cardiovascular events (MACE). The aim of this systematic review and meta-analysis was to examine whether DMARDs reduce the risk of MACE. A systematic literature search was performed to identify randomized controlled trials (RCTs) testing the effect of DMARDs on cardiovascular events. The primary outcome was MACE defined as the first occurrence of non-fatal myocardial infarction (MI), non-fatal stroke or cardiovascular death. Secondary outcomes were myocardial infarction or stroke alone and all-cause mortality. Safety was assessed by fatal or life threatening infection. Meta-analyses were performed using random effect models and reported as risk ratios (RR) and 95% confidence intervals (CI). Study quality and publication bias were assessed using the Cochrane Collaboration’s tool for assessing risk of bias and funnel plots. Twelve RCTs involving 18,056 participants testing three different DMARDs subclasses (Tumor Necrosis Factor inhibitors—4 trials; Janus Kinase inhibitors—5 trials; Interleukin inhibitors—3 trials) were included. Meta-analysis suggested that none of the DMARD subclasses had any effect on MACE, MI alone, stroke alone, risk of fatal or life threatening infection or death. Risk of bias was high, low and unclear in five, six and one studies respectively. Funnel plots suggested a low possibility of publication bias. This meta-analysis suggests that DMARDs do not affect the incidence of MACE. More trials are needed for firm conclusions.
Collapse
Affiliation(s)
- Shivshankar Thanigaimani
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | - James Phie
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | - Smriti Murali Krishna
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | - Joseph Moxon
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | - Jonathan Golledge
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia. .,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia. .,The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, QLD, Australia.
| |
Collapse
|
31
|
Biasucci LM, Pedicino D, Liuzzo G. Promises and challenges of targeting inflammation to treat cardiovascular disease: the post-CANTOS era. Eur Heart J 2021; 41:2164-2167. [PMID: 31504422 DOI: 10.1093/eurheartj/ehz586] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Luigi M Biasucci
- Dipartimento di Scienze Cardiovascolari e Toraciche, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Daniela Pedicino
- Dipartimento di Scienze Cardiovascolari e Toraciche, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanna Liuzzo
- Dipartimento di Scienze Cardiovascolari e Toraciche, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
32
|
Alam A, Ansari MA, Badrealam KF, Pathak S. Molecular approaches to lung cancer prevention. Future Oncol 2021; 17:1793-1810. [PMID: 33653087 DOI: 10.2217/fon-2020-0789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Lung cancer is generally diagnosed at advanced stages when surgical resection is not possible. Late diagnosis, along with development of chemoresistance, results in high mortality. Preventive approaches, including smoking cessation, chemoprevention and early detection are needed to improve survival. Smoking cessation combined with low-dose computed tomography screening has modestly improved survival. Chemoprevention has also shown some promise. Despite these successes, most lung cancer cases remain undetected until advanced stages. Additional early detection strategies may further improve survival and treatment outcome. Molecular alterations taking place during lung carcinogenesis have the potential to be used in early detection via noninvasive methods and may also serve as biomarkers for success of chemopreventive approaches. This review focuses on the utilization of molecular biomarkers to increase the efficacy of various preventive approaches.
Collapse
Affiliation(s)
- Asrar Alam
- Department of Preventive Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Mohammad A Ansari
- Department of Epidemic Disease Research, Institute of Research & Medical Consultation, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Khan F Badrealam
- Cardiovascular & Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Sujata Pathak
- Department of Preventive Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW To review recent literature with relevance to the management of multimorbid patients with gout, i.e., gout medication repurposed for comorbidities and vice versa. RECENT FINDINGS Adding to the previous success of interleukin-1 inhibition, two trials on low-dose colchicine's role in cardiovascular disease (CVD) demonstrated potential benefits in patients with or without gout. In Colchicine Cardiovascular Outcomes Trial, a composite CVD endpoint was reduced by 23% among patients who had experienced a recent myocardial infarction. In Low-Dose Colchicine 2, the composite CVD endpoint was reduced 31% among those with stable coronary artery disease. Use of urate-lowering therapy (ULT) for renal protection in patients without gout produced null results. Allopurinol did not benefit the glomerular filtration rate in two trials (Controlled trial of slowing of Kidney Disease progression From the Inhibition of Xanthine oxidase and Preventing Early Renal Function Loss) among patients with chronic kidney disease (with or without hyperuricemia, but not gout). SGLT-2 inhibitors, a medication recommended for patients with diabetes and CVD, diabetic kidney disease, or heart failure, demonstrated a protective effect against gout flares in a secondary trial analysis and a large observational study. SUMMARY The role of colchicine may expand beyond gout flare prevention to patients with existing CVD. The renal benefit of ULT among patients with gout remains unclear. SGLT-2 inhibitors may benefit diabetic patients who have gout as a comorbidity.
Collapse
Affiliation(s)
- Kazuki Yoshida
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital
- Harvard Medical School
| | - Hyon K Choi
- Harvard Medical School
- Division of Rheumatology, Allergy & Immunology, Department of Medicine, Massachusetts General Hospital
| | - Daniel H Solomon
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital
- Harvard Medical School
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Wu P, Gao W, Su M, Nice EC, Zhang W, Lin J, Xie N. Adaptive Mechanisms of Tumor Therapy Resistance Driven by Tumor Microenvironment. Front Cell Dev Biol 2021; 9:641469. [PMID: 33732706 PMCID: PMC7957022 DOI: 10.3389/fcell.2021.641469] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/05/2021] [Indexed: 02/05/2023] Open
Abstract
Cancer is a disease which frequently has a poor prognosis. Although multiple therapeutic strategies have been developed for various cancers, including chemotherapy, radiotherapy, and immunotherapy, resistance to these treatments frequently impedes the clinical outcomes. Besides the active resistance driven by genetic and epigenetic alterations in tumor cells, the tumor microenvironment (TME) has also been reported to be a crucial regulator in tumorigenesis, progression, and resistance. Here, we propose that the adaptive mechanisms of tumor resistance are closely connected with the TME rather than depending on non-cell-autonomous changes in response to clinical treatment. Although the comprehensive understanding of adaptive mechanisms driven by the TME need further investigation to fully elucidate the mechanisms of tumor therapeutic resistance, many clinical treatments targeting the TME have been successful. In this review, we report on recent advances concerning the molecular events and important factors involved in the TME, particularly focusing on the contributions of the TME to adaptive resistance, and provide insights into potential therapeutic methods or translational medicine targeting the TME to overcome resistance to therapy in clinical treatment.
Collapse
Affiliation(s)
- Peijie Wu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wei Gao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Miao Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Wenhui Zhang
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jie Lin
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
35
|
Domen A, Quatannens D, Zanivan S, Deben C, Van Audenaerde J, Smits E, Wouters A, Lardon F, Roeyen G, Verhoeven Y, Janssens A, Vandamme T, van Dam P, Peeters M, Prenen H. Cancer-Associated Fibroblasts as a Common Orchestrator of Therapy Resistance in Lung and Pancreatic Cancer. Cancers (Basel) 2021; 13:987. [PMID: 33673405 PMCID: PMC7956441 DOI: 10.3390/cancers13050987] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer arises from mutations accruing within cancer cells, but the tumor microenvironment (TME) is believed to be a major, often neglected, factor involved in therapy resistance and disease progression. Cancer-associated fibroblasts (CAFs) are prominent and key components of the TME in most types of solid tumors. Extensive research over the past decade revealed their ability to modulate cancer metastasis, angiogenesis, tumor mechanics, immunosuppression, and drug access through synthesis and remodeling of the extracellular matrix and production of growth factors. Thus, they are considered to impede the response to current clinical cancer therapies. Therefore, targeting CAFs to counteract these protumorigenic effects, and overcome the resistance to current therapeutic options, is an appealing and emerging strategy. In this review, we discuss how CAFs affect prognosis and response to clinical therapy and provide an overview of novel therapies involving CAF-targeting agents in lung and pancreatic cancer.
Collapse
Affiliation(s)
- Andreas Domen
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
- Department of Oncology, Antwerp University Hospital (UZA), 2650 Edegem, Belgium
| | - Delphine Quatannens
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
| | - Sara Zanivan
- Cancer Research UK, Beatson Institute, Glasgow G611BD, UK;
- Institute of Cancer Sciences, University of Glasgow, Glasgow G611QH, UK
| | - Christophe Deben
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
| | - Jonas Van Audenaerde
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
| | - Evelien Smits
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
| | - An Wouters
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
| | - Filip Lardon
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
| | - Geert Roeyen
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
- Department of Hepatobiliary Transplantation and Endocrine Surgery, Antwerp University Hospital (UZA), 2650 Edegem, Belgium
| | - Yannick Verhoeven
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
| | - Annelies Janssens
- Department of Pulmonology & Thoracic Oncology, Antwerp University Hospital (UZA), 2650 Edegem, Belgium;
| | - Timon Vandamme
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
- Department of Oncology, Antwerp University Hospital (UZA), 2650 Edegem, Belgium
| | - Peter van Dam
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
- Gynaecologic Oncology Unit, Antwerp University Hospital (UZA), 2650 Edegem, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
- Department of Oncology, Antwerp University Hospital (UZA), 2650 Edegem, Belgium
| | - Hans Prenen
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, B2610 Antwerp, Belgium; (A.D.); (D.Q.); (C.D.); (J.V.A.); (E.S.); (A.W.); (F.L.); (G.R.); (Y.V.); (T.V.); (P.v.D.); (M.P.)
- Department of Oncology, Antwerp University Hospital (UZA), 2650 Edegem, Belgium
| |
Collapse
|
36
|
Ortega-Paz L, Capodanno D, Angiolillo DJ. Canakinumab for secondary prevention of coronary artery disease. Future Cardiol 2021; 17:427-442. [PMID: 33533289 DOI: 10.2217/fca-2020-0211] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease manifestations (CVD) are the world's leading cause of death, and their impact on morbidity requires effective prevention strategies of recurrent adverse events. For decades, inflammation has been proposed as a key promoter for atherosclerosis and its complications. However, studies on the use of drugs to target the excess inflammation in CVD are limited. In 2017, the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial confirmed the key role of inflammation on atherosclerotic disease. Canakinumab is a monoclonal antibody that blocks an inflammatory pathway mediated by IL-1β. The results of the CANTOS trial opened a new era of investigating new therapeutics targeting inflammation for CVD secondary prevention. This review presents the canakinumab's pharmacology, current clinical development status and regulatory perspectives.
Collapse
Affiliation(s)
- Luis Ortega-Paz
- Cardiovascular Institute, Hospital Clinic, IDIBAPS, Barcelona, 08036, Spain
| | - Davide Capodanno
- Division of Cardiology, A.O.U. 'Policlinico-Vittorio Emanuele', University of Catania, Catania, 95124, Italy
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL 32209, USA
| |
Collapse
|
37
|
Xia M, Yang X, Qian C. Meta-analysis Evaluating the Utility of Colchicine in Secondary Prevention of Coronary Artery Disease. Am J Cardiol 2021; 140:33-38. [PMID: 33137319 DOI: 10.1016/j.amjcard.2020.10.043] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 01/17/2023]
Abstract
Colchicine has shown potential therapeutic benefits in cardiovascular conditions owing to its broad anti-inflammatory properties. Here, we performed a meta-analysis to determine the efficacy and safety of colchicine in patients with coronary artery disease (CAD). A systematical search in electronic databases of PubMed, The Cochrane Library, and Scopus were carried out to identify eligible studies. Only randomized controlled trials evaluating the cardiovascular effects of colchicine in CAD patients were included. Study-level data of cardiovascular outcomes or adverse events were pooled using random-effect models. We finally included 5 randomized controlled trials with follow-up duration ≥6 months, comprising a total of 11,790 patients with CAD. Compared with placebo or no treatment, colchicine administration was associated with a significantly lower incidence of major adverse cardiovascular events (relative risk [RR] 0.65, 95% confidence interval [CI] 0.52 to 0.82). Such a benefit was not modified by the clinical phenotype of CAD (p for interaction = 0.34). Colchicine treatment also decreased the risk of myocardial infarction (RR 0.73, 95% CI 0.55 to 0.98), coronary revascularization (RR 0.61, 95% CI 0.42 to 0.89) and stroke (RR 0.47, 95% CI 0.28 to 0.81) in CAD patients, but with no impact on cardiovascular mortality. In addition, the rates of common adverse events were generally similar between colchicine and control groups, including noncardiovascular deaths (RR 1.50, 95% CI 0.93 to 2.40) and gastrointestinal symptoms (RR 1.05, 95% CI 0.91 to 1.22). In conclusion, the results of our meta-analysis demonstrated that colchicine treatment may reduce the risk of future cardiovascular events in CAD patients.
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW The pathogenesis and progression of coronary artery disease (CAD) is now known to be largely driven by inflammation on top of the well-accepted role for the disequilibrium between cholesterol deposition and removal from the arterial wall. Recent clinical trials have supported the inflammatory hypothesis of CAD and will be discussed in this review. RECENT FINDINGS The clinical trial Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) found that treatment with canakinumab, an anti-interleukin-1β agent, resulted in a reduction in non-fatal myocardial infarction, non-fatal stroke, or death. This provided evidence for the inflammatory hypothesis of CAD. However, canakinumab is not cost-effective for widespread therapy and more cost-effective treatments are warranted. The Cardiovascular Inflammation Reduction Trial (CIRT), Colchicine Cardiovascular Outcomes Trial (COLCOT), and Low-Dose Colchicine 2 (LoDoCo2) are recent clinical trials that increased the understanding of the inflammatory hypothesis of CAD. Cost-effective therapies targeting inflammation are the future of preventative CAD treatment. Additional clinical trials with anti-inflammatory and anti-cytokine agents would help delineate the most beneficial target for CAD prevention.
Collapse
Affiliation(s)
- Julia Boland
- Department of Internal Medicine, George Washington University Hospital, Washington, DC, USA.
| | - Carlin Long
- Department of Cardiology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
39
|
Bershtein LL. New Possibilities to Reduce the Residual Risk in Patients with Ischemic Heart Disease. KARDIOLOGIIA 2020; 60:1370. [PMID: 33487157 DOI: 10.18087/cardio.2020.11.n1370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 10/09/2020] [Indexed: 06/12/2023]
Abstract
Despite a significant progress of the recent decades, incidence of cardiovascular complications in patients with manifest, stable ischemic heart disease (IHD) is still high. Furthermore, this patient group is heterogenous; individuals with a higher risk of cardiovascular complications can be isolated from this group based on the presence of comorbidities and acute IHD on the background of the therapy. Such patients require a more aggressive treatment to influence major components of the increased risk. Even after administration of an optimum therapy, the risk for complications in such patients remains high (residual risk). The article discusses the lipid, inflammatory, and thrombotic components of residual risk in IHD patients and possibilities of their control with drugs with a special focus on possibilities of pharmaceutical correction of the risk thrombotic component in IHD patients with diabetes mellitus.
Collapse
Affiliation(s)
- L L Bershtein
- North Western State Medical Universtiy n.a. I.I. Mechnikov, St-Peterburg
| |
Collapse
|
40
|
Karády J, Mayrhofer T, Ivanov A, Foldyna B, Lu MT, Ferencik M, Pursnani A, Salerno M, Udelson JE, Mark DB, Douglas PS, Hoffmann U. Cost-effectiveness Analysis of Anatomic vs Functional Index Testing in Patients With Low-Risk Stable Chest Pain. JAMA Netw Open 2020; 3:e2028312. [PMID: 33315111 PMCID: PMC7737090 DOI: 10.1001/jamanetworkopen.2020.28312] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
IMPORTANCE Both noninvasive anatomic and functional testing strategies are now routinely used as initial workup in patients with low-risk stable chest pain (SCP). OBJECTIVE To determine whether anatomic approaches (ie, coronary computed tomography angiography [CTA] and coronary CTA supplemented with noninvasive fractional flow reserve [FFRCT], performed in patients with 30% to 69% stenosis) are cost-effective compared with functional testing for the assessment of low-risk SCP. DESIGN, SETTING, AND PARTICIPANTS This cost-effectiveness analysis used an individual-based Markov microsimulation model for low-risk SCP. The model was developed using patient data from the Prospective Multicenter Imaging Study for Evaluation of Chest Pain (PROMISE) trial. The model was validated by comparing model outcomes with outcomes observed in the PROMISE trial for anatomic (coronary CTA) and functional (stress testing) strategies, including diagnostic test results, referral to invasive coronary angiography (ICA), coronary revascularization, incident major adverse cardiovascular event (MACE), and costs during 60 days and 2 years. The validated model was used to determine whether anatomic approaches are cost-effective over a lifetime compared with functional testing. EXPOSURE Choice of index test for evaluation of low-risk SCP. MAIN OUTCOMES AND MEASURES Downstream ICA and coronary revascularization, MACE (death, nonfatal myocardial infarction), cost, quality-adjusted life-years (QALYs), and incremental cost-effectiveness ratio (ICER) of competing strategies. RESULTS The model cohort included 10 003 individual patients (median [interquartile range] age, 60.0 [54.4-65.9] years; 5270 [52.7%] women; 7693 [77.4%] White individuals), who entered the model 100 times. The Markov model accurately estimated the test assignment, results of anatomic and functional index testing, referral to ICA, revascularization, MACE, and costs at 60 days and 2 years compared with observed data in PROMISE (eg, coronary CTA: ICA, 12.2% [95% CI, 10.9%-13.5%] vs 12.3% [95% CI, 12.2%-12.4%]; revascularization, 6.2% [95% CI, 5.5%-6.9%] vs 6.4% [95% CI, 6.3%-6.5%]; functional strategy: ICA, 8.1% [95% CI, 7.4%-8.9%] vs 8.2% [95% CI, 8.1%-8.3%]; revascularization, 3.2% [95% CI, 2.7%-3.7%] vs 3.3% [95% CI, 3.2%-3.4%]; 2-year MACE rates: coronary CTA, 2.1% [95% CI, 1.7%-2.5%] vs 2.3% [95% CI, 2.2%-2.4%]; functional strategy, 2.2% [95% CI, 1.8%-2.6%] vs 2.4% [95% CI, 2.3%-2.4%]). Anatomic approaches led to higher ICA and revascularization rates at 60 days, 2 years, and 5 years compared with functional testing but were more effective in patient selection for ICA (eg, 60-day revascularization-to-ICA ratio, CTA: 53.7% [95% CI, 53.3%-54.0%]; CTA with FFRCT: 59.5% [95% CI, 59.2%-59.8%]; functional testing: 40.7% [95% CI, 40.4%-50.0%]). Over a lifetime, anatomic approaches gained an additional 6 months in perfect health compared with functional testing (CTA, 25.16 [95% CI, 25.14-25.19] QALYs; CTA with FFRCT, 25.14 [95% CI, 25.12-25.17] QALYs; functional testing, 24.68 [95% CI, 24.66-24.70] QALYs). Anatomic strategies were less costly and more effective; thus, CTA with FFRCT dominated and CTA alone was cost-effective (ICERs ranged from $1912/QALY for women and $3,559/QALY for men) compared with functional testing. In probabilistic sensitivity analyses, anatomic approaches were cost-effective in more than 65% of scenarios, assuming a willingness-to-pay threshold of $100 000/QALY. CONCLUSIONS AND RELEVANCE The results of this study suggest that anatomic strategies may present a more favorable initial diagnostic option in the evaluation of low-risk SCP compared with functional testing.
Collapse
Affiliation(s)
- Júlia Karády
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- MTA-SE Cardiovascular Imaging Research Group, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Alexander Ivanov
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Michael T. Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Maros Ferencik
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland
| | - Amit Pursnani
- Cardiology Division, Evanston Hospital, Evanston, Illinois
| | - Michael Salerno
- Departments of Medicine and Radiology, University of Virginia Health System, Charlottesville
| | - James E. Udelson
- Division of Cardiology, Tufts Medical Center, Boston, Massachusetts
| | - Daniel B. Mark
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Pamela S. Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Udo Hoffmann
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
41
|
Aimo A, Pascual-Figal DA, Barison A, Cediel G, Vicente ÁH, Saccaro LF, Emdin M, Bayes-Genis A. Colchicine for the Treatment of Coronary Artery Disease. Trends Cardiovasc Med 2020; 31:497-504. [PMID: 33096241 DOI: 10.1016/j.tcm.2020.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 10/23/2022]
Abstract
Inflammation plays an important role in atherosclerosis. Acute coronary syndromes (ACS), and particularly myocardial infarction (MI), are associated with a systemic inflammatory response that may accelerate coronary atherosclerotic processes, leading to plaque destabilization and increased risk of further cardiovascular events. These considerations provide a conceptual framework for the use of anti-inflammatory therapies in patients with chronic coronary syndrome or ACS. Following the diverging results of trials on canakinumab and methotrexate, the Colchicine Cardiovascular Outcomes Trial (COLCOT) and the Low-Dose Colchicine trial-2 (LoDoCo2) have sparked new interest in the perspective of an anti-inflammatory therapy for CAD by showing that colchicine confers a prognostic benefit in patients with a recent MI or CCS, respectively. Colchicine blocks multiple steps of the inflammatory cascade and modulates also platelet function and endothelial activation. It has a better safety profile than canakinumab and is a very inexpensive drug throughout the world. We deemed it useful to reappraise the available literature on colchicine and coronary artery disease to assess the likelihood that it might become part of the therapeutic armamentarium of this condition.
Collapse
Affiliation(s)
- Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, University Hospital of Pisa, Pisa, Italy.
| | - Domingo A Pascual-Figal
- Cardiology Department, Virgen de la Arrixaca Hospital and School of Medicine, University of Murcia, Murcia, Spain
| | - Andrea Barison
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Germán Cediel
- Hospital Universitari Germans Trias i Pujol, Badalona (Barcelona), Spain
| | - Álvaro Hernández Vicente
- Cardiology Department, Virgen de la Arrixaca Hospital and School of Medicine, University of Murcia, Murcia, Spain
| | - Luigi F Saccaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Antoni Bayes-Genis
- Hospital Universitari Germans Trias i Pujol, Badalona (Barcelona), Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
42
|
Poredos P, Gregoric ID, Jezovnik MK. Inflammation of carotid plaques and risk of cerebrovascular events. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1281. [PMID: 33178813 PMCID: PMC7607075 DOI: 10.21037/atm-2020-cass-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Carotid atherosclerotic plaques represent a risk for ischemic stroke. The data indicate that the risk for distal embolization from atherosclerotic lesions in internal carotid arteries is not related only to the degree of stenosis but also to the composition of plaques. The stability of atherosclerotic plaque depends on the thickness of the fibrous cap and plaque hemorrhage. Recent research indicated that the inflammatory activity of atherosclerotic lesions is pivotal in the progression of atherosclerotic plaques. It also promotes the development of unstable atherosclerotic lesions and is related to thromboembolic cerebrovascular complications. Inflammation destabilizes atherosclerotic plaques through the degradation of their fibrotic structure. Inflammation of atherosclerotic plaques was confirmed by histopathologic findings and levels of circulating inflammatory markers which were correlated to the intensity of the inflammation in atherosclerotic lesions. Recently, new techniques like fluorodeoxyglucose positron emission tomography (18-FDG PET) were developed for the identification of inflammation of atherosclerotic lesions in the vessel wall in vivo. Systemic inflammatory markers, particularly interleukins, tumor necrosis factor-alpha and metalloproteinases were shown to be related to the intensity of the inflammatory process in atherosclerotic lesions and the cerebrovascular events. Identification of inflamed atherosclerotic plaques may help to identify unstable atherosclerotic lesions and subjects at high risk for cerebrovascular incidents who need intensive preventive measures including anti-inflammatory medication.
Collapse
Affiliation(s)
- Pavel Poredos
- Department of Vascular Disease, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Department of Advanced Cardiopulmonary Therapies and Transplantation, The University of Texas Health Science Centre at Houston, Houston, TX, USA
| | - Igor D Gregoric
- Department of Advanced Cardiopulmonary Therapies and Transplantation, The University of Texas Health Science Centre at Houston, Houston, TX, USA
| | - Mateja K Jezovnik
- Department of Advanced Cardiopulmonary Therapies and Transplantation, The University of Texas Health Science Centre at Houston, Houston, TX, USA
| |
Collapse
|
43
|
Rosas G, Gaffo A, Rahn EJ, Saag KG. Cardiovascular safety risks associated with gout treatments. Expert Opin Drug Saf 2020; 19:1143-1154. [PMID: 32731769 DOI: 10.1080/14740338.2020.1804551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Uric acid is the final byproduct of purine metabolism. The loss of the enzyme that hydrolyzes uric acid to allantoin was lost, leading to a decrease in uric acid excretion and its further accumulation. The buildup of uric acid leads to damage in different organ systems, including the cardiovascular system. With the increasing burden of cardiovascular disease worldwide, a growing body of evidence has addressed the relationship between urate, cardiovascular outcomes, and gout medication cardiovascular safety. Areas covered: We discuss the most common gout therapies used for the reduction of serum urate and management of gout flares in different observational and clinical trials and their effects on different aspects of cardiovascular disease. We selected the most representative clinical studies that evaluated cardiovascular outcomes with each gout therapy as well as recommendation given by the most representative guidelines from Rheumatology societies for the management of gout. EXPERT OPINION The treatment of gout reduces joint damage and it can also lessen CV morbidity. Allopurinol shows CV safety profile when compared to other ULTs. Evidence supporting CV safety with the use of colchicine and IL-1 agents is promising and research needs to be conducted to further assess this outcome.
Collapse
Affiliation(s)
- Giovanna Rosas
- Department of Medicine, University of Alabama at Birmingham , Birmingham, AL, USA
| | - Angelo Gaffo
- Department of Medicine, University of Alabama at Birmingham , Birmingham, AL, USA
| | - Elizabeth J Rahn
- Department of Medicine, University of Alabama at Birmingham , Birmingham, AL, USA
| | - Kenneth G Saag
- Department of Medicine, University of Alabama at Birmingham , Birmingham, AL, USA
| |
Collapse
|
44
|
Liberale L, Montecucco F, Schwarz L, Lüscher TF, Camici GG. Inflammation and cardiovascular diseases: lessons from seminal clinical trials. Cardiovasc Res 2020; 117:411-422. [PMID: 32666079 DOI: 10.1093/cvr/cvaa211] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammation has been long regarded as a key contributor to atherosclerosis. Inflammatory cells and soluble mediators play critical roles throughout arterial plaque development and accordingly, targeting inflammatory pathways effectively reduces atherosclerotic burden in animal models of cardiovascular (CV) diseases. Yet, clinical translation often led to inconclusive or even contradictory results. The Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) followed by the Colchicine Cardiovascular Outcomes Trial (COLCOT) were the first two randomized clinical trials to convincingly demonstrate the effectiveness of specific anti-inflammatory treatments in the field of CV prevention, while other phase III trials-including the Cardiovascular Inflammation Reduction Trial one using methotrexate-were futile. This manuscript reviews the main characteristics and findings of recent anti-inflammatory Phase III trials in cardiology and discusses their similarities and differences in order to get further insights into the contribution of specific inflammatory pathways on CV outcomes. CANTOS and COLCOT demonstrated efficacy of two anti-inflammatory drugs (canakinumab and colchicine, respectively) in the secondary prevention of major adverse CV events (MACE) thus providing the first confirmation of the involvement of a specific inflammatory pathway in human atherosclerotic CV disease (ASCVD). Also, they highlighted the NOD-, LRR-, and pyrin domain-containing protein 3 inflammasome-related pathway as an effective therapeutic target to blunt ASCVD. In contrast, other trials interfering with a number of inflammasome-independent pathways failed to provide benefit. Lastly, all anti-inflammatory trials underscored the importance of balancing the risk of impaired host defence with an increase in infections and the prevention of MACE in CV patients with residual inflammatory risk.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland.,First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Lena Schwarz
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland.,Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8092 Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Rämistrasse 100, 8092, Zurich, Switzerland
| |
Collapse
|
45
|
Samuel M, Tardif JC, Khairy P, Roubille F, Waters DD, Grégoire JC, Pinto FJ, Maggioni AP, Diaz R, Berry C, Koenig W, Ostadal P, Lopez-Sendon J, Gamra H, Kiwan GS, Dubé MP, Provencher M, Orfanos A, Blondeau L, Kouz S, L'Allier PL, Ibrahim R, Bouabdallaoui N, Mitchell D, Guertin MC, Lelorier J. Cost-effectiveness of low-dose colchicine after myocardial infarction in the Colchicine Cardiovascular Outcomes Trial (COLCOT). EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2020; 7:486-495. [PMID: 32407460 PMCID: PMC8445085 DOI: 10.1093/ehjqcco/qcaa045] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022]
Abstract
Aims In the randomized, placebo-controlled Colchicine Cardiovascular Outcomes Trial (COLCOT) of 4745 patients enrolled within 30 days after myocardial infarction (MI), low-dose colchicine (0.5 mg once daily) reduced the incidence of the primary composite endpoint of cardiovascular death, resuscitated cardiac arrest, MI, stroke, or urgent hospitalization for angina leading to coronary revascularization. To assess the in-trial period and lifetime cost-effectiveness of low-dose colchicine therapy compared to placebo in post-MI patients on standard-of-care therapy. Methods and results A multistate Markov model was developed incorporating the primary efficacy and safety results from COLCOT, as well as healthcare costs and utilities from the Canadian healthcare system perspective. All components of the primary outcome, non-cardiovascular deaths, and pneumonia were included as health states in the model as both primary and recurrent events. In the main analysis, a deterministic approach was used to estimate the incremental cost-effectiveness ratio (ICER) for the trial period (24 months) and lifetime (20 years). Over the in-trial period, the addition of colchicine to post-MI standard-of-care treatment decreased the mean overall per-patient costs by 47%, from $502 to $265 Canadian dollar (CAD), and increased the quality-adjusted life years (QALYs) from 1.30 to 1.34. The lifetime per-patient costs were further reduced (69%) and QALYs increased with colchicine therapy (from 8.82 to 11.68). As a result, both in-trial and lifetime ICERs indicated colchicine therapy was a dominant strategy. Conclusion Cost-effectiveness analyses indicate that the addition of colchicine to standard-of-care therapy after MI is economically dominant and therefore generates cost savings.
Collapse
Affiliation(s)
- Michelle Samuel
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montréal, Québec H1T 1C8, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montréal, Québec H1T 1C8, Canada
| | - Paul Khairy
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montréal, Québec H1T 1C8, Canada
| | - François Roubille
- Université de Montpellier, INSERM, CNRS, CHU de Montpellier, Cardiology Department, CHU Arnaud de Villeneuve, 371, avenue du Doyen Gaston-Giraud, 34090 Montpellier, France
| | - David D Waters
- San Francisco General Hospital, Zuckerberg San Francisco General Hospital, 1001 Potrero Avenue, San Francisco, CA 94110, USA
| | - Jean C Grégoire
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montréal, Québec H1T 1C8, Canada
| | - Fausto J Pinto
- Santa Maria University Hospital (Centro Hospitalar Universitário Lisboa Norte), Centro Académico de Medicina de Lisboa, Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Aldo P Maggioni
- ANMCO Research Center, Via La Marmora 34, 50121 Firenze, Italy
| | - Rafael Diaz
- Estudios Clinicos Latinoamerica, Paraguay 160, 2000, Rosario, Argentina
| | - Colin Berry
- University of Glasgow and NHS Glasgow Clinical Research Facility, 126 University Pl, University of Glasgow, Glasgow, G12 8TA, Scotland, UK
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Lazarettstr. 36, D-80636 Munchen, Germany
| | - Petr Ostadal
- Cardiovascular Center, Na Homolce Hospital, Roentgenova 2, 150 00 Prague, Czech Republic
| | - Jose Lopez-Sendon
- H La Paz, IdiPaz, UAM, Ciber-CV Madrid, La Paz University Hospital, Paseo de la Castellana, 261, 28046 Madrid, Spain
| | - Habib Gamra
- Fattouma Bourguiba University Hospital, 5000 Monastir, Tunisia
| | - Ghassan S Kiwan
- Bellevue Medical Center, Qanater Zubayda- Mansouriyeh, Mansourieh, Metn District, Beirut, Lebanon
| | - Marie-Pierre Dubé
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montréal, Québec H1T 1C8, Canada
| | - Mylène Provencher
- The Montreal Health Innovations Coordinating Center, 4100 Molson St. Suite 400 Montreal, Quebec H1Y 3N1, Canada
| | - Andreas Orfanos
- The Montreal Health Innovations Coordinating Center, 4100 Molson St. Suite 400 Montreal, Quebec H1Y 3N1, Canada
| | - Lucie Blondeau
- The Montreal Health Innovations Coordinating Center, 4100 Molson St. Suite 400 Montreal, Quebec H1Y 3N1, Canada
| | - Simon Kouz
- Centre Hospitalier Régional de Lanaudière, 1000 Sainte-Anne Blvd Saint-Charles-Borromée, Quebec J6E 6J2, Canada
| | - Philippe L L'Allier
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montréal, Québec H1T 1C8, Canada
| | - Reda Ibrahim
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montréal, Québec H1T 1C8, Canada
| | - Nadia Bouabdallaoui
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montréal, Québec H1T 1C8, Canada
| | - Dominic Mitchell
- Logimetrix Inc., 3600 Rhodes Drive Windsor, Ontario N8W 5A4, Canada
| | - Marie-Claude Guertin
- The Montreal Health Innovations Coordinating Center, 4100 Molson St. Suite 400 Montreal, Quebec H1Y 3N1, Canada
| | - Jacques Lelorier
- Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montréal, Québec H1T 1C8, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal, 900 St Denis St Montreal, Quebec H2X 0A9, Canada
| |
Collapse
|
46
|
Ospina-Quintero L, Jaramillo JC, Tabares-Guevara JH, Ramírez-Pineda JR. Reformulating Small Molecules for Cardiovascular Disease Immune Intervention: Low-Dose Combined Vitamin D/Dexamethasone Promotes IL-10 Production and Atheroprotection in Dyslipidemic Mice. Front Immunol 2020; 11:743. [PMID: 32395119 PMCID: PMC7197409 DOI: 10.3389/fimmu.2020.00743] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022] Open
Abstract
The targeting of proinflammatory pathways has a prophylactic and therapeutic potential on atherosclerotic cardiovascular diseases (CVD). An alternative/complementary strategy is the promotion of endogenous atheroprotective mechanisms that are impaired during atherosclerosis progression, such as the activity of tolerogenic dendritic cells (tolDC) and regulatory T cells (Treg). There is a need to develop novel low cost, safe and effective tolDC/Treg-inducing formulations that are atheroprotective and that can be of easy translation into clinical settings. We found that apolipoprotein E-deficient (ApoE–/–) mice treated with a low-dose combined formulation of Vitamin D and Dexamethasone (VitD/Dexa), delivered repetitively and subcutaneously (sc) promoted interleukin-10 (IL-10) production by dendritic cells and other antigen presenting cells in the lymph nodes draining the site of injection and the spleens. Expectedly, the treatment also increased the numbers of IL-10-producing CD4+ T cells. Concomitantly, the frequency of IFNγ-producing CD4+ and CD8+ T cells in the spleen, and the IFNγ response of splenocytes to polyclonal stimulation ex vivo were lower after VitD/Dexa treatment, indicating a reduced proatherogenic Th1 response. Interestingly, VitD/Dexa-treated mice had smaller atherosclerotic lesions, with reduced lipid content and lower inflammatory infiltrate of macrophages and T cells in the aortic root. No hypolipidemic or antioxidant effect could be detected, suggesting that a dominantly immunomodulatory mechanism of atheroprotection was engaged under the low-dose sc VitD/Dexa conditions used. Finally, no evidence of clinical, biochemical or immune toxicity was observed in treated ApoE–/– mice and, most importantly, C57BL/6 mice latently infected with Leishmania parasites and treated with an identical VitD/Dexa dose/scheme showed no clinical or microbiological signs of disease reactivation, suggesting the absence of general immunosuppression. Altogether, these results indicate that a non-toxic, non-immunosuppressive, low-dose of VitD/Dexa, administered subcutaneously and repetitively, exerts atheroprotective effects in dyslipidemic mice, apparently due to the induction of an IL-10-producing network of lymphoid and myeloid immune cells. These well known, widely available, and inexpensive small molecules can be easily co-formulated into a simple and accessible agent with a potential use as a prophylactic or therapeutic immune intervention for CVD and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Laura Ospina-Quintero
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellin, Colombia
| | - Julio C Jaramillo
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellin, Colombia
| | - Jorge H Tabares-Guevara
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellin, Colombia
| | - José R Ramírez-Pineda
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellin, Colombia
| |
Collapse
|
47
|
Bosmans LA, Bosch L, Kusters PJH, Lutgens E, Seijkens TTP. The CD40-CD40L Dyad as Immunotherapeutic Target in Cardiovascular Disease. J Cardiovasc Transl Res 2020; 14:13-22. [PMID: 32222950 PMCID: PMC7892683 DOI: 10.1007/s12265-020-09994-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022]
Abstract
Chronic inflammation drives the development of atherosclerosis. Despite optimal treatment of classical cardiovascular risk factors, a substantial portion of the population has elevated inflammatory biomarkers and develops atherosclerosis-related complications, indicating that a residual inflammatory risk drives atherosclerotic cardiovascular disease in these patients. Additional anti-inflammatory therapeutic strategies are therefore required. The co-stimulatory molecule CD40 and its ligand CD40L (CD154) have a central role in the regulation of the inflammatory response during the development of atherosclerosis by modulating the interaction between immune cells and between immune cells and non-immune cells. In this review, we discuss the role of the CD40-CD40L dyad in atherosclerosis, and we discuss recent studies on the therapeutic potential of novel CD40-CD40L targeting strategies in cardiovascular medicine.
Collapse
Affiliation(s)
- Laura A Bosmans
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands
| | - Lena Bosch
- Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pascal J H Kusters
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.,Department of Pathology, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Tom T P Seijkens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
48
|
Abstract
Atherosclerosis, a chronic inflammatory disorder of the vasculature that results in cardiovascular disease, continues to pose a significant health and economic burden on modern society. Whilst inflammation has generally been accepted as the key driver of all stages of the disease, it was not until recently that inhibition of a specific proinflammatory cytokine (IL-1β) yielded successful results in the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study trial. This article offers a perspective on targeting inflammation for atherosclerosis, focusing on results of recent Phase III clinical trials, and discusses other potential candidates together with future challenges and prospects.
Collapse
|
49
|
Li Z, Hu S, Huang K, Su T, Cores J, Cheng K. Targeted anti-IL-1β platelet microparticles for cardiac detoxing and repair. SCIENCE ADVANCES 2020; 6:eaay0589. [PMID: 32076644 PMCID: PMC7002120 DOI: 10.1126/sciadv.aay0589] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 11/22/2019] [Indexed: 05/12/2023]
Abstract
An acute myocardial infarction (AMI) induces a sterile inflammatory response that facilitates further heart injury and promotes adverse cardiac remodeling. Interleukin-1β (IL-1β) plays a central role in the sterile inflammatory response that results from AMI. Thus, IL-1β blockage is a promising strategy for treatment of AMI. However, conventional IL-1β blockers lack targeting specificity. This increases the risk of serious side effects. To address this problem herein, we fabricated platelet microparticles (PMs) armed with anti-IL-1β antibodies to neutralize IL-1β after AMI and to prevent adverse cardiac remodeling. Our results indicate that the infarct-targeting PMs could bind to the injured heart, increasing the number of anti-IL-1β antibodies therein. The anti-IL-1β platelet PMs (IL1-PMs) protect the cardiomyocytes from apoptosis by neutralizing IL-1β and decreasing IL-1β-driven caspase-3 activity. Our findings indicate that IL1-PM is a promising cardiac detoxification agent that removes cytotoxic IL-1β during AMI and induces therapeutic cardiac repair.
Collapse
Affiliation(s)
- Zhenhua Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
| | - Teng Su
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Jhon Cores
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
50
|
Mattke S, Hanson M, Bentele M, Kandzari DE. Cost and Mortality Implications of Lower Event Rates After Implantation of an Ultrathin-Strut Coronary Stent Compared With a Thin-Strut Stent Over Four Years. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2020; 21:835-842. [PMID: 31954661 DOI: 10.1016/j.carrev.2019.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/13/2019] [Accepted: 12/13/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND The recent BIOFLOW V trial (ClinicalTrials.gov: NCT02389946) showed that revascularization with an ultrathin strut, bioresorbable polymer sirolimus-eluting stent (BP SES) was associated with lower rates of clinically driven target lesion revascularization (TLR) and target vessel-related myocardial infarction (TVMI) at 24-month follow-up than that with a thin strut, durable polymer everolimus-eluting stent (DP EES). We simulated the impact on cost and mortality. METHODS AND RESULTS We projected the impact of the lower adverse event rates from a U.S. health system perspective over a 48-month horizon with a Markov model using event data from the BIOFLOW V trial and estimates for costs and excess mortality due to adverse events from published sources. All cost estimates were CPI-adjusted to 2018 US$ and future cost discounted by 3%. We estimated that use of BP SES compared to DP EES was associated with cumulative net reductions in medical cost of $2429 per patient over 48 months. Peri-procedural TVMI contributed $124 (5%), TLR in patients without TVMI $810 (33%) and spontaneous TVMI $1496 (62%) of cost. Use of BP SES compared to DP EES was associated with 2603 fewer deaths in one million patients over four years, corresponding to a relative risk reduction of 6%. CONCLUSIONS Lower adverse event rates associated with revascularization using BP SES translate into reductions in direct medical cost and mortality. Most of the cost reduction is attributed to reduction in spontaneous TVMI. Given the high volume of coronary procedures, such results are an important consideration for patients, clinicians and payers.
Collapse
Affiliation(s)
- Soeren Mattke
- University of Southern California, Los Angeles, CA, USA; Benecit Research, Newton, MA, USA.
| | - Mark Hanson
- University of Southern California, Los Angeles, CA, USA; Benecit Research, Newton, MA, USA
| | | | | |
Collapse
|