101
|
Inflammation, Frailty and Cardiovascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1216:55-64. [PMID: 31894547 DOI: 10.1007/978-3-030-33330-0_7] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic inflammation, which is called "inflamm-aging" , is characterized by an increased level of inflammatory cytokines in response to physiological and environmental stressors, and causes the immune system to function consistently at a low level, even though it is not effective. Possible causes of inflammaging include genetic susceptibility, visceral obesity, changes in gut microbiota and permeability, chronic infections and cellular senescence. Inflammation has a role in the development of many age-related diseases, such as frailty. Low grade chronic inflammation can also increase the risk of atherosclerosis and insulin resistance which are the leading mechanisms in the development of cardiovascular diseases (CVD). As it is well known that the risk of CVD is higher in older people with frailty and the risk of frailty is higher in patients with CVD, there may be relationship between inflammation and the development of CVD and frailty. Therefore, this important issue will be discussed in this chapter.
Collapse
|
102
|
Giles JT, Sattar N, Gabriel S, Ridker PM, Gay S, Warne C, Musselman D, Brockwell L, Shittu E, Klearman M, Fleming TR. Cardiovascular Safety of Tocilizumab Versus Etanercept in Rheumatoid Arthritis: A Randomized Controlled Trial. Arthritis Rheumatol 2019; 72:31-40. [DOI: 10.1002/art.41095] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 08/27/2019] [Indexed: 01/30/2023]
Affiliation(s)
- Jon T. Giles
- Columbia University College of Physicians & Surgeons New York New York
| | | | - Sherine Gabriel
- Rutgers Robert Wood Johnson Medical School New Brunswick New Jersey
| | - Paul M. Ridker
- Brigham and Women’s HospitalHarvard Medical School Boston Massachusetts
| | - Steffen Gay
- University Hospital Zurich Zurich Switzerland
| | | | | | | | | | | | | |
Collapse
|
103
|
The Giants (biologicals) against the Pigmies (small molecules), pros and cons of two different approaches to the disease modifying treatment in rheumatoid arthritis. Autoimmun Rev 2019; 19:102421. [PMID: 31733368 DOI: 10.1016/j.autrev.2019.102421] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 06/13/2019] [Indexed: 02/08/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease that, if untreated, can lead to disability and reduce the life expectancy of affected patients. Over the last two decades the improvement of knowledge of the pathogenetic mechanisms leading to the development of the disease has profoundly changed the treatment strategies of RA through the development of biotechnological drugs (bDMARDs) directed towards specific pro-inflammatory targets involved in the RA network. To date, the therapeutic armamentarium for RA includes ten bDMARDs able to produce the depletion B-cells, the blockade of three different pro-inflammatory cytokines (tumour necrosis factor alpha, interleukin-6 and interleukin-1), or the inhibition of T-cell co-stimulation. The introduction of these new compounds has dramatically improved outcomes in the short and long term, although still a significant proportion of patients are unable to reach or maintain the treatment target over time. The identification of the fundamental role of Janus kinases in the process of transduction of the inflammatory signal within the immune cells has recently provided the opportunity to use the new pharmacological class of small molecules for the therapy of RA, further increasing the number of treatment options. In this review the PROS and CONS of these two drug classes will be discussed, trying to provide the evidence currently available to make the right choice based on the analysis of the efficacy and safety profile of the different drugs on the market and close to marketing.
Collapse
|
104
|
Martínez-Hervás S, González-Navarro H. Terapias antiinflamatorias para la enfermedad cardiovascular: vías de señalización y mecanismos. Rev Esp Cardiol 2019. [DOI: 10.1016/j.recesp.2019.02.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
105
|
Trott DW, Fadel PJ. Inflammation as a mediator of arterial ageing. Exp Physiol 2019; 104:1455-1471. [PMID: 31325339 DOI: 10.1113/ep087499] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022]
Abstract
NEW FINDINGS What is the topic of this review? This review summarizes and synthesizes what is known about the contribution of inflammation to age-related arterial dysfunction. What advances does it highlight? This review details observational evidence for the relationship of age-related inflammation and arterial dysfunction, insight from autoimmune inflammatory diseases and their effects on arterial function, interventional evidence linking inflammation and age-related arterial dysfunction, insight into age-related arterial inflammation from preclinical models and interventions to ameliorate age-related inflammation and arterial dysfunction. ABSTRACT Advanced age is a primary risk factor for cardiovascular disease, the leading cause of death in the industrialized world. Two major components of arterial ageing are stiffening of the large arteries and impaired endothelium-dependent dilatation in multiple vascular beds. These two alterations are major contributors to the development of overt cardiovascular disease. Increasing inflammation with advanced age is likely to play a role in this arterial dysfunction. The purpose of this review is to synthesize what is known about inflammation and its relationship to age-related arterial dysfunction. This review discusses both the initial observational evidence for the relationship of age-related inflammation and arterial dysfunction and the evidence that inflammatory autoimmune diseases are associated with a premature arterial ageing phenotype. We next discuss interventional and mechanistic evidence linking inflammation and age-related arterial dysfunction in older adults. We also attempt to summarize the relevant evidence from preclinical models. Lastly, we discuss interventions in both humans and animals that have been shown to ameliorate age-related arterial inflammation and dysfunction. The available evidence provides a strong basis for the role of inflammation in both large artery stiffening and impairment of endothelium-dependent dilatation; however, the specific inflammatory mediators, the initiating factors and the relative importance of the endothelium, smooth muscle cells, perivascular adipose tissue and immune cells in arterial inflammation are not well understood. With the expansion of the ageing population, ameliorating age-related arterial inflammation represents an important potential strategy for preserving vascular health in the elderly.
Collapse
Affiliation(s)
- Daniel W Trott
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| | - Paul J Fadel
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
106
|
Charles-Schoeman C, DeMasi R, Valdez H, Soma K, Hwang LJ, Boy MG, Biswas P, McInnes IB. Risk Factors for Major Adverse Cardiovascular Events in Phase III and Long-Term Extension Studies of Tofacitinib in Patients With Rheumatoid Arthritis. Arthritis Rheumatol 2019; 71:1450-1459. [PMID: 31385441 PMCID: PMC6754249 DOI: 10.1002/art.40911] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 04/11/2019] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Tofacitinib is an oral JAK inhibitor for the treatment of rheumatoid arthritis (RA). This study was undertaken to evaluate the risk of major adverse cardiovascular events (MACE) in patients with RA receiving tofacitinib. METHODS Data were pooled from patients with moderately to severely active RA receiving ≥1 tofacitinib dose in 6 phase III and 2 long-term extension studies over 7 years. MACE (myocardial infarction, stroke, cardiovascular death) were independently adjudicated. Cox regression models were used to evaluate associations between baseline variables and time to first MACE. Following 24 weeks of tofacitinib, changes in variables and time to future MACE were evaluated after adjusment for age, baseline values, and time-varying tofacitinib dose. Hazard ratios and 95% confidence intervals were calculated. RESULTS Fifty-two MACE occurred in 4,076 patients over 12,873 patient-years of exposure (incidence rate 0.4 patients with events per 100 patient-years). In univariable analyses of baseline variables, traditional cardiovascular risk factors and glucocorticoid and statin use were associated with MACE risk; disease activity and inflammation measures were not. In subsequent multivariable analyses, baseline age, hypertension, and the total cholesterol to high-density lipoprotein (HDL) cholesterol ratio remained significantly associated with risk of MACE. After 24 weeks of treatment, an increase in HDL cholesterol and a decrease in the total to HDL cholesterol were associated with decreased MACE risk; changes in total cholesterol, low-density lipoprotein (LDL) cholesterol, and disease activity measures were not. Increased erythrocyte sedimentation rates trended with increased future MACE risk. CONCLUSION In this post hoc analysis, after 24 weeks of tofacitinib treatment, increased HDL cholesterol, but not increased LDL cholesterol or total cholesterol, appeared to be associated with lower future MACE risk. Further data are needed to test the cardiovascular safety of tofacitinib.
Collapse
|
107
|
Mackey RH, Kuller LH, Moreland LW. Inflammatory joint diseases and atherosclerosis: time to look beyond the 'lipid paradox'. Curr Opin Lipidol 2019; 30:342-349. [PMID: 31145122 DOI: 10.1097/mol.0000000000000620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Uncertainty persists about the contribution of lipids to the increased risk of cardiovascular disease (CVD) among rheumatoid arthritis and other inflammatory joint disease (IJD) patients. In reviewing recent research, we consider potential insights gained by quantifying lipoprotein particles directly, rather than by their lipid content. RECENT FINDINGS Although inflammation often decreases LDL cholesterol (LDL-C), and anti-inflammatory medications often increase LDL-C, both inflammation and anti-inflammatory medications can increase atherogenic Apolipoprotein B (ApoB)-containing lipoprotein particles, attenuated by statins. CVD risk factors, that is, smoking, obesity, ApoB, may increase years prior to IJD diagnosis. Increased risks of nonatherosclerotic myocardial and pulmonary disease, heart failure and mortality may be directly related to disease activity, inflammation, and possibly to HDL particles and function. SUMMARY For IJD patients, higher cumulative lifetime exposure to CVD risk factors accelerates atherosclerosis and subsequent CVD risk that is underestimated by current risk factor levels. CVD risk reduction in IJD requires aggressive and earlier reduction in CVD risk factors (ApoB lipoproteins, smoking, hypertension, diabetes, lack of physical activity), in addition to control of disease activity and inflammation. Lipid-lowering medications can attenuate anti-inflammatory medication-induced increases in ApoB and LDL-C, but can also reduce CVD risk due to cumulative lifetime exposure.
Collapse
Affiliation(s)
- Rachel H Mackey
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh
| | - Lewis H Kuller
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh
| | - Larry W Moreland
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
108
|
Castagné B, Viprey M, Martin J, Schott AM, Cucherat M, Soubrier M. Cardiovascular safety of tocilizumab: A systematic review and network meta-analysis. PLoS One 2019; 14:e0220178. [PMID: 31369575 PMCID: PMC6675055 DOI: 10.1371/journal.pone.0220178] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 07/10/2019] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Our objective was to compare the cardiovascular safety of tocilizumab and other biological disease-modifying antirheumatic drugs (bDMARD) in rheumatoid arthritis using a network meta-analysis (NMA). METHODS A systematic literature search through May 2018 identified randomized controlled trials (RCT) or observational studies (cohort only) reporting cardiovascular outcomes of tocilizumab (TCZ) and/or abatacept (ABA) and/or rituximab (RTX) and/or tumor necrosis factor inhibitors (TNFi) in rheumatoid arthritis patients. The composite primary outcome was the rate of major adverse cardiovascular outcomes (MACE, myocardial infarction (MI), peripheral artery disease (PAD) and cardiac heart failure (CHF)). RESULTS 19 studies were included in the NMA, including 11 RCTs and 8 cohort studies. We found less events with RTX (5.41 [1.70;17.26]. We found no difference between TCZ and other treatments. Concerning MI, we found no difference between TCZ and csDMARD (4.23 [0.22;80.64]), no difference between TCZ and TNFi (2.00 [0.18;21.84]). There was no difference between TCZ and csDMARD (1.51[0.02;103.50] and between TCZ and TNFi (1.00 [0.06;15.85]) for stroke event. With cohorts and RCT NMA, we found no difference between TCZ and other treatments for MACE (0.66 [0.42;1.03] with ABA, 1.04 [0.60;1.81] with RTX, 0.78[0.53;1.16] and 0.91 [0.54;1.51] with csDMARD), but the risk of myocardial infarction was lower with TCZ compared to ABA (0.67 [0.47;0.97]). We lacked data to compare TCZ and other bDMARD for stoke and MI. Not enough data was available to perform a NMA for CHF and PAD. CONCLUSIONS Despite an increase in cholesterol levels, TCZ has safe cardiovascular outcomes compared to other bDMARD.
Collapse
Affiliation(s)
- Benjamin Castagné
- Rheumatology Department, Gabriel-Montpied University Hospital, Clermont-Ferrand, France
- HESPER EA 7425, University of Lyon, Claude Bernard University Lyon 1, Lyon, France
| | - Marie Viprey
- HESPER EA 7425, University of Lyon, Claude Bernard University Lyon 1, Lyon, France
- Public Health Centre, Hospices Civils de Lyon, Lyon, France
| | - Julie Martin
- HESPER EA 7425, University of Lyon, Claude Bernard University Lyon 1, Lyon, France
- Public Health Centre, Hospices Civils de Lyon, Lyon, France
| | - Anne-Marie Schott
- HESPER EA 7425, University of Lyon, Claude Bernard University Lyon 1, Lyon, France
- Public Health Centre, Hospices Civils de Lyon, Lyon, France
| | - Michel Cucherat
- University Lyon, UMR 5558, Laboratory of Biometry and Evolutionary Biology, CNRS, Villeurbanne, France
- Department of Pharmacology and Toxicology, Hospices Civils de Lyon, Lyon, France
| | - Martin Soubrier
- Rheumatology Department, Gabriel-Montpied University Hospital, Clermont-Ferrand, France
| |
Collapse
|
109
|
Giles JT, Wasko MCM, Chung CP, Szklo M, Blumenthal RS, Kao A, Bokhari S, Zartoshti A, Stein CM, Bathon JM. Exploring the Lipid Paradox Theory in Rheumatoid Arthritis: Associations of Low Circulating Low-Density Lipoprotein Concentration With Subclinical Coronary Atherosclerosis. Arthritis Rheumatol 2019; 71:1426-1436. [PMID: 30883031 DOI: 10.1002/art.40889] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 03/12/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) patients with the lowest circulating low-density lipoprotein (LDL) concentrations are at heightened risk of cardiovascular events. However, the atherosclerosis burden within this subgroup is unknown. METHODS RA patients pooled from 4 cohort studies of cardiovascular disease (CVD; n = 546) were compared with non-RA controls from the Multi-Ethnic Study of Atherosclerosis (n = 5,279). Those taking lipid-lowering medications were excluded. Differences in cardiac computed tomography-derived Agatston coronary artery calcium (CAC) scores between the RA and control groups were compared across strata of LDL concentration. RESULTS Among those with low LDL concentrations (<70 mg/dl), mean adjusted CAC scores were >4-fold higher for RA patients than for controls (18.6 versus 4.6 Agatston units, respectively; P < 0.001), a difference significantly greater than that in any other LDL concentration stratum except LDL concentration ≥160 mg/dl. Similarly, 32% of the RA patients with low LDL concentration had a CAC score of ≥100 Agatston units compared with only 7% of controls in the same LDL concentration stratum (odds ratio 5.97; P < 0.001), a difference significantly greater than that in all of the other LDL concentration strata. Low LDL concentration was most strongly associated with higher CAC score among RA patients who were white, had ever smoked, or were not obese. Other than a higher frequency of current smokers, RA patients with low LDL concentrations did not have more CVD risk factors or higher measures of RA disease activity or severity than RA patients with higher LDL concentrations. CONCLUSION RA patients with low LDL concentration may represent a group for whom heightened screening and prevention of atherosclerotic CVD is appropriate.
Collapse
Affiliation(s)
| | - Mary Chester M Wasko
- Allegheny Health Network, Temple University School of Medicine, Pittsburgh, Pennsylvania
| | | | | | | | - Amy Kao
- EMD Serono Research & Development Institute, Inc., Billerica, Maryland
| | | | | | | | | |
Collapse
|
110
|
Virone A, Bastard JP, Fellahi S, Capeau J, Rouanet S, Sibilia J, Ravaud P, Berenbaum F, Gottenberg JE, Sellam J. Comparative effect of tumour necrosis factor inhibitors versus other biological agents on cardiovascular risk-associated biomarkers in patients with rheumatoid arthritis. RMD Open 2019; 5:e000897. [PMID: 31413865 PMCID: PMC6667971 DOI: 10.1136/rmdopen-2019-000897] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/28/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022] Open
Abstract
Background To comparatively investigate the differential effect of second-line tumour necrosis factor inhibitors (TNFis) versus other biological agents on cardiovascular disease (CVD) risk-associated biomarkers in patients with rheumatoid arthritis (RA). Methods We evaluated the serum levels of lipoprotein-associated apoproteins ApoA1 and ApoB100 and lipoprotein(a) (Lp(a)) and the leptin/adiponectin ratio (LAR) as an insulin resistance proxy in patients with RA from the Rotation Or Change (ROC) trial treated with either a second-line TNFi or another biologic (tocilizumab (TCZ), rituximab or abatacept) at baseline and week 24. We compared the changes in biomarker levels in each group and according to the EULAR response. Results Of the 300 patients enrolled in the ROC trial, 203 were included in the study, including 96 in the second-line TNFi group and 107 in the other biological group. The measured biomarkers did not deteriorate between baseline and week 24 regardless of the group. A greater improvement in the LAR was noted in the other biological group (median (IQR) -0.12 ng/µg (-0.58 to 0.31) vs 0.04 (-0.19 to 0.43), p=0.033), and a greater improvement in the Lp(a) level was observed following treatment with TCZ than with a TNFi (-0.05 g/L (-0.11 to -0.01) vs -0.01 g/L (-0.02 to 0.01), p<0.001). When considering the patients' responses to treatment, improved biomarkers were mainly observed in the EULAR responders in each treatment group. Conclusions TNFis and non-TNFis were neutral on improved CVD risk-associated biomarkers in patients with RA insufficiently controlled by TNFis. TCZ could be associated with a better improvement concerning Lp(a) and LAR than TNFis. This improvement could be related to a good therapeutic response, thereby supporting the need of good control of RA. Trial registration number ClinicalTrials.gov Identifier NCT01000441, registered on 22 October 2009.
Collapse
Affiliation(s)
- Alexandre Virone
- Rheumatology Department, Hôpital Saint-Antoine, Assistance Publique - Hopitaux de Paris (AP-HP), Paris, France
| | - Jean-Philippe Bastard
- Biochemistry Department, Hôpital Tenon, AP-HP, Paris, France.,Faculty of Medicine Sorbonne Université, CRSA INSERM UMRS_938, Paris, France
| | - Soraya Fellahi
- Biochemistry Department, Hôpital Tenon, AP-HP, Paris, France.,Faculty of Medicine Sorbonne Université, CRSA INSERM UMRS_938, Paris, France
| | - Jacqueline Capeau
- Faculty of Medicine Sorbonne Université, CRSA INSERM UMRS_938, Paris, France
| | | | - Jean Sibilia
- Rheumatology Department, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Philippe Ravaud
- Epidemiology and Statistics Research Centre, Inserm UMR 1153, Paris Descartes University, Paris, France
| | - Francis Berenbaum
- Rheumatology Department, Hôpital Saint-Antoine, Assistance Publique - Hopitaux de Paris (AP-HP), Paris, France.,Faculty of Medicine Sorbonne Université, CRSA INSERM UMRS_938, Paris, France
| | | | - Jérémie Sellam
- Rheumatology Department, Hôpital Saint-Antoine, Assistance Publique - Hopitaux de Paris (AP-HP), Paris, France.,Faculty of Medicine Sorbonne Université, CRSA INSERM UMRS_938, Paris, France
| |
Collapse
|
111
|
Cardiometabolic comorbidities in RA and PsA: lessons learned and future directions. Nat Rev Rheumatol 2019; 15:461-474. [PMID: 31292564 DOI: 10.1038/s41584-019-0256-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2019] [Indexed: 02/07/2023]
Abstract
Cardiometabolic comorbidities present a considerable burden for patients with rheumatoid arthritis (RA) or psoriatic arthritis (PsA). Both RA and PsA are associated with an increased risk of cardiovascular disease (CVD). PsA more often exhibits an increased risk of metabolically linked comorbidities such as obesity, insulin resistance, type 2 diabetes mellitus and non-alcoholic fatty liver disease. Although both RA and PsA are characterized by a state of chronic inflammation, the mechanisms that contribute to CVD risk in these conditions might not be identical. In RA, systemic inflammation is thought to directly contribute to CVD risk, whereas in PsA, adiposity is thought to contribute to a notable metabolic phenotype that, in turn, contributes to CVD risk. Hence, appropriate management strategies that consider the increased risk of cardiometabolic comorbidities in patients with inflammatory arthropathy are important. In RA, such strategies should focus on the prediction of CVD risk and its management through targeting chronic inflammation and traditional CVD risk factors. In PsA, management strategies should additionally focus on targeting metabolic components, including weight management, which might not only help improve disease activity in the joints, entheses and skin, but also reduce the risk of metabolic comorbidities and improve the quality of life of patients.
Collapse
|
112
|
Marchio P, Guerra-Ojeda S, Vila JM, Aldasoro M, Victor VM, Mauricio MD. Targeting Early Atherosclerosis: A Focus on Oxidative Stress and Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8563845. [PMID: 31354915 PMCID: PMC6636482 DOI: 10.1155/2019/8563845] [Citation(s) in RCA: 397] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/10/2019] [Accepted: 05/19/2019] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a chronic vascular inflammatory disease associated to oxidative stress and endothelial dysfunction. Oxidation of low-density lipoprotein (LDL) cholesterol is one of the key factors for the development of atherosclerosis. Nonoxidized LDL have a low affinity for macrophages, so they are not themselves a risk factor. However, lowering LDL levels is a common clinical practice to reduce oxidation and the risk of major events in patients with cardiovascular diseases (CVD). Atherosclerosis starts with dysfunctional changes in the endothelium induced by disturbed shear stress which can lead to endothelial and platelet activation, adhesion of monocytes on the activated endothelium, and differentiation into proinflammatory macrophages, which increase the uptake of oxidized LDL (oxLDL) and turn into foam cells, exacerbating the inflammatory signalling. The atherosclerotic process is accelerated by a myriad of factors, such as the release of inflammatory chemokines and cytokines, the generation of reactive oxygen species (ROS), growth factors, and the proliferation of vascular smooth muscle cells. Inflammation and immunity are key factors for the development and complications of atherosclerosis, and therefore, the whole atherosclerotic process is a target for diagnosis and treatment. In this review, we focus on early stages of the disease and we address both biomarkers and therapeutic approaches currently available and under research.
Collapse
Affiliation(s)
- Patricia Marchio
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Sol Guerra-Ojeda
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - José M. Vila
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Martín Aldasoro
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Victor M. Victor
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Maria D. Mauricio
- Department of Physiology, Faculty of Medicine and Odontology, Universitat de Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| |
Collapse
|
113
|
McMahon M, Skaggs B, Grossman J, Wong WK, Sahakian L, Chen W, Hahn B. Comparison of PREDICTS atherosclerosis biomarker changes after initiation of new treatments in patients with SLE. Lupus Sci Med 2019; 6:e000321. [PMID: 31321062 PMCID: PMC6606066 DOI: 10.1136/lupus-2019-000321] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/21/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022]
Abstract
Objective Patients with SLE have an increased risk of atherosclerosis (ATH) that is not adequately explained by traditional risk factors. We previously described the Predictors of Risk for Elevated Flares, Damage Progression, and Increased Cardiovascular disease in PaTients with SLE (PREDICTS) atherosclerosis-risk panel, which includes proinflammatory HDL (piHDL), leptin, soluble tumour necrosis factor-like weak inducer of apoptosis (sTWEAK) and homocysteine, as well as age and diabetes. A high PREDICTS score confers 28-fold increased odds for future atherosclerosis in SLE. The aim of this study is to determine whether PREDICTS biomarkers are modifiable by common lupus therapies. Methods This prospective observational study included SLE subjects started on new lupus treatments. Leptin, sTWEAK, homocysteine and antioxidant function of HDL were measured at baseline (prior to drug initiation), 6 weeks and 12 weeks. Results 16 subjects started mycophenolate (MMF), 18 azathioprine (AZA) and 25 hydroxychloroquine (HCQ). In MMF-treated subjects, HDL function progressively improved from 2.23 ± 1.32 at baseline to 1.37±0.81 at 6 weeks (p=0.02) and 0.93±0.54 at 12 weeks (p=0.009). sTWEAK levels also improved in MMF-treated subjects from 477.5±447.1 to 290.3±204.6 pg/mL after 12 weeks (p=0.04), but leptin and homocysteine levels were not significantly changed. In HCQ-treated subjects, only HDL function improved from 1.80±1.29 at baseline to 1.03±0.74 after 12 weeks (p=0.05). There were no changes in the AZA group. MMF treatment was still associated with significant improvements in HDL function after accounting for potential confounders such as total prednisone dose and changes in disease activity. Overall, the mean number of high-risk PREDICTS biomarkers at week 12 significantly decreased in the entire group of patients started on a new lupus therapy (2.1±0.9 to 1.8±0.9, p=0.02) and in the MMF-treated group (2.4±0.8 vs 1.8±0.9, p=0.003), but not in the AZA or HCQ groups. In multivariate analysis, the odds of having a high PREDICTS atherosclerosis risk score at 12 weeks were lower with MMF treatment (OR 0.002, 95% CI 0.000 to 0.55, p=0.03). Conclusions 12 weeks of MMF therapy improves the overall PREDICTS atherosclerosis biomarker profile. Further studies will determine whether biomarker changes reflect decreases in future cardiovascular events.
Collapse
Affiliation(s)
- Maureen McMahon
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Brian Skaggs
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Jennifer Grossman
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Weng Kee Wong
- Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California, USA
| | - Lori Sahakian
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Weiling Chen
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Bevra Hahn
- Internal Medicine, Division Rheumatology, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
114
|
Anti-inflammatory Therapies for Cardiovascular Disease: Signaling Pathways and Mechanisms. ACTA ACUST UNITED AC 2019; 72:767-773. [PMID: 31155366 DOI: 10.1016/j.rec.2019.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/15/2019] [Indexed: 12/23/2022]
Abstract
Cardiovascular diseases (CVD) are the clinical manifestation of atherosclerosis, a chronic inflammatory disease promoted by several risk factors such as dyslipidemia, type 2 diabetes mellitus, hypertension, and smoking. Acute CVD events are the result of an unresolved inflammatory chronic state that promotes the rupture of unstable plaque lesions. Of note, the existing intensive therapies modify risk factors but do not prevent life-threatening recurrent ischemic events in high-risk patients, who have a residual inflammatory risk displayed by increased C-reactive protein (CRP) levels. Better understanding of the role of innate and adaptive immunity in plaque development and rupture has led to intensive investigation of anti-inflammatory strategies for CVD. Some of them are being tested in specific clinical trials and use lower doses of existing medications originally developed for other inflammatory diseases such as rheumatoid arthritis and psoriasis, which have high CVD risk. Other investigations are retrospective and meta-analyses of existing clinical trials that evaluate the incidence of CVD in these inflammatory diseases. Others are based on preclinical testing such as vaccines. In this article, we summarize the main anti-inflammatory strategies and associated molecular mechanisms that are being evaluated in preclinical or clinical CVD studies.
Collapse
|
115
|
Rheumatoid arthritis (RA) and cardiovascular disease. Autoimmun Rev 2019; 18:679-690. [PMID: 31059840 DOI: 10.1016/j.autrev.2019.05.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 01/27/2019] [Indexed: 12/13/2022]
Abstract
Patients with rheumatoid arthritis (RA) suffer cardiovascular events 1.5-2 fold than the general population, and cardiovascular (CV) events are leading cause of death in patients with RA. It is known that patients with RA have endothelial dysfunction, related with impaired function of endothelial progenitor cells (EPCs). The mechanistic pathways leading to endothelial function are complicated, but understanding these mechanisms may open new frontiers of management and therapies to patients suffering from atherosclerosis. Inflammation is a key factor in atherosclerosis, including endothelial function, plaque stabilization and post infarct remodeling; thus, inhibition of TNF-α may affect the inflammatory burden and plaque vulnerability leading to less cardiovascular events and myocardial infarctions. An aggressive management of inflammation may lead to a significant improvement in the clinical cardiovascular outcome of patients with RA. The clinical evidence that showed a reduced risk of CV events following treatment with anti-inflammatory agents may suggest a new approach to treat atherosclerosis, i.e., inhibition of inflammation using biological medications that were primarily aimed to treat the high scale inflammation of RA and other autoimmune-inflammatory diseases, but may be useful also to prevent progression of atherosclerosis.
Collapse
|
116
|
Ursini F, Ruscitti P, Caio GPI, Manfredini R, Giacomelli R, De Giorgio R. The effect of non-TNF-targeted biologics on vascular dysfunction in rheumatoid arthritis: A systematic literature review. Autoimmun Rev 2019; 18:501-509. [DOI: 10.1016/j.autrev.2019.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 11/25/2018] [Indexed: 02/08/2023]
|
117
|
Ellis KL, Chakraborty A, Moses EK, Watts GF. To test, or not to test: that is the question for the future of lipoprotein(a). Expert Rev Cardiovasc Ther 2019; 17:241-250. [PMID: 30916582 DOI: 10.1080/14779072.2019.1596799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Lipoprotein(a) [Lp(a)] is a potent, highly heritable and common risk factor for atherosclerotic cardiovascular disease (ASCVD). Evidence for a causal association between elevated Lp(a) and ASCVD has been provided by large epidemiological investigations that have demonstrated a curvilinear association with increased risk, as well as from genetic examinations and cellular and transgenic animal studies. Although there are several therapies available for lowering Lp(a), none are selective for Lp(a) and there is no clinical trial data that has specifically shown that lowering Lp(a) reduces the risk of ASCVD. Hence, screening for elevated Lp(a) is not routinely incorporated into clinical practice. Areas covered: This paper reviews the current evidence supporting the causal role of Lp(a) in the primary and secondary prevention of ASCVD, screening approaches for high Lp(a), current guidelines on testing Lp(a), and barriers to the routine screening of elevated Lp(a) in clinical practice. Expert opinion: At present, there is a moderate level of evidence supporting the routine screening of elevated Lp(a). Current guidelines recommend testing for elevated Lp(a) in individuals at intermediate or high risk of ASCVD.
Collapse
Affiliation(s)
- Katrina L Ellis
- a Centre for Genetic Origins of Health and Disease, School of Biomedical Sciences, The University of Western Australia and School of Biomedical Sciences , Curtin University , Perth , Australia.,b School of Medicine, Faculty of Medicine and Health Sciences , University of Western Australia , Perth , Australia
| | - Anindita Chakraborty
- b School of Medicine, Faculty of Medicine and Health Sciences , University of Western Australia , Perth , Australia
| | - Eric K Moses
- a Centre for Genetic Origins of Health and Disease, School of Biomedical Sciences, The University of Western Australia and School of Biomedical Sciences , Curtin University , Perth , Australia
| | - Gerald F Watts
- b School of Medicine, Faculty of Medicine and Health Sciences , University of Western Australia , Perth , Australia.,c Lipid Disorders Clinic, Department of Cardiology , Royal Perth Hospital , Perth , Australia
| |
Collapse
|
118
|
Ikonomidis I, Pavlidis G, Katsimbri P, Andreadou I, Triantafyllidi H, Tsoumani M, Varoudi M, Vlastos D, Makavos G, Kostelli G, Βenas D, Lekakis J, Parissis J, Boumpas D, Alexopoulos D, Iliodromitis E. Differential effects of inhibition of interleukin 1 and 6 on myocardial, coronary and vascular function. Clin Res Cardiol 2019; 108:1093-1101. [PMID: 30859382 DOI: 10.1007/s00392-019-01443-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/18/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Anakinra, an interleukin-1 receptor antagonist and tocilizumab, an interleukin-6 receptor blocker, are used for the treatment of rheumatoid arthritis. We investigated the differential effects of anakinra and tocilizumab on myocardial and vascular function in an atherosclerosis model of patients with rheumatoid arthritis. METHODS 120 patients with rheumatoid arthritis were randomized to anakinra (n = 40), tocilizumab (n = 40) or prednisolone (n = 40) for 3 months. Primary outcome measure was the change of left ventricular longitudinal strain after 3 months of treatment. Additionally, we measured coronary flow reserve, flow-mediated dilatation of the brachial artery, carotid-femoral pulse wave velocity, malondialdehyde and protein carbonyls as oxidative stress markers and C-reactive protein blood levels at baseline and post-treatment. RESULTS At baseline, patients among the three treatment arms had similar age, sex, disease activity score and atherosclerotic risk factors. Compared with baseline, all patients had improved longitudinal strain (- 16% vs. - 17.8%), coronary flow reserve (2.56 vs. 2.9), malondialdehyde (2.0 vs. 1.5 µM/L), protein carbonyls (0.0132 vs. 0.0115 nmol/mg), and C-reactive protein post-treatment. In all patients, the percent decrease of malondialdehyde was correlated with percent increase of longitudinal strain (p < 0.001). Compared with tocilizumab and prednisolone, anakinra treatment resulted in a greater improvement of longitudinal strain (18.7% vs. 9.7% vs. 6%) and coronary flow reserve (29% vs. 13% vs. 1%), while pulse wave velocity and brachial blood pressure were improved only after tocilizumab treatment (11 ± 3 vs. 10.3 ± 2 m/s p < 0.05 for all comparisons). CONCLUSIONS Anakinra is associated with an improvement in cardiac function and tocilizumab with improvement in vascular function. CLINICAL TRIAL REGISTRATION URL: https:// http://www.clinicaltrials.gov . Unique identifier: NCT03288584.
Collapse
Affiliation(s)
- Ignatios Ikonomidis
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - George Pavlidis
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Pelagia Katsimbri
- 4th Department of Internal Medicine, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Andreadou
- Department of Pharmaceutical Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Helen Triantafyllidi
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Tsoumani
- Department of Pharmaceutical Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Varoudi
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Vlastos
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - George Makavos
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Gavriella Kostelli
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Βenas
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - John Lekakis
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - John Parissis
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Boumpas
- 4th Department of Internal Medicine, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Alexopoulos
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Iliodromitis
- 2nd Department of Cardiology, Medical School, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
119
|
Borrelli MJ, Youssef A, Boffa MB, Koschinsky ML. New Frontiers in Lp(a)-Targeted Therapies. Trends Pharmacol Sci 2019; 40:212-225. [PMID: 30732864 DOI: 10.1016/j.tips.2019.01.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/13/2022]
Abstract
Interest in lipoprotein (a) [Lp(a)] has exploded over the past decade with the emergence of genetic and epidemiological studies pinpointing elevated levels of this unique lipoprotein as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and calcific aortic valve disease (CAVD). This review summarizes the most recent discoveries regarding therapeutic approaches to lower Lp(a) and presents these findings in the context of an emerging, although far from complete, understanding of the biosynthesis and catabolism of Lp(a). Application of Lp(a)-specific lowering agents to outcome trials will be the key to opening this new frontier in the battle against CVD.
Collapse
Affiliation(s)
- Matthew J Borrelli
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Amer Youssef
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Michael B Boffa
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada; Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Marlys L Koschinsky
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
120
|
Balogh E, Pusztai A, Hamar A, Végh E, Szamosi S, Kerekes G, McCormick J, Biniecka M, Szántó S, Szűcs G, Nagy Z, Fearon U, Veale DJ, Szekanecz Z. Autoimmune and angiogenic biomarkers in autoimmune atherosclerosis. Clin Immunol 2019; 199:47-51. [DOI: 10.1016/j.clim.2018.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
121
|
Metz AK, Diaz JA, Obi AT, Wakefield TW, Myers DD, Henke PK. Venous Thrombosis and Post-Thrombotic Syndrome: From Novel Biomarkers to Biology. Methodist Debakey Cardiovasc J 2019; 14:173-181. [PMID: 30410646 DOI: 10.14797/mdcj-14-3-173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Deep vein thrombosis (DVT) is a common disease that carries serious ramifications for patients, including pulmonary embolism and post-thrombotic syndrome (PTS). Although standard treatment for DVT is anticoagulation, this carries an added risk of bleeding and increased medication monitoring. Identifying those at risk for DVT and PTS can be difficult, and current research with murine models is helping to illuminate the biologic changes associated with these two disorders. Potential novel biomarkers for improving the diagnosis of DVT and PTS include ICAM-1, P-selectin, and cell-free DNA. Inhibition of factor XI, P- and E-selectin, and neutrophil extracellular traps holds promise for novel clinical treatment of DVT. Experimental research on PTS suggests potential cellular and mediator therapy targets of TLR9, MMP-2 and-9, PAI-1, and IL-6. Although many important concepts and mechanisms have been elucidated through research on DVT and PTS, more work must be done to translate experimental findings to the clinical arena. This review examines the currently used murine models of DVT, biomarkers involved in the pathophysiology and diagnosis of DVT and PTS, and potential pharmacologic targets for PTS treatment.
Collapse
|
122
|
Jones SA, Takeuchi T, Aletaha D, Smolen J, Choy EH, McInnes I. Interleukin 6: The biology behind the therapy. ACTA ACUST UNITED AC 2019. [DOI: 10.1136/conmed-2018-000005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The cytokine interleukin (IL)−6 performs a diverse portfolio of functions in normal physiology and disease. These functions extend beyond the typical role for an inflammatory cytokine, and IL-6 often displays hormone-like properties that affect metabolic processes associated with lipid metabolism, insulin resistance, and the neuroendocrine system. Consequently, the biology of IL-6 is complex. Recent advances in the field have led to novel interpretations of how IL-6 delivers immune homeostasis in health and yet drives disease pathology during infection, autoimmunity, and cancer. Various biological drugs that target IL-6 are in clinical practice or emerging in clinical trials and pre-clinical development programmes. The challenge is knowing how and when to apply these therapies. In this review, we will explore the biology behind IL-6 directed therapies and identify some key hurdles for future investigation.
Collapse
|
123
|
Tuñón J, Badimón L, Bochaton-Piallat ML, Cariou B, Daemen MJ, Egido J, Evans PC, Hoefer IE, Ketelhuth DFJ, Lutgens E, Matter CM, Monaco C, Steffens S, Stroes E, Vindis C, Weber C, Bäck M. Identifying the anti-inflammatory response to lipid lowering therapy: a position paper from the working group on atherosclerosis and vascular biology of the European Society of Cardiology. Cardiovasc Res 2019; 115:10-19. [PMID: 30534957 PMCID: PMC6302260 DOI: 10.1093/cvr/cvy293] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/21/2018] [Accepted: 12/04/2018] [Indexed: 12/14/2022] Open
Abstract
Dysregulated lipid metabolism induces an inflammatory and immune response leading to atherosclerosis. Conversely, inflammation may alter lipid metabolism. Recent treatment strategies in secondary prevention of atherosclerosis support beneficial effects of both anti-inflammatory and lipid-lowering therapies beyond current targets. There is a controversy about the possibility that anti-inflammatory effects of lipid-lowering therapy may be either independent or not of a decrease in low-density lipoprotein cholesterol. In this Position Paper, we critically interpret and integrate the results obtained in both experimental and clinical studies on anti-inflammatory actions of lipid-lowering therapy and the mechanisms involved. We highlight that: (i) besides decreasing cholesterol through different mechanisms, most lipid-lowering therapies share anti-inflammatory and immunomodulatory properties, and the anti-inflammatory response to lipid-lowering may be relevant to predict the effect of treatment, (ii) using surrogates for both lipid metabolism and inflammation as biomarkers or vascular inflammation imaging in future studies may contribute to a better understanding of the relative importance of different mechanisms of action, and (iii) comparative studies of further lipid lowering, anti-inflammation and a combination of both are crucial to identify effects that are specific or shared for each treatment strategy.
Collapse
Affiliation(s)
- José Tuñón
- Department of Cardiology, Fundación Jiménez Díaz, Autónoma University and CiberCV, Avenida Reyes Católicos 2, Madrid, Spain
| | - Lina Badimón
- Cardiovascular Sciences Institute (ICCC) and CiberCV, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Bertrand Cariou
- L’Institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Mat J Daemen
- Academic Medical Center, Amsterdam, The Netherlands
| | - Jesus Egido
- Fundación Jiménez Díaz, Autónoma University and CIBERDEM, Madrid, Spain
| | | | - Imo E Hoefer
- University Medical Centre Utrecht, Utrecht, Netherlands
| | | | - Esther Lutgens
- Academic Medical Center, Amsterdam, The Netherlands
- University of Amsterdam, Amsterdam, The Netherlands
- Ludwig-Maximilians-University, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian M Matter
- University Heart Center, University Hospital Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Claudia Monaco
- Kennedy Institute, NDORMS, University of Oxford, Oxford, UK
| | - Sabine Steffens
- Ludwig-Maximilians-University, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | | | - Cécile Vindis
- INSERM UMR-1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Christian Weber
- Ludwig-Maximilians-University, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Magnus Bäck
- Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
124
|
Jialal I, Chaudhuri A. Targeting inflammation to reduce ASCVD in type 2 diabetes. J Diabetes Complications 2019; 33:1-3. [PMID: 30514609 DOI: 10.1016/j.jdiacomp.2018.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 01/12/2023]
Abstract
ASCVD is the leading cause of mortality in T2DM. Inflammation appears to be pivotal in the genesis of ASCVD. As T2DM is also a pro-inflammatory state, our aim was to determine the benefit of anti-inflammatory strategies on ASCVD in T2DM. PubMed searches were conducted using the keywords of T2DM, ASCVD, Inflammation and clinical trials. Our data review suggests that the Mediterranean diet, GLP1 receptor agonists and a monoclonal antibody against IL-1 reduces ASCVD events in T2DM. The former 2 therapies appear to be safe. Anti-IL-1 therapy resulted in an increase mortality from infections. We conclude that only the Mediterranean diet and GLP1 receptor agonists can be safely incorporated into mainstay therapy for patients with T2DM to reduce ASCVD. Further studies are required with respect to biologics targeting Inflammation to establish benefit to risk ratio.
Collapse
Affiliation(s)
- Ishwarlal Jialal
- VA Medical Center, Mather, CA, United States of America; California Northstate University College of Medicine, Elk Grove, CA, United States of America.
| | - Ajay Chaudhuri
- University at Buffalo, Buffalo, NY, United States of America
| |
Collapse
|
125
|
Moriya J. Critical roles of inflammation in atherosclerosis. J Cardiol 2019; 73:22-27. [DOI: 10.1016/j.jjcc.2018.05.010] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 01/02/2023]
|
126
|
Bartoloni E, Alunno A, Valentini V, Luccioli F, Valentini E, La Paglia GMC, Leone MC, Cafaro G, Marcucci E, Gerli R. Targeting Inflammation to Prevent Cardiovascular Disease in Chronic Rheumatic Diseases: Myth or Reality? Front Cardiovasc Med 2018; 5:177. [PMID: 30619884 PMCID: PMC6297850 DOI: 10.3389/fcvm.2018.00177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/29/2018] [Indexed: 01/10/2023] Open
Abstract
Evidence for increased risk of cardiovascular morbidity and mortality in chronic inflammatory rheumatic diseases has accumulated during the last years. Traditional cardiovascular risk factors contribute in part to the excess of cardiovascular risk in these patients and several mechanisms, including precocious acceleration of subclinical atherosclerotic damage, inflammation, and immune system deregulation factors, have been demonstrated to strictly interplay in the induction and progression of atherosclerosis. In this setting, chronic inflammation is a cornerstone of rheumatic disease pathogenesis and exerts also a pivotal role in all stages of atherosclerotic damage. The strict link between inflammation and atherosclerosis suggests that cardiovascular risk may be reduced by rheumatic disease activity control. There are data to suggest that biologic therapies, in particular TNFα antagonists, may improve surrogate markers of cardiovascular disease and reduce CV adverse outcome. Thus, abrogation of inflammation is considered an important outcome for achieving not only control of rheumatic disease, but also reduction of cardiovascular risk. However, the actual effect of anti-rheumatic therapies on atherosclerosis progression and CV outcome in these patients is rather uncertain due to great literature inconsistency. In this paper, we will summarize some of the main mechanisms linking the inflammatory pathogenic background underlying rheumatic diseases and the vascular damage observed in these patients, with a particular emphasis on the pathways targeted by currently available therapies. Moreover, we will analyze current evidence on the potential atheroprotective effects of these treatments on cardiovascular outcome pointing out still unresolved questions.
Collapse
Affiliation(s)
- Elena Bartoloni
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Alessia Alunno
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Valentina Valentini
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Filippo Luccioli
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Eleonora Valentini
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | | | - Maria Comasia Leone
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Giacomo Cafaro
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Elisa Marcucci
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| | - Roberto Gerli
- Rheumatology Unit, Department of Perugia, University of Perugia, Perugia, Italy
| |
Collapse
|
127
|
Schulte DM, Paulsen K, Türk K, Brandt B, Freitag-Wolf S, Hagen I, Zeuner R, Schröder JO, Lieb W, Franke A, Nikolaus S, Mrowietz U, Gerdes S, Schreiber S, Laudes M. Small dense LDL cholesterol in human subjects with different chronic inflammatory diseases. Nutr Metab Cardiovasc Dis 2018; 28:1100-1105. [PMID: 30143407 DOI: 10.1016/j.numecd.2018.06.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/06/2018] [Accepted: 06/27/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND AND AIMS Chronic inflammatory diseases (CID) are associated with a profound increase in cardiovascular (CV) risk resulting in reduced life expectancy. However, LDL-cholesterol is reported to be low in CID patients which is referred to as the "LDL paradoxon". The aim of the present study was to investigate whether LDL-particles in CID exhibit an increased content of the highly atherogenic small-dense LDL subfraction (sdLDL). METHODS AND RESULTS In this prospective, single center, observational study we enrolled 141 patients with CID (RA n = 59, inflammatory bowel disease (IBD) n = 35, ankylosing spondylitis (SpA) n = 25, Psoriasis n = 22) in 2011 through 2013 to evaluate sdLDL levels before as well as 6 and 26 weeks after initiation of different anti-cytokine therapies (anti-TNFα, anti-IL-6R antibodies). sdLDL levels were compared to 141 healthy individuals in a case control design. Compared to healthy controls, all CID patients displayed a significantly higher sdLDL content within the LDL cholesterol fraction: RA 35.0 ± 9.2% (p < 0.001), SpA 42.5 ± 10.5% (p < 0.001), IBD 37.5 ± 7.1% (p < 0.001), Psoriasis 33.6 ± 4.6% (p < 0.01). Furthermore, the sdLDL/LDL ratio was significantly higher in male compared to female RA subjects (p < 0.05). Neither anti-TNFα nor anti-IL6R medication altered sdLDL levels despite a significant improvement of disease activity. CONCLUSION In several different chronic inflammatory disease entities, LDL-cholesterol is shifted toward a pro-atherogenic phenotype due to an increased sdLDL content which might in part explain the LDL paradoxon. Since premature CV disease is a major burden of affected patients, specifically targeting lipid metabolism should be considered routinely in clinical patient care. CLINICAL TRIALS Registration at German Clinical Trial Register (DRKS): DRKS00005285.
Collapse
Affiliation(s)
- D M Schulte
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - K Paulsen
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - K Türk
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - B Brandt
- Institute of Clinical Chemistry, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - S Freitag-Wolf
- Institute of Medical Informatics and Statistics, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - I Hagen
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - R Zeuner
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - J O Schröder
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - W Lieb
- Institute of Epidemiology, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - A Franke
- Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Institute of Clinical Molecular Biology, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - S Nikolaus
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - U Mrowietz
- Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Psoriasis-Center at the Department of Dermatology, University Medical Center Schleswig-Holstein, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - S Gerdes
- Psoriasis-Center at the Department of Dermatology, University Medical Center Schleswig-Holstein, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - S Schreiber
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany
| | - M Laudes
- Department of Internal Medicine I, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany; Cluster of Excellence Inflammation at Interfaces, University of Kiel, Arnold-Heller-Strasse 3, D-24105, Kiel, Germany.
| |
Collapse
|
128
|
Braun J, Krüger K, Manger B, Schneider M, Specker C, Trappe HJ. Cardiovascular Comorbidity in Inflammatory Rheumatological Conditions. DEUTSCHES ARZTEBLATT INTERNATIONAL 2018; 114:197-203. [PMID: 28407841 DOI: 10.3238/arztebl.2017.0197] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 05/03/2016] [Accepted: 01/18/2017] [Indexed: 01/12/2023]
Abstract
BACKGROUND Approximately 1.5 million adults in Germany suffer from an inflammatory rheumatological condition. The most common among these are rheumatoid arthritis and spondyloarthritis-above all axial spondyloarthritis, including ankylosing spondylitis (Bekhterev's disease) and psoriatic arthritis. These systemic inflammatory diseases often affect the heart as well. METHODS This review is based on pertinent articles retrieved by a selective literature search, on current European guidelines, and on the authors' clinical experience. RESULTS Rheumatic inflammation of cardiac structures can manifest itself as pericarditis, myocarditis, or endocarditis. The heart valves and the intracardiac conduction system can be affected as well, leading to AV block. Functional sequelae, e.g., congestive heart failure, can arise as a consequence of any inflammatory rheumatic disease. The long-term mortality of rheumatic diseases is elevated predominantly because of the increased risk for cardiovascular comorbidities. The cardiovascular risk profile should therefore be re-evaluated regularly (e.g., at 5-year intervals) in cooperation with the patient's primary care physician. The cardiovascular manifestations of rheumatic disease, such as pericarditis, myocarditis, and vasculitis, are treated initially with high-dose glucocorticoids and then over the long term with maintenance drugs such as methotrexate and azathioprine. Biological agents are sometimes used as well. CONCLUSION In patients with inflammatory rheumatic diseases, the elevated cardiovascular risk should be kept in mind and preventive measures should be initiated early. This subject should be further studied in controlled trials so that the treatment options for patients with cardiac involvement can be evaluated.
Collapse
Affiliation(s)
- Jürgen Braun
- Rheumazentrum Ruhrgebiet, Herne; Rheumazentrum München, Munich; Department of Medicine 3, Universitätsklinikum Erlangen; Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf; Department of Rheumatology and Clinical Immunology, St. Josef Krankenhaus, Essen University Hospital; Department of Cardiology, Marien-Hospital Herne, University Hospitals of the Ruhr University of Bochum
| | | | | | | | | | | |
Collapse
|
129
|
Lipids and Atherogenic Indices Fluctuation in Rheumatoid Arthritis Patients on Long-Term Tocilizumab Treatment. Mediators Inflamm 2018; 2018:2453265. [PMID: 30405318 PMCID: PMC6204176 DOI: 10.1155/2018/2453265] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 09/07/2018] [Accepted: 09/19/2018] [Indexed: 12/05/2022] Open
Abstract
Rheumatoid arthritis (RA) patients are at high risk of cardiovascular (CV) events, and the chronic inflammatory state may generate quantitative and qualitative changes in lipoprotein fractions. The anti-IL-6 receptor tocilizumab (TCZ), even if effective in inflammation and joint damage prevention, determined significant alterations to RA patients' lipid levels in randomized controlled trials, but real-world data are lacking. We evaluated the changes in lipid fraction levels and disease activity in a longitudinal cohort of RA patients on long-term treatment with tocilizumab (TCZ) in a community setting. We retrospectively selected 40 naïve-biologic RA patients on treatment with intravenous TCZ compared to 20 RA patients on methotrexate treatment as the control group. Total cholesterol (Tot-Chol), low-density lipoproteins (LDL), high-density lipoprotein (HDL), and triglyceride (TG) levels were measured at the baseline and at 12, 24, and 52 weeks thereafter. At the same points, 28-joint disease activity score (DAS28), clinical disease activity index (CDAI), and EULAR clinical responses were also assessed. During the first 24 weeks, we observed in TCZ-treated patients a progressive statistically significant (p < 0.001) increase in Tot-Chol, LDL, HDL, and TG, which returned close to the baseline at 52 weeks. But no changes in the lipid-related CV risk indices Tot-Chol/HDL and LDL/HDL ratios and the atherogenic index (log10 TG/HDL) were detectable. Notably, we observed a statistically significant negative correlation between changes in lipid fractions and DAS28 or CDAI. The prolonged treatment with TCZ was associated to a transient increase in cholesterol's fractions during the first 6 months of treatment, with inverse correlation to disease activity, but with no impact on surrogate lipid indices of atherogenic risk. These findings may aid clinicians in interpreting the RA patient's lipid profile in daily clinical practice.
Collapse
|
130
|
Abstract
Lipoprotein (a) is a low-density lipoprotein-like particle covalently bound to a glycoprotein called apolipoprotein(a) that is under potent genetic control. Plasma levels of lipoprotein (a) vary by up to 1000-fold among individuals, with 1 in 4 having levels that increase the risk of atherosclerotic cardiovascular disease. New evidence supports a causal role for lipoprotein (a) in atherosclerotic cardiovascular disease and aortic valve stenosis. Individuals with elevated lipoprotein (a) have a high life-time burden of atherosclerotic cardiovascular disease. This notion is important for coronary prevention. But is lipoprotein (a) ready for prime-time use in coronary prevention clinics?
Collapse
Affiliation(s)
- Katrina L Ellis
- School of Medicine, University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia; School of Biomedical Sciences, University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Gerald F Watts
- School of Medicine, University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia; Department of Cardiology, Lipid Disorders Clinic, Royal Perth Hospital, GPO Box X2213, Perth, WA 6001, Australia.
| |
Collapse
|
131
|
Cai T, Zhang Y, Ho YL, Link N, Sun J, Huang J, Cai TA, Damrauer S, Ahuja Y, Honerlaw J, Huang J, Costa L, Schubert P, Hong C, Gagnon D, Sun YV, Gaziano JM, Wilson P, Cho K, Tsao P, O’Donnell CJ, Liao KP. Association of Interleukin 6 Receptor Variant With Cardiovascular Disease Effects of Interleukin 6 Receptor Blocking Therapy: A Phenome-Wide Association Study. JAMA Cardiol 2018; 3:849-857. [PMID: 30090940 PMCID: PMC6233652 DOI: 10.1001/jamacardio.2018.2287] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/13/2018] [Indexed: 12/30/2022]
Abstract
Importance Electronic health record (EHR) biobanks containing clinical and genomic data on large numbers of individuals have great potential to inform drug discovery. Individuals with interleukin 6 receptor (IL6R) single-nucleotide polymorphisms (SNPs) who are not receiving IL6R blocking therapy have biomarker profiles similar to those treated with IL6R blockers. This gene-drug pair provides an example to test whether associations of IL6R SNPs with a broad range of phenotypes can inform which diseases may benefit from treatment with IL6R blockade. Objective To determine whether screening for clinical associations with the IL6R SNP in a phenome-wide association study (PheWAS) using EHR biobank data can identify drug effects from IL6R clinical trials. Design, Setting, and Participants Diagnosis codes and routine laboratory measurements were extracted from the VA Million Veteran Program (MVP); diagnosis codes were mapped to phenotype groups using published PheWAS methods. A PheWAS was performed by fitting logistic regression models for testing associations of the IL6R SNPs with 1342 phenotype groups and by fitting linear regression models for testing associations of the IL6R SNP with 26 routine laboratory measurements. Significance was reported using a false discovery rate of 0.05 or less. Findings were replicated in 2 independent cohorts using UK Biobank and Vanderbilt University Biobank data. The Million Veteran Program included 332 799 US veterans; the UK Biobank, 408 455 individuals from the general population of the United Kingdom; and the Vanderbilt University Biobank, 13 835 patients from a tertiary care center. Exposures IL6R SNPs (rs2228145; rs4129267). Main Outcomes and Measures Phenotypes defined by International Classification of Diseases, Ninth Revision codes. Results Of the 332 799 veterans included in the main cohort, 305 228 (91.7%) were men, and the mean (SD) age was 66.1 (13.6) years. The IL6R SNP was most strongly associated with a reduced risk of aortic aneurysm phenotypes (odds ratio, 0.87-0.90; 95% CI, 0.84-0.93) in the MVP. We observed known off-target effects of IL6R blockade from clinical trials (eg, higher hemoglobin level). The reduced risk for aortic aneurysms among those with the IL6R SNP in the MVP was replicated in the Vanderbilt University Biobank, and the reduced risk for coronary heart disease was replicated in the UK Biobank. Conclusions and Relevance In this proof-of-concept study, we demonstrated application of the PheWAS using large EHR biobanks to inform drug effects. The findings of an association of the IL6R SNP with reduced risk for aortic aneurysms correspond with the newest indication for IL6R blockade, giant cell arteritis, of which a major complication is aortic aneurysm.
Collapse
Affiliation(s)
- Tianxi Cai
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Yichi Zhang
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Yuk-Lam Ho
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Nicholas Link
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Jiehuan Sun
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Jie Huang
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| | - Tianrun A. Cai
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| | - Scott Damrauer
- Corporal Michael Crescenz Veterans Affairs Medical Center, Perlman School of Medicine, University of Pennsylvania, Philadelphia
| | - Yuri Ahuja
- Harvard Medical School, Boston, Massachusetts
| | | | - Jie Huang
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Lauren Costa
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Petra Schubert
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Chuan Hong
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - David Gagnon
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Boston University School of Public Health, Boston, Massachusetts
| | - Yan V. Sun
- Emory University Schools of Medicine and Public Health, Atlanta, Georgia
| | - J. Michael Gaziano
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| | - Peter Wilson
- Emory University Schools of Medicine and Public Health, Atlanta, Georgia
- Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Kelly Cho
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| | - Philip Tsao
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Department of Medicine, Stanford University of Medicine, Stanford, California
| | - Christopher J. O’Donnell
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Associate Editor, JAMA Cardiology
| | - Katherine P. Liao
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
132
|
Abstract
Most older individuals develop inflammageing, a condition characterized by elevated levels of blood inflammatory markers that carries high susceptibility to chronic morbidity, disability, frailty, and premature death. Potential mechanisms of inflammageing include genetic susceptibility, central obesity, increased gut permeability, changes to microbiota composition, cellular senescence, NLRP3 inflammasome activation, oxidative stress caused by dysfunctional mitochondria, immune cell dysregulation, and chronic infections. Inflammageing is a risk factor for cardiovascular diseases (CVDs), and clinical trials suggest that this association is causal. Inflammageing is also a risk factor for chronic kidney disease, diabetes mellitus, cancer, depression, dementia, and sarcopenia, but whether modulating inflammation beneficially affects the clinical course of non-CVD health problems is controversial. This uncertainty is an important issue to address because older patients with CVD are often affected by multimorbidity and frailty - which affect clinical manifestations, prognosis, and response to treatment - and are associated with inflammation by mechanisms similar to those in CVD. The hypothesis that inflammation affects CVD, multimorbidity, and frailty by inhibiting growth factors, increasing catabolism, and interfering with homeostatic signalling is supported by mechanistic studies but requires confirmation in humans. Whether early modulation of inflammageing prevents or delays the onset of cardiovascular frailty should be tested in clinical trials.
Collapse
Affiliation(s)
- Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA.
| | - Elisa Fabbri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
133
|
Research Progress on the Relationship between Atherosclerosis and Inflammation. Biomolecules 2018; 8:biom8030080. [PMID: 30142970 PMCID: PMC6163673 DOI: 10.3390/biom8030080] [Citation(s) in RCA: 524] [Impact Index Per Article: 74.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/03/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease; unstable atherosclerotic plaque rupture, vascular stenosis, or occlusion caused by platelet aggregation and thrombosis lead to acute cardiovascular disease. Atherosclerosis-related inflammation is mediated by proinflammatory cytokines, inflammatory signaling pathways, bioactive lipids, and adhesion molecules. This review discusses the effects of inflammation and the systemic inflammatory signaling pathway on atherosclerosis, the role of related signaling pathways in inflammation, the formation of atherosclerosis plaques, and the prospects of treating atherosclerosis by inhibiting inflammation.
Collapse
|
134
|
Tornero Molina J, Balsa Criado A, Blanco García F, Blanco Alonso R, Bustabad S, Calvo Alen J, Corominas H, Fernández Nebro A, Román Ivorra JA, Sanmartí R. Expert Recommendations on the Interleukin 6 Blockade in Patients with Rheumatoid Arthritis. ACTA ACUST UNITED AC 2018; 16:272-281. [PMID: 30098882 DOI: 10.1016/j.reuma.2018.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/06/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To draft recommendations on interleukin 6 (IL-6) blockade in rheumatoid arthritis (RA), based on best evidence and experience. METHODS A group of 10 experts on IL-6 blockade in RA was selected. The 2 coordinators formulated 23 questions about IL-6 blockade (indications, efficacy, safety, etc.). A systematic review was conducted to answer the questions. Using this information, inclusion and exclusion criteria were established, as were the search strategies (Medline, EMBASE and the Cochrane Library were searched). Two different reviewers selected the articles. Evidence tables were created. At the same time, European League Against Rheumatism and American College of Rheumatology abstracts were evaluated. Based on this evidence, the coordinators proposed preliminary recommendations that the experts discussed and voted on in a nominal group meeting. The level of evidence and grade of recommendation were established using the Oxford Centre for Evidence Based Medicine and the level of agreement with the Delphi technique (2 rounds). Agreement was established if at least 80% of the experts voted yes (yes/no). RESULTS The 8 preliminary recommendations were accepted after the Delphi process. They covered aspects such as the use of these therapies in monotherapy, in combination, in patients with refractory disease or intolerant patients, response evaluation, optimization and risk management. CONCLUSIONS The manuscript aims to solve frequently asked questions and aid in decision making strategies when treating RA patients with IL-6 blockade.
Collapse
Affiliation(s)
- Jesús Tornero Molina
- Servicio de Reumatología, Hospital Universitario de Guadalajara; Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, España.
| | | | - Francisco Blanco García
- Servicio de Reumatología, Complexo Hospitalario Universitario A Coruña (CHUAC), La Coruña, España
| | - Ricardo Blanco Alonso
- Servicio de Reumatología, Hospital Universitario Marqués de Valdecilla, Santander, España
| | - Sagrario Bustabad
- Servicio de Reumatología, Hospital Universitario de Canarias, Santa Cruz de Tenerife, España
| | - Jaime Calvo Alen
- Servicio de Reumatología, Hospital Universitario Araba, Vitoria-Gasteiz, España
| | - Héctor Corominas
- Servicio de Reumatología, Hospital Universitari de la Santa Creu i Sant Pau, Barcelona, España
| | - Antonio Fernández Nebro
- Unidad de Gestión Clínica de Reumatología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, España
| | | | - Raimon Sanmartí
- Servicio de Reumatología, Hospital Universitari Clínic, Barcelona, España
| |
Collapse
|
135
|
Impact of obesity on autoimmune arthritis and its cardiovascular complications. Autoimmun Rev 2018; 17:821-835. [PMID: 29885537 PMCID: PMC9996646 DOI: 10.1016/j.autrev.2018.02.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 02/25/2018] [Indexed: 02/06/2023]
Abstract
Obesity can instigate and sustain a systemic low-grade inflammatory environment that can amplify autoimmune disorders and their associated comorbidities. Metabolic changes and inflammatory factors produced by the adipose tissue have been reported to aggravate autoimmunity and predispose the patient to cardiovascular disease (CVD) and metabolic comorbidities. Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) are autoimmune arthritic diseases, often linked with altered body mass index (BMI). Severe joint inflammation and bone destruction have a debilitating impact on the patient's life; there is also a staggering risk of cardiovascular morbidity and mortality. Furthermore, these patients are at risk of developing metabolic symptoms, including insulin resistance resulting in type 2 diabetes mellitus (T2DM). In addition, arthritis severity, progression and response to therapy can be markedly affected by the patient's BMI. Hence, a complex integrative pathogenesis interconnects autoimmunity with metabolic and cardiovascular disorders. This review aims to shed light on the network that connects obesity with RA, PsA, systemic lupus erythematosus and Sjӧgren's syndrome. We have focused on clarifying the mechanism by which obesity affects different cell types, inflammatory factors and traditional therapies in these autoimmune disorders. We conclude that to further optimize arthritis therapy and to prevent CVD, it is imperative to uncover the intricate relation between obesity and arthritis pathology.
Collapse
|
136
|
Bergström U, Jovinge S, Persson J, Jacobsson LTH, Turesson C. Effects of Treatment with Adalimumab on Blood Lipid Levels and Atherosclerosis in Patients with Rheumatoid Arthritis. Curr Ther Res Clin Exp 2018; 89:1-6. [PMID: 30128057 PMCID: PMC6097545 DOI: 10.1016/j.curtheres.2018.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 07/15/2018] [Accepted: 07/17/2018] [Indexed: 01/04/2023] Open
Abstract
Background Treatment with tumor necrosis factor inhibitors for rheumatoid arthritis has been associated with a decreased risk of cardiovascular disease in observational studies. There are conflicting data on the influence of tumor necrosis factor inhibitors on lipid levels. Objectives To evaluate the effect of treatment with adalimumab on blood lipid levels, lipoproteins, and atherosclerosis of the carotid artery. Methods Fourteen patients with active rheumatoid arthritis (11 women and 3 men; mean age 63.7 years; median disease duration 9.0 years; and 78% rheumatoid factor positive) were treated with adalimumab 40 mg subcutaneously every 2 weeks and followed for 3 months. The patients had not been treated with adalimumab previously and had not received other tumor necrosis factor inhibitors within the past 3 months or moderate/high dose corticosteroids within the past 2 weeks. The intima-media thickness of the common carotid artery was assessed using B mode ultrasonography. Triglycerides, total cholesterol, LDL cholesterol, and HDL cholesterol levels were analyzed in fresh fasting blood samples, whereas apolipoprotein B and apolipoprotein A1 (apoA1) levels were determined in thawed plasma samples using standard turbidimetric immunoassays. Results Total cholesterol (mean = 5.36 vs 5.96 mmol/L; P = 0.005), LDL cholesterol (mean = 3.33 vs 3.77 mmol/L; P = .005), HDL cholesterol (mean = 1.43 vs 1.55 mmol/L; P = 0.048), apolipoprotein B (mean = 1.04 vs 1.13 g/L; P = .012), and apoA1 (mean = 1.42 vs 1.58 g/L; P = 0.005) all increased, but there were no major changes in the LDL to HDL cholesterol ratio (median = 2.56 vs 2.35; P = 0.27) or the apolipoprotein B to apoA1 ratio (mean = 0.76 vs 0.74; P = 0.46). There was no change in triglyceride levels (P = 0.55). Disease activity decreased significantly from baseline to the 3-month evaluation (disease activity score based on 28 joints mean = 5.6 vs 4.1; P = 0.007). An increase in apoA1 correlated with decreases in the patient global assessment of disease severity (r = 0.79; P = 0.001) and C-reactive protein level (r = 0.74; P = 0.003). Changes in the apoliprotein B to apoA1 ratio correlated with changes in erythrocyte sedimentation rate (r = 0.54; P = 0.046). There was no major change in the common carotid artery intima-media thickness (mean = 0.78 vs 0.80 mm; P = 0.48). Conclusions Although these results suggest that control of inflammation could have a beneficial effect on the lipid profile through an increase in HDL cholesterol levels, the observed protective effect on cardiovascular disease events by tumor necrosis factor blockers is likely to be explained by other mechanisms than changes in lipid levels or short-term effects on atherosclerosis of the carotid artery.
Collapse
Affiliation(s)
- Ulf Bergström
- Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden.,Department of Rheumatology, Skåne University Hospital, Malmö, Sweden
| | - Stefan Jovinge
- Fred and Lena Meijer Heart and Vascular Institute, Spectrum Health, Grand Rapids, Michigan.,Van Andel Institute, Grand Rapids, Michigan
| | - Jerker Persson
- Department of Medicine, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Lennart T H Jacobsson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| | - Carl Turesson
- Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden.,Department of Rheumatology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
137
|
Khambhati J, Engels M, Allard-Ratick M, Sandesara PB, Quyyumi AA, Sperling L. Immunotherapy for the prevention of atherosclerotic cardiovascular disease: Promise and possibilities. Atherosclerosis 2018; 276:1-9. [PMID: 30006321 DOI: 10.1016/j.atherosclerosis.2018.07.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/27/2018] [Accepted: 07/05/2018] [Indexed: 12/23/2022]
Abstract
Cardiovascular disease remains the leading cause of death worldwide with coronary atherosclerotic heart disease being the largest contributor. The mechanisms behind the presence and progression of atherosclerosis remain an area of intense scientific focus. Immune dysregulation and inflammation are key contributors to the development of an atherosclerotic plaque and its progression to acute coronary syndromes. Increased circulating levels of biomarkers of systemic inflammation including hsCRP are correlated with a higher cardiovascular risk. Targeting specific inflammatory pathways implicated in atherosclerotic plaque formation is an exciting area of ongoing research. Target specific therapies directed at pro-inflammatory cytokines such as IL-1β, IL-6, TNFα, and CCL2 have demonstrated slowing in the progression of atherosclerosis in animal models and improved cardiovascular outcomes in human subjects. Most notably, treatment with the monoclonal antibody canakinumab, which directly targets and neutralizes IL-1β, was recently shown to be associated with reduced risk of adverse cardiovascular events compared to placebo in a randomized, placebo-controlled trial. Several other therapies including colchicine, methotrexate and leukotriene inhibitors demonstrate the potential for lowering cardiovascular risk through immunomodulation, though further studies are needed. Understanding the role of inflammation in atherosclerosis and the development of targeted immunotherapies continues to be an evolving area of research that is rapidly becoming clinically relevant for the 21st century cardiac patient.
Collapse
Affiliation(s)
- Jay Khambhati
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| | - Marc Engels
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Marc Allard-Ratick
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Pratik B Sandesara
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Laurence Sperling
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
138
|
Taylor PC, Kremer JM, Emery P, Zuckerman SH, Ruotolo G, Zhong J, Chen L, Witt S, Saifan C, Kurzawa M, Otvos JD, Connelly MA, Macias WL, Schlichting DE, Rooney TP, de Bono S, McInnes IB. Lipid profile and effect of statin treatment in pooled phase II and phase III baricitinib studies. Ann Rheum Dis 2018; 77:988-995. [PMID: 29463520 PMCID: PMC6029633 DOI: 10.1136/annrheumdis-2017-212461] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/02/2018] [Accepted: 02/03/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Lipid profiles are altered by active disease in patients with rheumatoid arthritis (RA) and may be further modified by treatment with Janus kinase inhibitors and other disease-modifying antirheumatic drugs. METHODS Lipid data were analysed from phase II and III studies of 4 mg (n=997) and 2 mg (n=479) oral baricitinib administered once daily in patients with moderate-to-severe active RA. Lipoprotein particle size and number and GlycA were evaluated with nuclear magnetic resonance in one phase III study. The effect of statin therapy on lipid levels was evaluated in patients on statins at baseline and in patients who initiated statins during the study. RESULTS Treatment with baricitinib was associated with increased levels of total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C) and triglycerides, but no significant change in LDL-C:HDL-C ratio. Lipid levels plateaued after 12 weeks of treatment. Baricitinib treatment increased large LDL and decreased small, dense LDL particle numbers and GlycA. Lipid changes from baseline were not significantly different between baseline statin users and non-users. In patients who initiated statin therapy during the study, LDL-C, triglycerides (baricitinib 4 mg only) and apolipoprotein B decreased to pre-baricitinib levels; HDL-C and apolipoprotein A-I levels remained elevated. CONCLUSIONS Baricitinib was associated with increased LDL-C, HDL-C and triglyceride levels, but did not alter the LDL-C:HDL-C ratio. Evaluation of cardiovascular event rates during long-term treatment is warranted to further characterise these findings and their possible clinical implications. TRIAL REGISTRATION NUMBER NCT00902486, NCT01469013, NCT01185353, NCT01721044, NCT01721057, NCT01711359, NCT01710358, NCT01885078.
Collapse
Affiliation(s)
- Peter C Taylor
- Botnar Research Centre, Nuffield Department ofOrthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Joel M Kremer
- Center for Rheumatology, Albany Medical College, Albany, New York, USA
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | | | | | | | - Lei Chen
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Sarah Witt
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Chadi Saifan
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - James D Otvos
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, North Carolina, USA
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, North Carolina, USA
| | | | | | | | | | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
139
|
Ueland T, Kleveland O, Michelsen AE, Wiseth R, Damås JK, Holven KB, Aukrust P, Gullestad L, Yndestad A, Halvorsen B. Serum lipoprotein(a) is not modified by interleukin-6 receptor antagonism or associated with inflammation in non-ST-elevation myocardial infarction. Int J Cardiol 2018; 274:348-350. [PMID: 29961573 DOI: 10.1016/j.ijcard.2018.06.093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/02/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND The IL-6 receptor antagonist tocilizumab has been shown to attenuate the proatherogenic lipoprotein a [Lp(a)] in rheumatoid arthritis. We evaluated if a single dose of tocilizumab reduced Lp(a) in patients with non-ST-elevation myocardial infarction (NSTEMI). METHODS Lp(a) was assessed by immunoassay (n = 117 patients) at 7 consecutive time-points between day 1 and 3 and at 3 and 6 months follow-up. RESULTS Tocilizumab did not affect Lp(a) at any time-point during the study and was not associated with cardiovascular risk factors. CONCLUSIONS Short-time inhibition of IL-6 with tocilizumab in patients with NSTEMI did not influence Lp(a) levels.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway; K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway.
| | - Ola Kleveland
- Clinic of Cardiology, St. Olavs Hospital, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology NTNU, Trondheim, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway
| | - Rune Wiseth
- Clinic of Cardiology, St. Olavs Hospital, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology NTNU, Trondheim, Norway
| | - Jan Kristian Damås
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology NTNU, Trondheim, Norway
| | - Kirsten B Holven
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity, and Preventive Medicine, Norway; Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Norway; K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
| | - Lars Gullestad
- Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway; Department of Cardiology, Oslo University Hospital Rikshospitalet, Norway; KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway; KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway
| |
Collapse
|
140
|
Giollo A, Bissell LA, Buch MH. Cardiovascular outcomes of patients with rheumatoid arthritis prescribed disease modifying anti-rheumatic drugs: a review. Expert Opin Drug Saf 2018; 17:697-708. [PMID: 29871535 DOI: 10.1080/14740338.2018.1483331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is associated with a heightened risk of cardiovascular disease (CVD), with both traditional CV risk factors and inflammation contributing to this risk. AREAS COVERED This review highlights the burden of CVD in RA and associated traditional CV risk factors, including the complexity of dyslipidemia in RA and the so-called 'lipid paradox.' Furthermore, the recognized RA-disease-specific factors associated with higher risk of CVD and the role of systemic inflammation in the pathogenesis of CVD in RA will be addressed. With the advent of biologic and targeted synthetic therapies in the treatment of RA, the effect of conventional and newer generation disease modifying anti-rheumatic therapies (DMARDs) on CV risk and associated risk factors will also be discussed. EXPERT OPINION Identifying the RA phenotype at greatest risk of CVD, understanding the interplay of increased traditional risk factors, common inflammatory processes and RA-specific factors, and personalized use of DMARDs according to disease phenotype and comorbidity to reduce this risk are key areas for future research.
Collapse
Affiliation(s)
- Alessandro Giollo
- a Leeds Institute of Rheumatic and Musculoskeletal Medicine , University of Leeds, Chapel Allerton Hospital , Leeds , UK.,b NIHR Leeds Biomedical Research Centre , Leeds Teaching Hospitals NHS Trust , Leeds , LS7 4SA , UK.,c Department of Medicine, Rheumatology Unit , University of Verona , Verona , Italy
| | - Lesley-Anne Bissell
- a Leeds Institute of Rheumatic and Musculoskeletal Medicine , University of Leeds, Chapel Allerton Hospital , Leeds , UK.,b NIHR Leeds Biomedical Research Centre , Leeds Teaching Hospitals NHS Trust , Leeds , LS7 4SA , UK
| | - Maya H Buch
- a Leeds Institute of Rheumatic and Musculoskeletal Medicine , University of Leeds, Chapel Allerton Hospital , Leeds , UK.,b NIHR Leeds Biomedical Research Centre , Leeds Teaching Hospitals NHS Trust , Leeds , LS7 4SA , UK
| |
Collapse
|
141
|
Cardiovascular Safety of Biologics and JAK Inhibitors in Patients with Rheumatoid Arthritis. Curr Rheumatol Rep 2018; 20:42. [DOI: 10.1007/s11926-018-0752-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
142
|
Kottoor SJ, Arora RR. The Utility of Anti-Inflammatory Agents in Cardiovascular Disease: A Novel Perspective on the Treatment of Atherosclerosis. J Cardiovasc Pharmacol Ther 2018; 23:483-493. [PMID: 29783850 DOI: 10.1177/1074248418778548] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Approximately 40% of heart attack survivors remain at increased risk of recurrent cardiovascular events, despite the current treatment options showing that atherothrombosis is not exclusively a disorder of lipoprotein aggregation in the arterial wall. Clinical and experimental data suggest that inflammation plays an important role in atherothrombosis independent of the cholesterol level. Acute-phase reactants, such as C-reactive protein, increase in patients with coronary artery disease and are known to predict adverse outcomes in such patients. The recent CANTOS trial published in The New England Journal of Medicine provides evidence that interleukin-1β along with other cytokines play central roles in the inflammatory reaction that drives the interleukin-6 signaling pathway and have profound effects on cardiovascular outcomes. Several other ongoing studies are focused on multiple immune mediators involved in this process to support the inflammatory hypothesis of cardiovascular diseases. These new classes of drugs could represent the biggest breakthrough in cardiovascular medicine, which could have the greatest impact on cardiovascular mortality since the advent of statins. The drug canakinumab has shown promise in lowering atherosclerosis, and other drugs, such as colchicine and methotrexate, are gaining interest and are being investigated in multiple ongoing trials. A major concern is the affordability of these drugs, as most cardiovascular diseases are noted among people of lower socioeconomic statuses. The LoDoCo trial showed some benefits of colchicine, and whether this old drug can be marketed with a new label for cardiovascular disease remains in question. Therefore, a clear understanding of the different inflammatory pathways involved in atherosclerosis is needed to help develop more effective treatment modalities that will benefit humankind.
Collapse
Affiliation(s)
- Santhosh J Kottoor
- 1 Department of Cardiology, Samaritan Heart Institute, Pazhanganad, Ernakulam, Kerala, India
| | - Rohit R Arora
- 2 Department of Medicine, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA
| |
Collapse
|
143
|
Nurmohamed M, Choy E, Lula S, Kola B, DeMasi R, Accossato P. The Impact of Biologics and Tofacitinib on Cardiovascular Risk Factors and Outcomes in Patients with Rheumatic Disease: A Systematic Literature Review. Drug Saf 2018; 41:473-488. [PMID: 29318514 PMCID: PMC5938314 DOI: 10.1007/s40264-017-0628-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Rheumatic diseases are autoimmune, inflammatory diseases often associated with cardiovascular (CV) disease, a major cause of mortality in these patients. In recent years, treatment with biologic and targeted synthetic disease-modifying anti-rheumatic drugs (DMARDs), either as monotherapy or in combination with other drugs, have become the standard of treatment. In this systematic literature review, we evaluated the effect of treatment with biologic or tofacitinib on the CV risk and outcomes in these patients. METHODS A systematic search was performed in MEDLINE, Embase, the Cochrane Central Register of Controlled Trials, and Cochrane Database of Systematic Reviews for articles reporting on CV risk and events in patients with rheumatic disease treated with a biologic agent or tofacitinib. Articles identified were subjected to two levels of screening. Articles that passed the first level based on title and abstract were assessed on full-text evaluation. The quality of randomized clinical trials was assessed by Jadad scoring system and the quality of the other studies and abstracts was assessed using the Downs and Black instrument. The data extracted included study design, baseline patient characteristics, and measurements of CV risk and events. RESULTS Of the 5722 articles identified in the initial search, screening yielded 105 unique publications from 90 unique studies (33 clinical trials, 39 prospective cohort studies, and an additional 18 retrospective studies) that reported CV risk outcomes. A risk of bias analysis for each type of report indicated that they were of good or excellent quality. Importantly, despite some limitations in data reported, there were no indications of significant increase in adverse CV events or risk in response to treatment with the agents evaluated. CONCLUSIONS Treatment with biologic or tofacitinib appears to be well-tolerated with respect to CV outcomes in these patients.
Collapse
Affiliation(s)
- Michael Nurmohamed
- Department of Rheumatology, Reade, VU University Medical Center, 3A50, Amsterdam Rheumatology and Immunology Center, Dr. Jan van Breemenstraat 2, 1056 AB, Amsterdam, The Netherlands.
| | - Ernest Choy
- Cardiff University School of Medicine, Cardiff, Wales, UK
| | - Sadiq Lula
- Envision Pharma Group, London, UK
- IQVIA, London, UK
| | | | | | | |
Collapse
|
144
|
Effects of tumor necrosis factor inhibitors and tocilizumab on the glycosylated hemoglobin levels in patients with rheumatoid arthritis; an observational study. PLoS One 2018; 13:e0196368. [PMID: 29694426 PMCID: PMC5918963 DOI: 10.1371/journal.pone.0196368] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/11/2018] [Indexed: 12/13/2022] Open
Abstract
Rheumatoid arthritis (RA) and diabetes mellitus (DM) are associated with inflammation. We tried to investigate the influence of tumor necrosis factor inhibitors (TNFi) and tocilizumab (TCZ) on the glucose metabolism of RA patients. RA patients in whom treatment with TNFi or TCZ was initiated from 2008 to 2015 were studied based on their medical records. We analyzed patients whose glycosylated hemoglobin (HbA1c) levels were measured both before and 3 months after the initiation of these biologic agents. The association between HbA1c reduction and the treatment was evaluated. From 971 cases treated with these biologic agents, 221 cases whose medical records of HbA1c were available, were included (TNFi, n = 154; TCZ, n = 67). Both the TNFi and TCZ groups had significantly lower HbA1c values at 1 month and 3 months after the initiation of treatment (TNFi, p<0.001; TCZ, p<0.001). Although the pretreatment HbA1c values did not differ (TNFi, 6.2%; TCZ, 6.2%; p = 0.532), the 3-month treatment HbA1c values were lower (TNFi, 6.1%; TCZ, 5.8%; p = 0.010) and the changes in HbA1c (ΔHbA1c) were greater (TNFi, 0.1%; TCZ, 0.4%; p<0.001) in the TCZ group. The reduction of HbA1c—defined by the achievement of a ΔHbA1c of ≥0.5%—was associated with baseline diagnosis of diabetes mellitus, baseline diabetes treatment, hospitalization, medical change during the observation period, and TCZ. In the multivariate logistic regression analysis, TCZ was associated with the reduction of HbA1c in comparison to TNFi (adjusted OR = 5.59, 95% CI = 2.56–12.2; p<0.001). The HbA1c levels in RA patients were significantly lower after the initiation of TNFi or TCZ. Our study suggests that TCZ decreases the HbA1c levels in RA patients to a greater extent than TNFi.
Collapse
|
145
|
England BR, Thiele GM, Anderson DR, Mikuls TR. Increased cardiovascular risk in rheumatoid arthritis: mechanisms and implications. BMJ 2018; 361:k1036. [PMID: 29685876 PMCID: PMC6889899 DOI: 10.1136/bmj.k1036] [Citation(s) in RCA: 305] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Rheumatoid arthritis is a systemic autoimmune disease characterized by excess morbidity and mortality from cardiovascular disease. Mechanisms linking rheumatoid arthritis and cardiovascular disease include shared inflammatory mediators, post-translational modifications of peptides/proteins and subsequent immune responses, alterations in the composition and function of lipoproteins, increased oxidative stress, and endothelial dysfunction. Despite a growing understanding of these mechanisms and their complex interplay with conventional cardiovascular risk factors, optimal approaches of risk stratification, prevention, and treatment in the context of rheumatoid arthritis remain unknown. A multifaceted approach to reduce the burden posed by cardiovascular disease requires optimal management of traditional risk factors in addition to those intrinsic to rheumatoid arthritis such as increased disease activity. Treatments for rheumatoid arthritis seem to exert differential effects on cardiovascular risk as well as the mechanisms linking these conditions. More research is needed to establish whether preferential rheumatoid arthritis therapies exist in terms of prevention of cardiovascular disease. Ultimately, understanding the unique mechanisms for cardiovascular disease in rheumatoid arthritis will aid in risk stratification and the identification of novel targets for meaningful reduction of cardiovascular risk in this patient population.
Collapse
Affiliation(s)
- Bryant R England
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Daniel R Anderson
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ted R Mikuls
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
146
|
Towards the Clinical Management of Cardiac Involvement in Systemic Inflammatory Conditions—a Central Role for CMR. CURRENT CARDIOVASCULAR IMAGING REPORTS 2018. [DOI: 10.1007/s12410-018-9451-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
147
|
Sanmartí R, Ruiz-Esquide V, Bastida C, Soy D. Tocilizumab in the treatment of adult rheumatoid arthritis. Immunotherapy 2018; 10:447-464. [PMID: 29495891 DOI: 10.2217/imt-2017-0173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is the most prevalent immune-mediated chronic rheumatic disease and is associated with joint destruction and disability. Therapeutic strategies, including biological disease-modifying antirheumatic drugs (bDMARDs) have improved the prognosis and quality of life of RA patients. Tocilizumab (TCZ) is a humanized monoclonal antibody against IL-6 receptor licensed in 2009 that has demonstrated clinical efficacy in various adult RA populations. RA management guidelines and recommendations consider TCZ as one of the bDMARDS indicated after methotrexate or other conventional synthetic DMARDs and/or TNF inhibitors failure in adult RA. Of particular interest is the demonstration of its effectiveness in monotherapy in comparison with other bDMARDs. Recent observational studies have shown good results for the safety profile of TCZ with no new alert signals.
Collapse
Affiliation(s)
- Raimon Sanmartí
- Arthritis Unit, Rheumatology Service, Hospital Clinic of Barcelona, Barcelona, Catalonia, Spain
| | - Virginia Ruiz-Esquide
- Arthritis Unit, Rheumatology Service, Hospital Clinic of Barcelona, Barcelona, Catalonia, Spain
| | - Carla Bastida
- Pharmacy Service, Hospital Clinic of Barcelona, Barcelona, Catalonia, Spain
| | - Dolor Soy
- Pharmacy Service, Hospital Clinic of Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
148
|
Chistiakov DA, Melnichenko AA, Grechko AV, Myasoedova VA, Orekhov AN. Potential of anti-inflammatory agents for treatment of atherosclerosis. Exp Mol Pathol 2018; 104:114-124. [PMID: 29378168 DOI: 10.1016/j.yexmp.2018.01.008] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 12/30/2017] [Accepted: 01/17/2018] [Indexed: 12/23/2022]
Abstract
Chronic inflammation is a central pathogenic mechanism of atherosclerosis induction and progression. Vascular inflammation is associated with accelerated onset of late atherosclerosis complications. Atherosclerosis-related inflammation is mediated by a complex cocktail of pro-inflammatory cytokines, chemokines, bioactive lipids, and adhesion molecules, and blocking the key pro-atherogenic inflammatory mechanisms can be beneficial for treatment of atherosclerosis. Therapeutic agents that specifically target some of the atherosclerosis-related inflammatory mechanisms have been evaluated in preclinical and clinical studies. The most promising anti-inflammatory compounds for treatment of atherosclerosis include non-specific anti-inflammatory drugs, phospholipase inhibitors, blockers of major inflammatory cytokines, leukotrienes, adhesion molecules, and pro-inflammatory signaling pathways, such as CCL2-CCR2 axis or p38 MAPK pathway. Ongoing studies attempt evaluating therapeutic utility of these anti-inflammatory drugs for treatment of atherosclerosis. The obtained results are important for our understanding of atherosclerosis-related inflammatory mechanisms and for designing randomized controlled studies assessing the effect of specific anti-inflammatory strategies on cardiovascular outcomes.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Neurochemistry, Division of Basic and Applied Neurobiology, Serbsky Federal Medical Research Center of Psychiatry and Narcology, Moscow 119991, Russia
| | - Alexandra A Melnichenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia
| | - Andrey V Grechko
- Federal Scientific Clinical Center for Resuscitation and Rehabilitation, Moscow 109240, Russia
| | - Veronika A Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow 121609, Russia.
| |
Collapse
|
149
|
Imamura H, Momohara S, Yano K, Sakuma Y, Nakayama M, Tobimatsu H, Ikari K. Tocilizumab treatment in patients with rheumatoid arthritis is associated with reduced fibrinogen levels and increased blood loss after total knee arthroplasty. Mod Rheumatol 2018; 28:976-980. [PMID: 29325462 DOI: 10.1080/14397595.2018.1428041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Since IL-6 has been associated with activation of the coagulation cascade and upregulation of fibrinogen transcription, we retrospectively tested the hypothesis that patients with rheumatoid arthritis (RA) treated with tocilizumab (TCZ) may lose more blood when undergoing total knee arthroplasty (TKA). METHODS This study included 115 RA patients who underwent primary TKA and were preoperatively tested for fibrinogen levels. The blood volume of each patient was calculated using the Nadler formula, and estimated blood loss after TKA was calculated as the change between pre-operative and post-operative hematocrits. If salvaged blood was reinfused, the volume was measured and added to the volume of the estimated blood loss. RESULTS We observed that patients treated with TCZ had significantly lower pre-operative fibrinogen levels than those not treated with TCZ (190.0 mg/dL versus 347.0 mg/dL, respectively; p = .00018). We also observed a statistically significant increase in mean total volume of estimated blood loss after TKA in RA patients who had been treated with TCZ compared with those not treated with TCZ (797.1 mL versus 511.4 mL, respectively; p = .0039). CONCLUSION TCZ treatment in patients with RA may increase the risk of blood loss after TKA because of decreased fibrinogen levels.
Collapse
Affiliation(s)
- Hitoshi Imamura
- a Department of Orthopedic Surgery , Institute of Rheumatology, Tokyo Women's Medical University , Shinjuku , Tokyo , Japan
| | - Shigeki Momohara
- a Department of Orthopedic Surgery , Institute of Rheumatology, Tokyo Women's Medical University , Shinjuku , Tokyo , Japan.,b Hakkeikai Incorporated Medical Institution , Kusanagi , Shizuoka , Japan
| | - Koichiro Yano
- a Department of Orthopedic Surgery , Institute of Rheumatology, Tokyo Women's Medical University , Shinjuku , Tokyo , Japan
| | - Yu Sakuma
- a Department of Orthopedic Surgery , Institute of Rheumatology, Tokyo Women's Medical University , Shinjuku , Tokyo , Japan
| | - Masanori Nakayama
- a Department of Orthopedic Surgery , Institute of Rheumatology, Tokyo Women's Medical University , Shinjuku , Tokyo , Japan
| | - Haruki Tobimatsu
- a Department of Orthopedic Surgery , Institute of Rheumatology, Tokyo Women's Medical University , Shinjuku , Tokyo , Japan
| | - Katsunori Ikari
- a Department of Orthopedic Surgery , Institute of Rheumatology, Tokyo Women's Medical University , Shinjuku , Tokyo , Japan
| |
Collapse
|
150
|
Currie G, Delles C. Precision Medicine and Personalized Medicine in Cardiovascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1065:589-605. [PMID: 30051409 DOI: 10.1007/978-3-319-77932-4_36] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Precision medicine aims to offer "the right treatment to the right patient at the right time." In cardiovascular medicine the potential of precision medicine applies to all stages of the disease development and includes risk prediction, preventative measures, and targeted therapeutic approaches. Precision medicine will benefit from new developments in the area of genomics and other omics but equally heavily depends on established biomarkers, functional tests, and imaging. Cardiovascular medicine often relies on noninvasive diagnostic procedures and symptom-based disease management. In contrast, other clinical disciplines including oncology and immunology have already moved to molecular diagnostics that lend themselves to precision medicine approaches. There are opportunities to implement precision medicine approaches by focusing on common diseases such as hypertension, conditions with diagnostic and prognostic uncertainty such as angina, and conditions that are associated with high mortality and involve costly and potentially harmful interventions such as dilated cardiomyopathy and cardiac resynchronization therapy. Sex and gender issues have not yet been fully explored in precision medicine although the opportunity to use molecular data to more accurately manage men and women with cardiovascular disease has been acknowledged. A mindshift is required in order to fully exploit the potential of precision medicine to tackle the global burden of cardiovascular diseases.
Collapse
Affiliation(s)
- Gemma Currie
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, Glasgow, Scotland, UK
| | - Christian Delles
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, Glasgow, Scotland, UK.
| |
Collapse
|