1
|
Alebna PL, Ambrosio M, Martin M, Martey S, Spitz JA, Sharma G, Van Tassell B, Dixon DL, Hundley WG, Salloum FN, Mehta A. Association of Lipoprotein(a) with cardiovascular events among individuals with autoimmune conditions. Atherosclerosis 2025; 406:119244. [PMID: 40398292 DOI: 10.1016/j.atherosclerosis.2025.119244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Autoimmune conditions are associated with systemic inflammation, which elevates the risk of major adverse cardiovascular events (MACE). Systemic inflammation can increase plasma lipoprotein(a) [Lp(a)] levels by activating the interleukin-6 response element within the LPA gene promoter region. However, the association between elevated plasma Lp(a) and MACE risk in individuals with autoimmune conditions remains unclear. METHODS We analyzed data from 353,035 UK Biobank participants without cardiovascular disease, including 11,229 individuals with autoimmune conditions such as systemic lupus erythematosus, rheumatoid arthritis, psoriasis, multiple sclerosis, and others. Cox proportional hazards regression models and Kaplan-Meier survival curves assessed the association between elevated Lp(a) (≥125 nmol/L), autoimmune status, and time to MACE (myocardial infarction, stroke, cardiovascular death). RESULTS Over a median follow-up of 14 years, 19,091 MACEs occurred. Autoimmune conditions (HR, 1.30; 95 % CI, 1.20-1.41; P < 0.001) and elevated Lp(a) (HR, 1.24; 95 % CI, 1.18-1.30; P < 0.001) were independently associated with increased MACE risk. The interaction between autoimmune conditions and elevated Lp(a) was not significant (p = 0.40). Compared to participants with neither risk factor, those with both autoimmune conditions and elevated Lp(a) had the highest MACE risk (HR, 1.77; 95 % CI, 1.46-2.15; P < 0.001). Elevated MACE risk was also observed in individuals with only elevated Lp(a) (HR, 1.23; 95 % CI, 1.17-1.29; P < 0.001) or only autoimmune conditions (HR, 1.28; 95 % CI, 1.18-1.40; P < 0.001). CONCLUSION Autoimmune disease status and elevated Lp(a) level have an independent and additive joint association with MACE risk. This may have implications for Lp(a) lowering therapy use in high-risk primary prevention populations.
Collapse
Affiliation(s)
- Pamela L Alebna
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Mathew Ambrosio
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Maxwell Martin
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Sarah Martey
- School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jared A Spitz
- Inova Schar Heart and Vascular, Inova Health, Fairfax, VA, USA
| | - Garima Sharma
- Inova Schar Heart and Vascular, Inova Health, Fairfax, VA, USA
| | - Benjamin Van Tassell
- Department of Pharmacotherapy & Outcomes Science, VCU School of Pharmacy, Richmond, VA, USA
| | - Dave L Dixon
- Department of Pharmacotherapy & Outcomes Science, VCU School of Pharmacy, Richmond, VA, USA
| | - W Gregory Hundley
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Fadi N Salloum
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Anurag Mehta
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
2
|
Cacciatore S, Andaloro S, Bernardi M, Oterino Manzanas A, Spadafora L, Figliozzi S, Asher E, Rana JS, Ecarnot F, Gragnano F, Calabrò P, Gallo A, Andò G, Manzo-Silberman S, Roeters van Lennep J, Tosato M, Landi F, Biondi-Zoccai G, Marzetti E, Sabouret P. Chronic Inflammatory Diseases and Cardiovascular Risk: Current Insights and Future Strategies for Optimal Management. Int J Mol Sci 2025; 26:3071. [PMID: 40243756 PMCID: PMC11989023 DOI: 10.3390/ijms26073071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Chronic inflammation is a pivotal driver in the progression of atherosclerosis, significantly contributing to the burden of cardiovascular disease (CVD). Patients with chronic inflammatory diseases, such as inflammatory bowel diseases (IBDs) (e.g., ulcerative colitis and Crohn's disease), rheumatological disorders, as well as individuals with auto-immune diseases (such as systemic lupus erythematosus), present a higher risk of major adverse cardiac events (MACEs). Despite their elevated CVD risk, these populations remain underrepresented in cardiovascular research, leading to a critical underestimation of their cardiovascular risk (CVR) in clinical practice. Furthermore, even recent CVR scores poorly predict the risk of events in these specific populations. This narrative review examines the physiopathological mechanisms linking chronic inflammation, immunomodulation, atherosclerosis, thrombosis and cardiovascular events. We review data from epidemiological studies and clinical trials to explore the potential cardiovascular benefits of anti-inflammatory and immunomodulatory therapies. Despite existing evidence, significant gaps in knowledge remain. Future research is mandatory, focusing on innovative strategies for risk stratification and optimization, including lipidomics, proteomics, advanced inflammatory markers, microbiota profiling, and cardiovascular imaging. Addressing these unmet needs will enhance understanding of cardiovascular risk in chronic inflammatory diseases, enabling tailored interventions and better outcomes.
Collapse
Affiliation(s)
- Stefano Cacciatore
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Silvia Andaloro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
| | - Marco Bernardi
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy; (M.B.); (L.S.); (G.B.-Z.)
| | - Armando Oterino Manzanas
- Department of Cardiology, Hospital Universitario de Salamanca-IBSAL, Paseo de San Vicente, 58-182, 37007 Salamanca, Spain;
| | - Luigi Spadafora
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy; (M.B.); (L.S.); (G.B.-Z.)
| | - Stefano Figliozzi
- IRCCS Humanitas Research Hospital, Via Alessandro Manzoni, 56, Rozzano, 20089 Milano, Italy;
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, Pieve Emanuele, 20090 Milano, Italy
| | - Elad Asher
- Jesselson Integrated Heart Center, The Eisenberg R&D Authority, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Shmuel (Hans) Beyth St. 12, Jerusalem 9103102, Israel;
| | - Jamal S. Rana
- Division of Cardiology, Kaiser Permanente Northern California, 1 Kaiser Plaza, Oakland, CA 94612, USA;
- Division of Research, Kaiser Permanente Northern California, 1 Kaiser Plaza, Oakland, CA 94612, USA
| | - Fiona Ecarnot
- Department of Cardiology, University Hospital, Boulevard Fleming, 25000 Besançon, France;
- SINERGIES Unit, University Marie & Louis Pasteur, 19 Rue Ambroise Paré, 25000 Besançon, France
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Via Leonardo Bianchi, Ospedale Monaldi, 80131 Naples, Italy; (F.G.); (P.C.)
- Division of Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, Via Ferdinando Palasciano, 81100 Caserta, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Via Leonardo Bianchi, Ospedale Monaldi, 80131 Naples, Italy; (F.G.); (P.C.)
- Division of Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, Via Ferdinando Palasciano, 81100 Caserta, Italy
| | - Antonio Gallo
- INSERM UMR1166, IHU ICAN, Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, Pitié-Salpêtrière Hospital, Sorbonne University, AP-HP, 47–83 Bd de l’Hôpital, 75013 Paris, France;
| | - Giuseppe Andò
- Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “Gaetano Martino”, Via Consolare Valeria, 1, 98124 Messina, Italy;
| | - Stephane Manzo-Silberman
- ACTION Study Group, Inserm UMRS1166, Heart Institute, Pitié-Salpetriere Hospital, Sorbonne University, 47-83 Bd de l’Hôpital, 75013 Paris, France; (S.M.-S.); (P.S.)
| | - Jeanine Roeters van Lennep
- Department of Internal Medicine, Cardiovascular Institute, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands;
| | - Matteo Tosato
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy; (M.B.); (L.S.); (G.B.-Z.)
- Maria Cecilia Hospital, GVM Care & Research, Via Corriera, 1, 48033 Cotignola, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Pierre Sabouret
- ACTION Study Group, Inserm UMRS1166, Heart Institute, Pitié-Salpetriere Hospital, Sorbonne University, 47-83 Bd de l’Hôpital, 75013 Paris, France; (S.M.-S.); (P.S.)
| |
Collapse
|
3
|
Ti Y, Xu D, Qin X, Hu Y, Xu Y, Zhao Q, Bu P, Li J. Mendelian randomization analysis does not support a causal influence between lipoprotein(A) and immune-mediated inflammatory diseases. Sci Rep 2025; 15:3834. [PMID: 39885280 PMCID: PMC11782540 DOI: 10.1038/s41598-025-88375-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/28/2025] [Indexed: 02/01/2025] Open
Abstract
Observational studies have reported an association between lipoprotein(a) (Lp(a)) and immune-mediated inflammatory diseases (IMIDs). This study used Mendelian Randomization (MR) and multivariable MR (MVMR) to explore the causal relationship between lipoprotein(a) [Lp(a)] and immune-mediated inflammatory diseases (IMIDs). We performed a bidirectional two-sample mendelian randomization analyses based on genome-wide association study (GWAS) summary statistics of Lp(a) and nine IMIDs, specifically celiac disease (CeD), Crohn's disease (CD), ulcerative colitis (UC), inflammatory bowel disease (IBD), multiple sclerosis (MS), psoriasis (Pso), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D), and summary-level data for lipid traits. Furthermore, we performed MVMR to examine the independence of relationship between Lp(a) and IMIDs after controlling other lipid traits, namely high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C) and triglycerides (TG). We didn't observe a causal association between Lp(a) and the risk of IMIDs in univariable and multivariable MR analysis, challenging previous observational studies. However, genetically predicted lipid traits HDL-C was associated with increased risk of Type 1 diabetes (T1D). The identification of potential mechanisms underlying the observed associations in observational studies necessitates further investigation.
Collapse
Affiliation(s)
- Yun Ti
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Dan Xu
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoning Qin
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yang Hu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuru Xu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qingzhao Zhao
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Peili Bu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jingyuan Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
4
|
Kattamuri L, Duggal S, Aparece JP, Sairam S. Cardiovascular Risk Factor and Atherosclerosis in Rheumatoid Arthritis (RA). Curr Cardiol Rep 2025; 27:31. [PMID: 39831939 DOI: 10.1007/s11886-025-02198-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE OF REVIEW To highlight advancements in managing traditional and rheumatoid arthritis (RA) specific risk factors and the impact of RA treatments on cardiovascular outcomes. RECENT FINDINGS Advancements in rheumatoid arthritis management have paralleled declining trends in cardiovascular disease risks. Biomarkers like CRP, Lipoprotein(a), Apolipoprotein B 100, and imaging tools such as coronary artery calcium scoring enhance cardiovascular risk stratification, particularly in intermediate-risk RA patients. While effective RA treatments, have demonstrated substantial cardiovascular benefits, subclass differences were noted in high-risk patients. Increased risk of cardiovascular disease is driven by chronic inflammation, altered lipid metabolism, and traditional risk factors. Effective RA treatment significantly lowers cardiovascular events. Standard treatment of hypertension, diabetes and hypercholesterolemia are effective and lowers RA disease activity and inflammatory markers. While RA is considered a risk enhancing state in calculating CV risk scores, currently there exists no RA disease -specific blood pressure, blood sugar or lipid targets.
Collapse
Affiliation(s)
- Lakshmi Kattamuri
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, 4800 Alberta Ave, El Paso, TX, 79935, USA
| | - Shivangini Duggal
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, 4800 Alberta Ave, El Paso, TX, 79935, USA
| | - John Paul Aparece
- Department of Internal Medicine, Texas Tech University Health Sciences Center El Paso, Paul L. Foster School of Medicine, 4800 Alberta Ave, El Paso, TX, 79935, USA
| | - Shrilekha Sairam
- Division of Rheumatology, Department of Internal Medicine, Texas Tech Health Sciences Center El Paso, Paul L. Foster School of Medicine, El Paso, TX, USA.
| |
Collapse
|
5
|
Lian PA, Zhu WQ, Zhao WX, Huang PP, Ran JL, Tang YX, Huang XS, Li R. Lipoprotein(a) in atherosclerotic cardiovascular disease and proprotein convertase subtilisin/kexin-type 9 inhibitors. Clin Chim Acta 2025; 565:119982. [PMID: 39366516 DOI: 10.1016/j.cca.2024.119982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
High plasma lipoprotein(a) (Lp(a)) levels increase the cardiovascular risk in populations with atherosclerotic cardiovascular disease (ASCVD). Apolipoprotein (a) [apo(a)], a unique protein component of Lp(a), plays an important role in the pathogenesis of atherosclerosis. Statins, the primary medication in managing ASCVD, lower low-density lipoprotein cholesterol (LDL-C) but concurrently elevate plasma Lp(a) levels, contributing to an increased residual cardiovascular risk. In turn, proprotein convertase subtilisin/kexin-type 9 (PCSK9) inhibitors, a novel class of LDL-C lowering drugs, effectively reduce plasma Lp(a) levels, which is believed to decrease residual cardiovascular risk. However, the mechanism by which PCSK9 inhibitors reduce Lp(a) levels remains unknown. In addition, there are some clinical limitations of PCSK9 inhibitors. Here, we systematically review the past, present, and prospects of studies pertaining to Lp(a), PCSK9 inhibitors, and ASCVD.
Collapse
Affiliation(s)
- Ping-An Lian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen-Qiang Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei-Xin Zhao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Piao-Piao Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan-Li Ran
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya-Xin Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xian-Sheng Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cardiovascular Medicine, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China
| | - Rong Li
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Stomatology, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China.
| |
Collapse
|
6
|
Fichtner I, Macchi C, Rizzuto AS, Carugo S, Corsini A, Ruscica M. Lipoprotein(a) and the atherosclerotic burden - Should we wait for clinical trial evidence before taking action? ATHEROSCLEROSIS PLUS 2024; 58:16-23. [PMID: 39435317 PMCID: PMC11492331 DOI: 10.1016/j.athplu.2024.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/23/2024]
Abstract
The fact that lipoprotein(a) levels should be regarded as a causal residual risk factor in the atherosclerotic cardiovascular diseases (ASCVD) is now a no-brainer. This review article aims to summarize the latest evidence supporting the causal role of lipoprotein(a) in ASCVD and the potential strategies to reduce the lipoprotein(a) burden until clinical trial results are available. Epidemiological and genetic data demonstrate the causal link between lipoprotein(a) and increased ASCVD risk. That being said, a specific question comes to mind: "must we wait for outcome trials in order to take action?". Given that lipoprotein(a) levels predict incident ASCVD in both primary and secondary prevention contexts, with a linear risk gradient across its distribution, measuring lipoprotein(a) can unequivocally help identify patients who may later benefit from specific lipoprotein(a)-lowering therapies. This understanding has led various National Societies to recommend dosing lipoprotein(a) in high-risk individuals and to support the recommendation of measuring lipoprotein(a) levels at least once in every adult for risk stratification.
Collapse
Affiliation(s)
- Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | | | - Stefano Carugo
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
7
|
Kozieł-Siołkowska M, Mitręga K, Podolecki T, Olma A, Kalarus Z, Streb W. Lipoprotein(a) as an Independent Predictor of Elevated SYNTAX Score. J Clin Med 2024; 13:7109. [PMID: 39685569 DOI: 10.3390/jcm13237109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Increased lipoprotein(a) [Lp(a)] level is associated with elevated possibility of atherosclerosis progression. SYNTAX score enables to grade the anatomy of coronary arteries. To identify the impact of increased Lp(a) level on SYNTAX score in individuals with acute myocardial infarction (AMI). Methods: In our analysis, we enrolled 173 consecutive adult patients hospitalized for AMI in a tertiary cardiology center from December 2022 to August 2023. Patient characteristics were compared for patients with SYNTAX score ≥ 23 (64 patients) and SYNTAX score < 23 (109 patients). The SYNTAX score was estimated based on the results of coronary angiography. Logistic regression analyses were performed to evaluate the factors associated with SYNTAX score. Results: Individuals with the SYNTAX score ≥ 23 were more likely to have arterial hypertension, diabetes mellitus, significant stenosis in the left main coronary artery, and higher Lp(a) levels than those with SYNTAX < 23 (all p < 0.05). On univariate analysis, age (OR 1.05, 95% CI 1.02-1.08, p = 0.001), Lp(a) levels (OR 1.04, 95% CI 1.01-1.06, p = 0.001), and arterial hypertension (OR 2.69, 95% CI 1.26-5.74, p = 0.011) were associated with SYNTAX score ≥ 23. Multivariable determinants of SYNTAX score ≥ 23 were as follows: Lp(a) levels (OR 1.03, 95% CI 1.01-1.08, p = 0.029), and age (OR 1.04, 95% CI 1.01-1.07, p = 0.005). The cut-off value for Lp(a) 166.16 nmol/L identifies patients with SYNTAX score ≥ 23 with 97% sensitivity and 44% specificity (area under curve 0.78, p < 0.001). Conclusions: Elevated Lp(a) concentration is associated with a higher SYNTAX score. A cut-off value of Lp(a) above 166.16 nmol/L allows us to identify subjects with SYNTAX score ≥ 23 with good specificity and sensitivity.
Collapse
Affiliation(s)
- Monika Kozieł-Siołkowska
- 1st Department of Cardiology and Angiology, Silesian Center for Heart Diseases, 41-800 Zabrze, Poland
| | - Katarzyna Mitręga
- 1st Department of Cardiology and Angiology, Silesian Center for Heart Diseases, 41-800 Zabrze, Poland
| | - Tomasz Podolecki
- Department of Cardiology, Congenital Heart Diseases and Electrotherapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | - Anna Olma
- 1st Department of Cardiology and Angiology, Silesian Center for Heart Diseases, 41-800 Zabrze, Poland
| | - Zbigniew Kalarus
- Department of Cardiology, Congenital Heart Diseases and Electrotherapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | - Witold Streb
- Department of Cardiology, Congenital Heart Diseases and Electrotherapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
8
|
Ma Z, Zhong J, Tu W, Li S, Chen J. The functions of apolipoproteins and lipoproteins in health and disease. MOLECULAR BIOMEDICINE 2024; 5:53. [PMID: 39465476 PMCID: PMC11513782 DOI: 10.1186/s43556-024-00218-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
Lipoproteins and apolipoproteins are crucial in lipid metabolism, functioning as essential mediators in the transport of cholesterol and triglycerides and being closely related to the pathogenesis of multiple systems, including cardiovascular. Lipoproteins a (Lp(a)), as a unique subclass of lipoproteins, is a low-density lipoprotein(LDL)-like particle with pro-atherosclerotic and pro-inflammatory properties, displaying high heritability. More and more strong evidence points to a possible link between high amounts of Lp(a) and cardiac conditions like atherosclerotic cardiovascular disease (ASCVD) and aortic stenosis (AS), making it a risk factor for heart diseases. In recent years, Lp(a)'s role in other diseases, including neurological disorders and cancer, has been increasingly recognized. Although therapies aimed at low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) have achieved significant success, elevated Lp(a) levels remain a significant clinical management problem. Despite the limited efficacy of current lipid-lowering therapies, major clinical advances in new Lp(a)-lowering therapies have significantly advanced the field. This review, grounded in the pathophysiology of lipoproteins, seeks to summarize the wide-ranging connections between lipoproteins (such as LDL-C and HDL-C) and various diseases, alongside the latest clinical developments, special emphasis is placed on the pivotal role of Lp(a) in cardiovascular disease, while also examining its future potential and mechanisms in other conditions. Furthermore, this review discusses Lp(a)-lowering therapies and highlights significant recent advances in emerging treatments, advocates for further exploration into Lp(a)'s pathogenic mechanisms and its potential as a therapeutic target, proposing new secondary prevention strategies for high-risk individuals.
Collapse
Affiliation(s)
- Zijun Ma
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, 430030, Hubei, China
| | - Wei Tu
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Shiliang Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun Chen
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China.
| |
Collapse
|
9
|
Taylor PC, Feist E, Pope JE, Nash P, Sibilia J, Caporali R, Balsa A. What have we learnt from the inhibition of IL-6 in RA and what are the clinical opportunities for patient outcomes? Ther Adv Musculoskelet Dis 2024; 16:1759720X241283340. [PMID: 39444594 PMCID: PMC11497505 DOI: 10.1177/1759720x241283340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/27/2024] [Indexed: 10/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterised by persistent inflammation of the synovial joints as well as other tissues and organs. Left untreated, it can lead to joint damage, disability and even increased mortality. The disease is driven by inflammatory cytokines that contribute to the chronic inflammation seen in RA. Interleukin-6 (IL-6) is a key pathological cytokine and a target for treatments aiming to alleviate local and systemic inflammation. Despite advances in understanding RA and the introduction of new treatments, achieving sustained remission remains challenging. This review explores the role of IL-6 in RA pathogenesis, its potential as a treatment target and the significance of personalised medicine in RA management. IL-6 has a dual signalling mechanism, classical and trans-signalling, which influences various intracellular pathways. While several targeted therapies have emerged, no single mechanism-based therapy is universally effective due to the diversity and complexity of the disease. Different approaches to targeting IL-6 have been tested, including biologic blockade of receptors or ligands, and inhibition of IL-6 signalling. IL-6 receptor inhibitors have been validated as RA therapeutics, either alone or in combination with other treatments. Tocilizumab, the first approved IL-6 inhibitor, blocks both soluble and membrane-bound IL-6 receptors, reducing the inflammatory cascade. Clinical trials confirm the efficacy and safety of tocilizumab and its role as a treatment option for patients unresponsive to conventional therapies. The benefits of IL-6 inhibition extend beyond reduced joint inflammation to the amelioration of comorbidities like anaemia, cardiovascular disease, depression and osteoporosis. Tailoring treatment to patients' profiles and comorbidities is essential for optimal outcomes. A 'treat-to-profile' approach, focusing on a holistic view of the patient, could improve personalised medicine strategies. Biosimilars - lower-cost alternatives to biologics - further enhance the accessibility and cost-effectiveness of treatment. IL-6 inhibitors present a valuable treatment option for RA management, particularly for patients with specific comorbidities.
Collapse
Affiliation(s)
- Peter C. Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Windmill Road, Oxford OX3 7LD, UK
| | - Eugen Feist
- Department of Rheumatology, Helios Clinic Vogelsang-Gommern, Cooperation Partner of the Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Janet E. Pope
- Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Peter Nash
- School of Medicine, Griffith University, Brisbane, QLD, Australia
| | - Jean Sibilia
- Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Roberto Caporali
- Department of Rheumatology and Medical Sciences, ASST Gaetano Pini-CTO, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Alejandro Balsa
- Rheumatology Department, Hospital Universitario La Paz, Madrid, Spain
- Inmuno-Rheumatology Research Group, Hospital La Paz, Institute for Health Research – IdiPAZ, Madrid, Spain
| |
Collapse
|
10
|
Kaur G, Abdelrahman K, Berman AN, Biery DW, Shiyovich A, Huck D, Garshick M, Blankstein R, Weber B. Lipoprotein(a): Emerging insights and therapeutics. Am J Prev Cardiol 2024; 18:100641. [PMID: 38646022 PMCID: PMC11033089 DOI: 10.1016/j.ajpc.2024.100641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/08/2024] [Accepted: 02/24/2024] [Indexed: 04/23/2024] Open
Abstract
The strong association between lipoprotein (a) [Lp(a)] and atherosclerotic cardiovascular disease has led to considerations of Lp(a) being a potential target for mitigating residual cardiovascular risk. While approximately 20 % of the population has an Lp(a) level greater than 50 mg/dL, there are no currently available pharmacological lipid-lowering therapies that have demonstrated substantial reduction in Lp(a). Novel therapies to lower Lp(a) include antisense oligonucleotides and small-interfering ribonucleic acid molecules and have shown promising results in phase 2 trials. Phase 3 trials are currently underway and will test the causal relationship between Lp(a) and ASCVD and whether lowering Lp(a) reduces cardiovascular outcomes. In this review, we summarize emerging insights related to Lp(a)'s role as a risk-enhancing factor for ASCVD, association with calcific aortic stenosis, effects of existing therapies on Lp(a) levels, and variations amongst patient populations. The evolving therapeutic landscape of emerging therapeutics is further discussed.
Collapse
Affiliation(s)
- Gurleen Kaur
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Adam N. Berman
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - David W. Biery
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Albert Einstein College of Medicine, New York, NY, USA
| | - Arthur Shiyovich
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel Huck
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Ron Blankstein
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Brittany Weber
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
11
|
Sharma A, Sharma C, Sharma L, Wal P, Mishra P, Sachdeva N, Yadav S, Vargas De-La Cruz C, Arora S, Subramaniyan V, Rawat R, Behl T, Nandave M. Targeting the vivid facets of apolipoproteins as a cardiovascular risk factor in rheumatoid arthritis. Can J Physiol Pharmacol 2024; 102:305-317. [PMID: 38334084 DOI: 10.1139/cjpp-2023-0259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Mostly, cardiovascular diseases are blamed for casualties in rheumatoid arthritis (RA) patients. Customarily, dyslipidemia is probably the most prevalent underlying cause of untimely demise in people suffering from RA as it hastens the expansion of atherosclerosis. The engagement of inflammatory cytokines like tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), interleukin-6 (IL-6), etc., is crucial in the progression and proliferation of both RA and abnormal lipid parameters. Thus, lipid abnormalities should be monitored frequently in patients with both primary and advanced RA stages. An advanced lipid profile examination, i.e., direct role of apolipoproteins associated with various lipid molecules is a more dependable approach for better understanding of the disease and selecting suitable therapeutic targets. Therefore, studying their apolipoproteins is more relevant than assessing RA patients' altered lipid profile levels. Among the various apolipoprotein classes, Apo A1 and Apo B are primarily being focused. In addition, it also addresses how calculating Apo B:Apo A1 ratio can aid in analyzing the disease's risk. The marketed therapies available to control lipid abnormalities are associated with many other risk factors. Hence, directly targeting Apo A1 and Apo B would provide a better and safer option.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Chakshu Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Pranay Wal
- Pranveer Singh Institute of Technology, Pharmacy, Kanpur, Uttar Pradesh, India
| | - Preeti Mishra
- Raja Balwant Singh Engineering Technical Campus, Bichpuri, Agra, India
| | - Nitin Sachdeva
- Department of Anesthesia, Mediclinic Aljowhara Hospital, Al Ain, United Arab Emirates
| | - Shivam Yadav
- School of Pharmacy, Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
| | - Celia Vargas De-La Cruz
- Department of Pharmacology, Bromatology and Toxicology, Faculty of Pharmacy and Biochemistry, Universidad Nacional Mayor de San Marcos, Lima 15001, Peru
- E-Health Research Center, Universidad de Ciencias y Humanidades, Lima 15001, Peru
| | - Sandeep Arora
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
- Centre for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 600077, India
| | - Ravi Rawat
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun, Uttarakhand, India
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Delhi, India
| |
Collapse
|
12
|
Sulu C, Dedeoglu SE, Gonen B, Hepokur M, Guzel AN, Sahin S, Demir AN, Kara Z, Konukoglu D, Damci T, Gonen MS. Serum Lipoprotein(a) Is Not Associated with Graves' Ophthalmopathy. Metab Syndr Relat Disord 2024; 22:69-76. [PMID: 37883657 DOI: 10.1089/met.2023.0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023] Open
Abstract
Aim: To investigate the relationship of serum lipoprotein(a) [Lp(a)] and other serum lipids with presence of Graves' ophthalmopathy (GO). Methods: A total of 99 consecutive patients diagnosed with Graves' disease (GD), aged 18-65 years, who had not received prior treatment for GO, thyroid surgery, or radioactive iodine therapy, were recruited between June 2020 and July 2022. In addition, 56 healthy controls (HCs) were included as the control group. All patients underwent an ophthalmological examination, and were classified based on the presence of GO into the GO group (n = 45) and no GO group (n = 54). Fasting blood samples were collected from all participants to analyze serum lipid parameters, including Lp(a), total cholesterol, low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, and triglycerides. Results: The median serum levels of Lp(a) were 5.7 [4.3-9.2] in the GO group, 6.7 [3.7-9.9] in the no GO group, and 4.7 [3-7.6] in the HC group. The intergroup comparisons of serum Lp(a) levels showed no significant result. The serum levels of total cholesterol, LDL cholesterol, HDL cholesterol, and triglycerides were also similar between the groups (P > 0.05 for all). However, when analyzing only euthyroid GD patients and the control group, the serum LDL cholesterol levels were found to be significantly higher in the euthyroid GO group [median: 132 interquartile range (IQR) (110-148) mg/dL] than in the HCs [median: 96 IQR (94-118) mg/dL] (P = 0.002). Conclusion: The findings of our study did not support the association between serum Lp(a) levels and GO.
Collapse
Affiliation(s)
- Cem Sulu
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Selin Ece Dedeoglu
- Department of Internal Medicine, Eyupsultan State Hospital, Istanbul, Turkiye
| | - Busenur Gonen
- Department of Ophthalmology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Mustafa Hepokur
- Department of Ophthalmology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Adnan Nuri Guzel
- Department of Internal Medicine, and Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Serdar Sahin
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Ahmet Numan Demir
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Zehra Kara
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Dildar Konukoglu
- Department of Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Taner Damci
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Mustafa Sait Gonen
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| |
Collapse
|
13
|
Makris A, Barkas F, Sfikakis PP, Liberopoulos E, Filippatos TD, Ray KK, Agouridis AP. Lipoprotein(a), Interleukin-6 inhibitors, and atherosclerotic cardiovascular disease: Is there an association? ATHEROSCLEROSIS PLUS 2023; 54:1-6. [PMID: 37720252 PMCID: PMC10500445 DOI: 10.1016/j.athplu.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/09/2023] [Accepted: 09/06/2023] [Indexed: 09/19/2023]
Abstract
Background and aims Lipoprotein(a) [Lp(a)] and interleuking-6 (IL-6), an inflammation biomarker, have been established as distinct targets of the residual atherosclerotic cardiovascular disease (ASCVD) risk. We aimed to investigate the association between them, and the potential clinical implications in ASCVD prevention. Methods A literature search was conducted in PubMed until December 31st, 2022, using relevant keywords. Results Elevated lipoprotein(a) [Lp(a)] levels constitute the most common inherited lipid disorder associated with ASCVD. Although Lp(a) levels are mostly determined genetically by the LPA gene locus, they may be altered by acute conditions of stress and chronic inflammatory diseases. Considering its resemblance with low-density lipoproteins, Lp(a) is involved in atherosclerosis, but it also exerts oxidative, thrombotic, antifibrinolytic and inflammatory properties. The cardiovascular efficacy of therapies lowering Lp(a) by >90% is currently investigated. On the other hand, interleukin (IL)-1b/IL-6 pathway also plays a pivotal role in atherosclerosis and residual ASCVD risk. IL-6 receptor inhibitors [IL-6(R)i] lower Lp(a) by 16-41%, whereas ongoing trials are investigating their potential anti-atherosclerotic effect. The Lp(a)-lowering effect of IL-6(R)i might be attributed to the inhibition of the IL-6 response elements in the promoter region of the LPA gene. Conclusions Although the effect of IL-6(R)i on Lp(a) levels is inferior to that of available Lp(a)-lowering therapies, the dual effect of the former on both inflammation and apolipoprotein (a) synthesis may prove of equal or even greater significance when it comes ASCVD outcomes. More trials are required to establish IL-6(R)i in ASCVD prevention and elucidate their interplay with Lp(a) as well as its clinical significance.
Collapse
Affiliation(s)
- Anastasios Makris
- School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Fotios Barkas
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Petros P. Sfikakis
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Evangelos Liberopoulos
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | | | - Kausik K. Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Aris P. Agouridis
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Department of Internal Medicine, German Oncology Center, Limassol, Cyprus
| |
Collapse
|
14
|
Popescu D, Rezus E, Badescu MC, Dima N, Seritean Isac PN, Dragoi IT, Rezus C. Cardiovascular Risk Assessment in Rheumatoid Arthritis: Accelerated Atherosclerosis, New Biomarkers, and the Effects of Biological Therapy. Life (Basel) 2023; 13:life13020319. [PMID: 36836675 PMCID: PMC9965162 DOI: 10.3390/life13020319] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Rheumatoid arthritis (RA), one of the most common of the chronic inflammatory autoimmune diseases (CIADs), is recognized as an independent cardiovascular risk factor. Traditional risk factors such as smoking, arterial hypertension, dyslipidemia, insulin resistance, and obesity are frequently found in RA. Given the increased risk of mortality and morbidity associated with cardiovascular disease (CVD) in RA patients, screening for risk factors is important. Moreover, there is a need to identify potential predictors of subclinical atherosclerosis. Recent studies have shown that markers such as serum homocysteine, asymmetric dimethylarginine, or carotid intima-media thickness (cIMT) are correlated with cardiovascular risk. Although RA presents a cardiovascular risk comparable to that of diabetes, it is not managed as well in terms of acute cardiovascular events. The introduction of biological therapy has opened new perspectives in the understanding of this pathology, confirming the involvement and importance of the inflammatory markers, cytokines, and the immune system. In addition to effects in inducing remission and slowing disease progression, most biologics have demonstrated efficacy in reducing the risk of major cardiovascular events. Some studies have also been conducted in patients without RA, with similar results. However, early detection of atherosclerosis and the use of targeted therapies are the cornerstone for reducing cardiovascular risk in RA patients.
Collapse
Affiliation(s)
- Diana Popescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology and Physiotherapy, “Grigore. T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Rheumatology Clinic, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
- Correspondence: (E.R.); (M.C.B.)
| | - Minerva Codruta Badescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
- Correspondence: (E.R.); (M.C.B.)
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Petronela Nicoleta Seritean Isac
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Ioan-Teodor Dragoi
- Department of Rheumatology and Physiotherapy, “Grigore. T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Rheumatology Clinic, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Internal Medicine Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| |
Collapse
|
15
|
Simantiris S, Antonopoulos AS, Papastamos C, Benetos G, Koumallos N, Tsioufis K, Tousoulis D. Lipoprotein(a) and inflammation- pathophysiological links and clinical implications for cardiovascular disease. J Clin Lipidol 2023; 17:55-63. [PMID: 36333256 DOI: 10.1016/j.jacl.2022.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
The role of lipoprotein(a) (Lp[a]) as a significant and possibly causal cardiovascular disease (CVD) risk factor has been well established. Many studies, mostly experimental, have supported inflammation as a mediator of Lp(a)-induced increase in CVD risk. Lp(a), mainly through oxidized phospholipids bound to its apolipoprotein(a) part, leads to monocyte activation and endothelial dysfunction. The relationship between Lp(a) and inflammation is bidirectional as Lp(a) levels, besides being associated with inflammatory properties, are regulated by inflammatory stimuli or anti-inflammatory treatment. Reduction of Lp(a) concentration, especially by potent siRNA agents, contributes to partial reversion of the Lp(a) related inflammatory profile. This review aims to present the current pathophysiological and clinical evidence of the relationship between Lp(a) and inflammation.
Collapse
Affiliation(s)
- Spyridon Simantiris
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Alexios S Antonopoulos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Charalampos Papastamos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Georgios Benetos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Nikolaos Koumallos
- Department of Cardiothoracic Surgery, Hippokration Hospital, Athens, Greece (Dr Koumallos)
| | - Konstantinos Tsioufis
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Dimitris Tousoulis
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis).
| |
Collapse
|
16
|
Wang ZH, Qiao S, Wang L, Wang K, Zhang R, Jin Y, Wu HK, Liu X. Plasma lipid profiles and homocysteine levels in anti-N-methyl-D-aspartate receptor encephalitis. Front Neurol 2023; 14:1148450. [PMID: 37122291 PMCID: PMC10133572 DOI: 10.3389/fneur.2023.1148450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/15/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction We aimed to investigate whether lipid profiles and homocysteine levels in patients with anti-N-methyl-D-aspartate receptor encephalitis are related to clinical presentation and prognosis, which may contribute to further research on the pathogenesis and treatment of this disease. Methods This study included a total of 43 patients with anti-N-methyl-D-aspartate receptor encephalitis and 43 sex-age-matched healthy controls. Baseline demography, clinical data, patient outcomes, and ancillary examination results were recorded. Patients were followed up every 2-3 months during the first year. The modified Rankin Scale score was used to evaluate the therapeutic effect and clinical outcome. Results Among the 43 patients included in this study, 55.81% were male, the mean age of onset was 27 years old, and the median modified Rankin Scale score on admission was 3.0. Apolipoprotein A-1 was significantly lower in patients with anti-N-methyl-D-aspartate receptor encephalitis compared with healthy controls (p = 0.004). Compared with healthy controls, homocysteine (p = 0.002), apolipoprotein B (p = 0.004), Lpa (p = 0.045), and apolipoprotein B/apolipoprotein A-1 (p = 0.001) were significantly increased in patients with anti-N-methyl-D-aspartate receptor encephalitis. According to the modified Rankin Scale scores, 6 months after discharge, 72.09% of patients had a good prognosis and 27.91% had a poor prognosis. In the good prognosis group, age (p = 0.031), lipoprotein a (p = 0.023), apolipoprotein A-1 (p = 0.027) at baseline, and the modified Rankin Scale score on admission (p = 0.019) were significantly higher than those in the poor prognosis group. Conclusion This study suggests the possibility that serum lipid profile and homocysteine play an important role in the pathogenesis of anti-N-methyl-D-aspartate receptor encephalitis, providing support for lipid-lowering treatment of anti-N-methyl-D-aspartate receptor encephalitis patients.
Collapse
Affiliation(s)
- Zhi-hao Wang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Shan Qiao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Lei Wang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Kemo Wang
- Department of Neurology of Stroke Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ranran Zhang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Yang Jin
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Huai-kuan Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xuewu Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Epilepsy, Shandong University, Jinan, China
- *Correspondence: Xuewu Liu
| |
Collapse
|
17
|
Farhat H, Irfan H, Muthiah K, Pallipamu N, Taheri S, Thiagaraj SS, Shukla TS, Gutlapalli SD, Giva S, Penumetcha SS. Increased Risk of Cardiovascular Diseases in Rheumatoid Arthritis: A Systematic Review. Cureus 2022; 14:e32308. [PMID: 36632250 PMCID: PMC9827945 DOI: 10.7759/cureus.32308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune condition in which the body's joints are attacked by the immune system, leaving the patient disabled in severe cases, with irreversible joint damage and a lower quality of life. RA patients are more likely to develop cardiovascular (CV) disease, which increases their risk of morbidity and mortality. This study systematically reviews various CV diseases that might occur with RA including heart failure (HF), coronary artery disease, acute coronary syndrome, ischemic heart disease, stroke, cardiac death, venous thromboembolism, and valvular diseases. The relation between these complications and RA is specifically assessed. Systematic search was carried out on literature reporting the risk of each of the CV diseases in RA patients from databases in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The databases searched were MEDLINE (through PubMed) and Google Scholar using a combination of keywords and medical subject headings (MeSH). Our keywords were mainly "cardiovascular diseases" and "arthritis and rheumatoid". We found a total of 33 articles reporting each CV comorbidity. Interestingly, a wide spectrum of CV diseases is reported in patients with RA. Many tools were implemented in the diagnosis of each disease such as carotid intima-media thickness for atherosclerosis and echocardiography for HF. We confirmed that RA is associated with an increased risk of different CV events, and prophylactic measures should be implemented.
Collapse
Affiliation(s)
- Hadi Farhat
- Cardiology and Rheumatology, University of Balamand, Beirut, LBN
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Huma Irfan
- Research, Larkin Community Hospital, South Miami, USA
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Kanmani Muthiah
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Namratha Pallipamu
- Internal Medicine, Franciscan Health, Lafayette, USA
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sogand Taheri
- Medical Science, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Suvedha S Thiagaraj
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Twisha S Shukla
- Pediatrics, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sai Dheeraj Gutlapalli
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sheiniz Giva
- Neonatology, Children's Health Ireland at Temple Street, Dublin, IRL
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sai Sri Penumetcha
- General Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- General Medicine, Chalmeda Anand Rao Institute of Medical Sciences, Karimnagar, IND
| |
Collapse
|
18
|
Dzobo KE, Kraaijenhof JM, Stroes ES, Nurmohamed NS, Kroon J. Lipoprotein(a): An underestimated inflammatory mastermind. Atherosclerosis 2022; 349:101-109. [DOI: 10.1016/j.atherosclerosis.2022.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/09/2022] [Accepted: 04/01/2022] [Indexed: 12/11/2022]
|
19
|
The Influence of Treatment with PCSK9 Inhibitors and Variants in the CRP (rs1800947), TNFA (rs1800629), and IL6 (rs1800795) Genes on the Corresponding Inflammatory Markers in Patients with Very High Lipoprotein(a) Levels. J Cardiovasc Dev Dis 2022; 9:jcdd9050127. [PMID: 35621838 PMCID: PMC9146305 DOI: 10.3390/jcdd9050127] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammation contributes significantly to the development and progression of atherosclerosis. However, the factors that lead to an inflammatory imbalance towards a proinflammatory state are not yet fully understood. The CRP rs1800947, TNFA rs1800629, and IL6 rs1800795 polymorphisms may play a role in the pathogenesis of atherosclerosis and were therefore selected to investigate the influence of genetic variability on the corresponding plasma levels after treatment with a proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor. A group of 69 patients with stable coronary artery disease after myocardial infarction before the age of 50 years and very high lipoprotein(a) levels were enrolled in the study. All patients received a PCSK9 inhibitor (evolocumab or alirocumab). Genotyping was performed using TaqMan assays (CRP rs1800947, TNFA rs1800629, and IL6 rs1800795). Consistent with previous studies, no significant change in levels of inflammatory biomarkers was observed after 6 months of treatment with PCSK9 inhibitors. We also did not detect any significant association between single nucleotide polymorphisms CRP rs1800947, TNFA rs1800629, and IL6 rs1800795 and plasma levels of high-sensitivity C-reactive protein (hsCRP), tumor necrosis factor-α (TNF-α), or interleukin 6 (IL6), respectively, at enrollment. However, the difference in IL6 levels after treatment with PCSK9 inhibitors was statistically significant (p = 0.050) in patients with IL6-74CC genotype, indicating the possible role of the IL6 rs1800795 polymorphism in modulating inflammation.
Collapse
|
20
|
Cantini F, Goletti D, Benucci M, Foti R, Damiani A, Niccoli L. Tailored first-line biologic and targeted synthetic disease modifying anti-rheumatic drugs therapy in patients with rheumatoid arthritis: 2021 updated ITABIO statements. Expert Opin Drug Saf 2021; 21:613-623. [PMID: 34937466 DOI: 10.1080/14740338.2022.2020247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
INTRODUCTION In 2015, the Italian board for the TAilored BIOlogic therapy (ITABIO) proposed evidence-based decisional statements for first-line tailored biologic therapy in patients with rheumatoid arthritis (RA). Taking into account the new licensed drugs, the aim of the present review was to update the previous statements. AREAS COVERED A narrative review of the most recent evidence on the efficacy and safety of old and newly licensed drugs for the treatment of articular and extra-articular RA was performed. In addition, host-related variables potentially driving the therapy choice, such as the infection risk, the cardiovascular risk, the risk of deep vein thrombosis, thromboembolism, pregnancy, and obesity were analyzed. Consequently, several statements for personalized therapy were formulated, thus providing a decisional algorithm useful for proper personalized therapy of RA patients in clinical practice. EXPERT OPINION Several clinical variables related to specific drug and host characteristics may drive the choice toward anti-TNF and non-anti-TNF biologics, or anti-JAKs, thus allowing to personalize the therapy. Consequently, the right therapy for the right patient would ensure a successful therapeutic intervention.
Collapse
Affiliation(s)
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Via,Portuense 292, 00149 Rome, Italy
| | - Maurizio Benucci
- Rheumatology Unit, Hospital S. Giovanni di Dio, Azienda USL-Toscana Centro, Florence, Italy
| | - Rosario Foti
- Rheumatology Unit, Vittorio-Emanuele University Hospital of Catania, Catania, Italy
| | - Arianna Damiani
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Laura Niccoli
- Rheumatology Department, Hospital of Prato, Prato Italy
| |
Collapse
|
21
|
Hamar A, Hascsi Z, Pusztai A, Czókolyová M, Végh E, Pethő Z, Gulyás K, Soós B, Kerekes G, Szekanecz É, Hodosi K, Szántó S, Szűcs G, Seres T, Szekanecz Z, Szamosi S. Prospective, simultaneous assessment of joint and vascular inflammation by PET/CT in tofacitinib-treated patients with rheumatoid arthritis: associations with vascular and bone status. RMD Open 2021; 7:e001804. [PMID: 34740980 PMCID: PMC8573670 DOI: 10.1136/rmdopen-2021-001804] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/27/2021] [Indexed: 12/04/2022] Open
Affiliation(s)
- Attila Hamar
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | | | - Anita Pusztai
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Monika Czókolyová
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Edit Végh
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Zsófia Pethő
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Katalin Gulyás
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Boglárka Soós
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - György Kerekes
- Intensive Care Unit, Department of Medicine, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Éva Szekanecz
- Department of Oncology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Katalin Hodosi
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Sándor Szántó
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
- Department of Sports Medicine, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Gabriella Szűcs
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Tamás Seres
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Zoltán Szekanecz
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Szilvia Szamosi
- Division of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| |
Collapse
|
22
|
Chen YJ, Liu SC, Lai KL, Tang KT, Lin CH, Chen YM, Tseng CW, Chang YM, Gotcher DF, Chiou CC, Weng SJ, Chen HH. Factors associated with risk of major adverse cardiovascular events in patients with rheumatoid arthritis: a nationwide, population-based, case-control study. Ther Adv Musculoskelet Dis 2021; 13:1759720X211030809. [PMID: 34471426 PMCID: PMC8404647 DOI: 10.1177/1759720x211030809] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives To investigate factors associated with major adverse cardiovascular events (MACEs) in patients with rheumatoid arthritis (RA). Methods We conducted a nationwide, population-based, case-control study using Taiwan's National Health Insurance Research Database for 2003-2013. From 2004 to 2012, we identified 108,319 newly diagnosed RA patients without previous MACEs, of whom 7,580 patients (7.0%) developed MACEs during follow-up. From these incident RA patients, we included 5,994 MACE cases and 1:4 matched 23,976 non-MACE controls for analysis. The associations of MACEs with comorbidities and use of anti-rheumatic medications within 1 year before the index date were examined using conditional logistic regression analyses. Results Using multivariable conditional logistic regression analysis, the risk of MACE in RA patients was associated with use of golimumab [odd's ratio (OR), 0.09; 95% confidence interval (CI), 0.01-0.67], abatacept (OR, 0.13; 95% CI, 0.02-0.93), hydroxychloroquine (OR, 0.90; 95% CI, 0.82-0.99), methotrexate (OR, 0.72; 95% CI, 0.64-0.81), cyclosporin (OR, 1.43; 95% CI, 1.07-1.91), nonsteroidal anti-inflammation drugs (NSAIDs) (OR, 1.36; 95% CI, 1.27-1.46), antiplatelet agent (OR, 2.47; 95% CI, 2.31-2.63), hypertension (without anti-hypertensive agents: OR, 1.04; 95% CI, 0.96-1.12; with anti-hypertensive agents: OR, 1.47; 95% CI, 1.36-1.59), diabetes (OR, 1.27; 95% CI, 1.18-1.37), hyperlipidemia without lipid-lowering agents (OR, 1.09; 95% CI, 1.01-1.17), ischemic heart disease (OR, 1.20; 95% CI, 1.10-1.31), and chronic obstructive pulmonary disease (COPD) (OR, 1.12; 95% CI, 1.03-1.23) in the parsimonious model. The risk of MACE in RA patients also increased markedly in participants younger than 65 years with some comorbidities. Conclusions This population-based case-control study revealed that the use of golimumab, abatacept, hydroxychloroquine, and methotrexate were associated with a decreased risk of MACE development in newly diagnosed RA patients, while the use of cyclosporin, NSAIDs, and antiplatelet agents, and comorbidities, including hypertension, diabetes, hyperlipidemia without lipid-lowering agent therapy, ischemic heart disease, and COPD, were associated with an increased risk of MACE development in RA patients.
Collapse
Affiliation(s)
- Yen-Ju Chen
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung
| | - Shih-Chia Liu
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung
| | - Kuo-Lung Lai
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung
| | - Kuo-Tung Tang
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung
| | - Ching-Heng Lin
- Department of Medical Research, Taichung Veterans General Hospital, Taichung
| | - Yi-Ming Chen
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung
| | - Chih-Wei Tseng
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung
| | - Yu-Mei Chang
- Department of Statistics, Tunghai University, Taichung
| | - Donald F Gotcher
- Department of International Business, Tunghai University, Taichung
| | - Chuang-Chun Chiou
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung
| | - Shao-Jen Weng
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung
| | - Hsin-Hua Chen
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung
| |
Collapse
|
23
|
Beyond Lipoprotein(a) plasma measurements: Lipoprotein(a) and inflammation. Pharmacol Res 2021; 169:105689. [PMID: 34033878 PMCID: PMC9247870 DOI: 10.1016/j.phrs.2021.105689] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022]
Abstract
Genome wide association, epidemiological, and clinical studies have established high lipoprotein(a) [Lp(a)] as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD). Lp(a) is an apoB100 containing lipoprotein covalently bound to apolipoprotein(a) [apo(a)], a glycoprotein. Plasma Lp(a) levels are to a large extent determined by genetics. Its link to cardiovascular disease (CVD) may be driven by its pro-inflammatory effects, of which its association with oxidized phospholipids (oxPL) bound to Lp(a) is the most studied. Various inflammatory conditions, such as rheumatoid arthritis (RA), systemic lupus erythematosus, acquired immunodeficiency syndrome, and chronic renal failure are associated with high Lp(a) levels. In cases of RA, high Lp(a) levels are reversed by interleukin-6 receptor (IL-6R) blockade by tocilizumab, suggesting a potential role for IL-6 in regulating Lp(a) plasma levels. Elevated levels of IL-6 and IL-6R polymorphisms are associated with CVD. Therapies aimed at lowering apo(a) and thereby reducing plasma Lp(a) levels are in clinical trials. Their results will determine if reductions in apo(a) and Lp(a) decrease cardiovascular outcomes. As we enter this new arena of available treatments, there is a need to improve our understanding of mechanisms. This review will focus on the role of Lp(a) in inflammation and CVD.
Collapse
|
24
|
Eraikhuemen N, Lazaridis D, Dutton MT. Emerging Pharmacotherapy to Reduce Elevated Lipoprotein(a) Plasma Levels. Am J Cardiovasc Drugs 2021; 21:255-265. [PMID: 32929693 PMCID: PMC8697690 DOI: 10.1007/s40256-020-00437-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lipoprotein(a) is a unique form of low-density lipoprotein. It is associated with a high incidence of premature atherosclerotic disease such as coronary artery disease, myocardial infarction, and stroke. Plasma levels of this lipoprotein and its activities are highly variable. This is because of a wide variability in the size of the apolipoprotein A moiety, which is determined by the number of repeats of cysteine-rich domains known as "kringles." Although the exact mechanism of lipoprotein(a)-induced atherogenicity is unknown, the lipoprotein has been found in the arterial walls of atherosclerotic plaques. It has been implicated in the formation of foam cells and lipid deposition in these plaques. Pharmacologic management of elevated levels of lipoprotein(a) with statins, fibrates, or bile acid sequestrants is ineffective. The newer and emerging lipid-lowering agents, such as the second-generation antisense oligonucleotides, cholesteryl ester transfer protein inhibitors, and proprotein convertase subtilisin/kexin type 9 inhibitors offer the most effective pharmacologic therapy.
Collapse
Affiliation(s)
- Nathaniel Eraikhuemen
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Davie, FL, 33324, USA
| | - Dovena Lazaridis
- Memorial Regional Hospital-Department of Pharmacy, 3501 Johnson Street, Hollywood, FL, 32301, USA.
| | - Matthew T Dutton
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, USA
| |
Collapse
|
25
|
Pierini FS, Botta E, Soriano ER, Martin M, Boero L, Meroño T, Saez MS, Lozano Chiappe E, Cerda O, Citera G, Gandino I, Rosa J, Sorroche P, Kontush A, Brites F. Effect of Tocilizumab on LDL and HDL Characteristics in Patients with Rheumatoid Arthritis. An Observational Study. Rheumatol Ther 2021; 8:803-815. [PMID: 33811316 PMCID: PMC8217399 DOI: 10.1007/s40744-021-00304-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/19/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND In patients with rheumatoid arthritis (RA), qualitative alterations of low and high-density lipoproteins (LDL and HDL, respectively) might partially explain their increased cardiovascular risk. Tocilizumab has been associated with an increase in lipids, including triglyceride (TG) and cholesterol levels. The aim of this study is to evaluate the effect of tocilizumab on certain LDL and HDL characteristics (oxidized LDL levels, HDL-associated enzymes, chemical composition of both total HDL and HDL3c subpopulation, and their capacity to promote cellular cholesterol efflux) at baseline and 3 months after the start of treatment in patients with RA. METHODS Twenty-eight RA patients (ACR/EULAR 2010 criteria) with indication of treatment with tocilizumab were included in the present study. Clinical assessment [Health assessment questionnaire (HAQ)], disease activity score 28 (DAS28), high-sensitivity C reactive protein (hsCRP) concentration, lipid profile, and lipoprotein (a) [Lp(a)] levels were evaluated in all patients at baseline and after 3 months of treatment with tocilizumab. Lipoprotein characteristics were evaluated through the levels of oxidized LDL (OxLDL), the activity of paraoxonase (PON) 1, the composition of total HDL and small, dense HDL3c subpopulation, and their ability to promote cellular cholesterol efflux. RESULTS After 3 months of treatment with tocilizumab, HAQ (- 23%, p < 0.05), DAS28 (- 49%, p < 0.001), and hsCRP (- 94%, p < 0.01) levels decreased significantly. Total cholesterol (TC), LDL-C, non-HDL-C, and apo B levels showed a significant increase after treatment (TC: + 7.0%, p < 0.01; LDL-C: + 10%, p < 0.01; non-HDL-C: + 9.9%, p < 0.01; and apo B: + 9.6%, p < 0.05). Decreases in Lp(a) and OxLDL levels were also observed after treatment [Lp(a): - 50%, p < 0.01; and oxLDL: - 5.4%, p < 0.05]. The latter was in accordance with the increment detected in PON activity. No changes were observed in HDL capacity to promote cholesterol efflux (p > 0.05) in the whole group. CONCLUSIONS Treatment with tocilizumab reduced hsCRP levels and displayed positive effects on certain lipoprotein-related parameters, such as a potent decrease inLp(a) and a reduction in OxLDL levels. Moreover, HDL capacity to promote cellular cholesterol efflux was maintained after 3 months of treatment.
Collapse
Affiliation(s)
- Florencia S Pierini
- Rheumatology Unit, Internal Medical Services, and University Institute, Hospital Italiano de Buenos Aires, Peron 4190, (1181), Buenos Aires, Argentina
| | - Eliana Botta
- Laboratory of Lipids and Atherosclerosis, School of Pharmacy and Biochemistry, INFIBIOC, University of Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Enrique R Soriano
- Rheumatology Unit, Internal Medical Services, and University Institute, Hospital Italiano de Buenos Aires, Peron 4190, (1181), Buenos Aires, Argentina.
| | - Maximiliano Martin
- Laboratory of Lipids and Atherosclerosis, School of Pharmacy and Biochemistry, INFIBIOC, University of Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Laura Boero
- Laboratory of Lipids and Atherosclerosis, School of Pharmacy and Biochemistry, INFIBIOC, University of Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Tomás Meroño
- Laboratory of Lipids and Atherosclerosis, School of Pharmacy and Biochemistry, INFIBIOC, University of Buenos Aires, CONICET, Buenos Aires, Argentina
| | - María Soledad Saez
- Central Laboratory, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Ezequiel Lozano Chiappe
- Laboratory of Lipids and Atherosclerosis, School of Pharmacy and Biochemistry, INFIBIOC, University of Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Osvaldo Cerda
- Rheumatology Service, Instituto de Rehabilitación Psicofísica, Buenos Aires, Argentina
| | - Gustavo Citera
- Rheumatology Service, Instituto de Rehabilitación Psicofísica, Buenos Aires, Argentina
| | - Ignacio Gandino
- Rheumatology Unit, Internal Medical Services, and University Institute, Hospital Italiano de Buenos Aires, Peron 4190, (1181), Buenos Aires, Argentina
| | - Javier Rosa
- Rheumatology Unit, Internal Medical Services, and University Institute, Hospital Italiano de Buenos Aires, Peron 4190, (1181), Buenos Aires, Argentina
| | - Patricia Sorroche
- Central Laboratory, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Anatol Kontush
- Faculty of Medicine Pitié-Salpêtrière, National Institute for Health and Medical Research (INSERM) Research Unit 1166 - ICAN, Sorbonne University, Paris, France
| | - Fernando Brites
- Laboratory of Lipids and Atherosclerosis, School of Pharmacy and Biochemistry, INFIBIOC, University of Buenos Aires, CONICET, Buenos Aires, Argentina
| |
Collapse
|
26
|
Taylor PC, Atzeni F, Balsa A, Gossec L, Müller-Ladner U, Pope J. The Key Comorbidities in Patients with Rheumatoid Arthritis: A Narrative Review. J Clin Med 2021; 10:509. [PMID: 33535498 PMCID: PMC7867048 DOI: 10.3390/jcm10030509] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 02/06/2023] Open
Abstract
Comorbidities in patients with rheumatoid arthritis (RA) are often associated with poor health outcomes and increased mortality. Treatment decisions should take into account these comorbidities due to known or suspected associations with certain drug classes. In clinical practice, it is critical to balance potential treatment benefit against the possible risks for comorbidities as well as the articular manifestations of RA. This review summarises the current literature relating to prevalence and risk factors for the important comorbidities of cardiovascular disease, infections, lymphomas and nonmelanoma skin cancers in patients with RA. The impact on patient outcomes and the interplay between these comorbidities and the therapeutic options currently available, including tumour necrosis factor inhibitors and newer biological therapies, are also explored. As newer RA therapies are developed, and patients gain wider and earlier access to advanced therapies, in part due to the emergence of biosimilars, it is important to consider the prevention or treatment of comorbidities as part of the overall management of RA.
Collapse
Affiliation(s)
- Peter C. Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Alejandro Balsa
- Rheumatology Unit, Hospital Universitario La Paz, La Paz Institute for Health Research IdiPAZ, Universidad Autónoma de Madrid, Paseo de la Castellana, 261, 28046 Madrid, Spain;
| | - Laure Gossec
- Institut Pierre Louis d’Epidémiologie et de Santé Publique, Sorbonne Université, 75006 Paris, France;
- Rheumatology Department, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne Université, 75013 Paris, France
| | - Ulf Müller-Ladner
- Department of Rheumatology and Clinical Immunology, Justus Liebig University Gießen, Campus Kerckhoff, 61231 Bad Nauheim, Germany;
| | - Janet Pope
- St. Joseph’s Health Care, Schulich School of Medicine, University of Western Ontario, London, ON N6A 5C1, Canada;
| |
Collapse
|
27
|
Halacoglu J, Shea LA. Cardiovascular Risk Assessment and Therapeutic Implications in Rheumatoid Arthritis. J Cardiovasc Transl Res 2020; 13:878-890. [PMID: 32080804 DOI: 10.1007/s12265-020-09964-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022]
Abstract
Patients with rheumatoid arthritis (RA) suffer from a magnitude of excess cardiovascular risk. A paradoxical lipid pattern has been observed in rheumatoid arthritis patients where low levels of total cholesterol and low-density lipoprotein are associated with a higher risk of cardiovascular disease. This paper aims to break down the evidence explaining why patients with low to normal LDL, and total cholesterol have such excess cardiovascular risk. A component of the enhanced cardiovascular risk is systemic inflammation and the subsequent pro-atherogenic dyslipidemia patterns. Due to this "lipid paradox," current risk algorithms and guidelines designed for the general population may underestimate cardiovascular risk in patients with rheumatoid arthritis. The purpose of this paper is to critically evaluate some of the discrepancies and layers of cardiovascular risk in RA patients, the role RA medication may have in mitigating or increasing cardiovascular risk, and the possible role of statin therapy.
Collapse
Affiliation(s)
- Juli Halacoglu
- Rueckert-Hartman College for Health Professions, School of Pharmacy, Regis University, 3333 Regis Blvd H-28, Denver, CO, 80221, USA
| | - Leticia A Shea
- Rueckert-Hartman College for Health Professions, School of Pharmacy, Regis University, 3333 Regis Blvd H-28, Denver, CO, 80221, USA.
| |
Collapse
|
28
|
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
|
29
|
Holm S, Oma I, Hagve TA, Saatvedt K, Brosstad F, Mikkelsen K, Rydningen H, Risnes I, Almdahl SM, Ueland T, Aukrust P, Halvorsen B, Hollan I. Levels of Lipoprotein (a) in patients with coronary artery disease with and without inflammatory rheumatic disease: a cross-sectional study. BMJ Open 2019; 9:e030651. [PMID: 31079089 PMCID: PMC6530453 DOI: 10.1136/bmjopen-2019-030651] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVES Patients with various inflammatory rheumatic diseases (IRDs) have increased risk of atherothrombotic disease. Lipoprotein (a) (Lp(a)) is a risk factor for atherosclerosis but its role in IRD with accompanying coronary artery disease (CAD) is still unclear. We aimed to examine if serum Lp(a) levels differed between CAD patients with and without accompanying IRD. DESIGN A cross-sectional observational, patient-based cohort study. SETTING Referred centre for coronary artery bypass grafting in the South Eastern part of Norway. PARTICIPANTS 67 CAD patients with IRD (CAD/IRD) and 52 CAD patients without IRD (CAD/non-IRD). All patients were Caucasians, aged >18 years, without any clinically significant infection or malignancy. METHODS Lp(a) levels in serum were analysed by particle enhanced immunoturbidimetric assay, and Lp(a) levels were related to clinical and biochemical characteristics of the patient population. RESULTS We found no differences in serum levels of Lp(a) between CAD patients with and without IRD. In general, we found that Lp(a) correlated poorly with clinical and biochemical parameters including C reactive protein with the same pattern in the CAD/non-IRD and CAD/IRD groups. CONCLUSIONS Our data do not support a link between inflammation and Lp(a) levels in CAD and in general Lp(a) levels were not correlated with other risk factors for cardiovascular disease.
Collapse
Affiliation(s)
- Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Rheumatology Department, Hospital for Rheumatic Diseases, Lillehammer, Norway
| | - Ingvild Oma
- Department of Pathology, Sykehuset Innlandet HF Divisjon Lillehammer, Lillehammer, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tor-Arne Hagve
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Diagnostics and Technology, Akershus University Hospital, Lorenskog, Norway
| | - Kjell Saatvedt
- Department of Thoracic Surgery, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Frank Brosstad
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Knut Mikkelsen
- Rheumatology Department, Hospital for Rheumatic Diseases, Lillehammer, Norway
| | - Hans Rydningen
- Department of Cardiac Surgery, Feiring Heart Clinic, Feiring, Norway
| | - Ivar Risnes
- Department of Cardiac Surgery, Feiring Heart Clinic, Feiring, Norway
| | - Sven Martin Almdahl
- Department of Cardiothoracic and Vascular Surgery, University Hospital of North Norway, Tromso, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ivana Hollan
- Rheumatology Department, Hospital for Rheumatic Diseases, Lillehammer, Norway
- Department of Pathology, Sykehuset Innlandet HF Divisjon Lillehammer, Lillehammer, Norway
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Harvard, Boston, USA
| |
Collapse
|
30
|
Castañeda S, Martínez-Quintanilla D, Martín-Varillas JL, García-Castañeda N, Atienza-Mateo B, González-Gay MA. Tocilizumab for the treatment of adult-onset Still’s disease. Expert Opin Biol Ther 2019; 19:273-286. [DOI: 10.1080/14712598.2019.1590334] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Santos Castañeda
- Rheumatology Division, Hospital de La Princesa, IIS-Princesa, Catedra de EPID-Futuro, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Dolores Martínez-Quintanilla
- Rheumatology Division, Hospital de La Princesa, IIS-Princesa, Catedra de EPID-Futuro, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - José L. Martín-Varillas
- Division of Rheumatology and Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Noelia García-Castañeda
- Rheumatology Division, Hospital de La Princesa, IIS-Princesa, Catedra de EPID-Futuro, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Belén Atienza-Mateo
- Division of Rheumatology and Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Miguel A. González-Gay
- Division of Rheumatology and Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
- Department of Medicine, University of Cantabria, Santander, Spain
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
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
|
34
|
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
|
35
|
Oxidized low density lipoproteins: The bridge between atherosclerosis and autoimmunity. Possible implications in accelerated atherosclerosis and for immune intervention in autoimmune rheumatic disorders. Autoimmun Rev 2018; 17:366-375. [DOI: 10.1016/j.autrev.2017.11.028] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/13/2017] [Indexed: 02/07/2023]
|
36
|
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
|