Therapeutic strategies targeting inflammation and immunity in atherosclerosis: how to proceed?

Suzanne E Engelen, Alice J B Robinson, Yasemin-Xiomara Zurke, Claudia Monaco, Suzanne E Engelen, Alice J B Robinson, Yasemin-Xiomara Zurke, Claudia Monaco

Abstract

Atherosclerosis is a chronic inflammatory disease of the arterial wall, characterized by the formation of plaques containing lipid, connective tissue and immune cells in the intima of large and medium-sized arteries. Over the past three decades, a substantial reduction in cardiovascular mortality has been achieved largely through LDL-cholesterol-lowering regimes and therapies targeting other traditional risk factors for cardiovascular disease, such as hypertension, smoking, diabetes mellitus and obesity. However, the overall benefits of targeting these risk factors have stagnated, and a huge global burden of cardiovascular disease remains. The indispensable role of immunological components in the establishment and chronicity of atherosclerosis has come to the forefront as a clinical target, with proof-of-principle studies demonstrating the benefit and challenges of targeting inflammation and the immune system in cardiovascular disease. In this Review, we provide an overview of the role of the immune system in atherosclerosis by discussing findings from preclinical research and clinical trials. We also identify important challenges that need to be addressed to advance the field and for successful clinical translation, including patient selection, identification of responders and non-responders to immunotherapies, implementation of patient immunophenotyping and potential surrogate end points for vascular inflammation. Finally, we provide strategic guidance for the translation of novel targets of immunotherapy into improvements in patient outcomes.

Conflict of interest statement

The authors declare no competing interests.

© 2022. Springer Nature Limited.

Figures

Fig. 1. History of research into the…
Fig. 1. History of research into the role of inflammation in atherosclerosis.
The timeline shows the main milestones in the past four decades of research into the role of inflammation in atherosclerosis. In the 1980s, the introduction of immunohistochemical techniques to study atherosclerotic plaques provided evidence of HLA-DR expression in human atherosclerotic plaques, followed by identification of monocytes, macrophages and T cells in the plaque,,–. In the 1990s, studies showed the presence of pro-inflammatory cytokines, such as tumour necrosis factor (TNF), in atherosclerotic plaques–, and the association between high plasma C-reactive protein (CRP) levels and coronary artery disease (CAD),. During this decade, the first mouse models of hypercholesterolaemia with an inflammatory gene knockout were developed,, and titres of antibodies against oxidized LDL (oxLDL) in the serum were shown to predict cardiovascular disease outcomes. In the 2000s, studies demonstrated the association between increased levels of inflammatory markers and increased risk of cardiovascular events,. An increased risk of cardiovascular disease was shown in patients with inflammatory diseases–, and several studies demonstrated the association between elevated levels of CRP, IL-6 and TNF in the plasma and worse clinical outcomes in patients with cardiovascular disease,,,. This finding led to the introduction of inflammation as a therapeutic target in cardiovascular disease. In the late 2010s, studies showed that immune checkpoint inhibitor treatment increased the risk of cardiovascular disease in patients with cancer,. In the past decade, clinical trials investigated whether targeting inflammation in cardiovascular disease is beneficial,,. Numerous studies also demonstrated the involvement of the bone marrow in atherosclerosis,, and performed single-cell analysis of plaque immune cells,. Preclinical discoveries are shown in blue boxes and clinical discoveries in red boxes. MI, myocardial infarction; VSMC, vascular smooth muscle cell.
Fig. 2. Inflammation in atherosclerosis.
Fig. 2. Inflammation in atherosclerosis.
In medium and large arteries, haemodynamic forces create areas of low shear stress that are often predictors of atherosclerotic plaque location. As the atherosclerotic plaque begins to form, circulating apolipoprotein B (ApoB)-containing lipoproteins (ApoB-LP) and ApoB peptides enter the subendothelial space, where they can be modified and recognized by innate immune cells as danger signals. These danger signals activate Toll-like receptor (TLR) signalling and the inflammasome in innate immune cells, eliciting responses that drive inflammation, including production and secretion of cytokines, release of neutrophil extracellular traps (NETs), upregulation of co-stimulatory molecules and promotion of monocyte recruitment to the plaque. Macrophages derived from monocyte differentiation, local proliferation or from transdifferentiation of vascular smooth muscle cells (VSMCs) take up lipoproteins present in the plaque and become lipid-laden foam cells that lay the foundation for the formation of the plaque necrotic core. At the immune synapse, antigen-presenting cells (APCs), including macrophages, dendritic cells and B cells, present lipid antigens to invariant natural killer T (iNKT) cells and peptide antigens to T cells, the latter engaging adaptive T cell and B cell responses. Antigen presentation occurs in the plaque and in secondary lymph organs, such as the lymph node. Together, all these processes contribute to endothelial dysfunction, leading to further aggravation of inflammation through continued monocyte recruitment, increased uptake of lipoproteins adding to the plaque lipid burden, VSMC activation and proliferation, and fibroblast migration contributing to fibrous cap formation. ECM, extracellular matrix; IFN, interferon; MHC, major histocompatibility complex; ROS, reactive oxygen species; TCR, T cell receptor; TH1, T helper 1; TNF, tumour necrosis factor; Treg cell, regulatory T cell.
Fig. 3. Targeting the immune system in…
Fig. 3. Targeting the immune system in atherosclerosis.
ad | Immunotherapies for the treatment of atherosclerosis that showed benefit (green), no benefit (red) or potential benefit (yellow) in reducing inflammation or cardiovascular events in clinical trials or currently being tested in ongoing clinical trials (blue) are shown. Therapeutics targeting innate immunity include IL-1 inhibitors, IL-6 inhibitors, tumour necrosis factor (TNF) blockers and p38 inhibitors (panel a). Therapeutics targeting adaptive immunity include local proliferation inhibitors in drug-eluting stents and low-dose IL-2 targeting regulatory T (Treg) cells (panel b). Therapeutics targeting lipoproteins to reduce inflammation include antibodies against oxidized LDL (oxLDL), lipoprotein-associated phospholipase A2 (Lp-PLA2), secretory phospholipase A2 (sPLA2) and lectin-like oxidized LDL receptor 1 (LOX1) (panel c). Therapeutics with broad immunosuppressive effects include colchicine, low-dose methotrexate, glucocorticoids and hydroxychloroquine (panel d). See Tables 1,2 and 3 and Supplementary Table 1 for further details. eg | Overview of therapeutics in preclinical development targeting innate immunity (panel e), co-stimulation pathways (panel f) and B cell and T cell regulation (panel g). APC, antigen-presenting cell; ApoB, apolipoprotein B; BAFF, B cell activating factor; BCMA, B cell maturation antigen; BTLA, B and T lymphocyte attenuator; CCR, C-C chemokine receptor; CD30L, CD30 ligand; CD40L, CD40 ligand; CTLA4, cytotoxic T lymphocyte antigen 4; CVD, cardiovascular disease; GLUT1, glucose transporter 1; HDAC, histone deacetylase; HSPC, haematopoietic stem and progenitor cell; IRF5, interferon regulatory factor 5; OX40L, OX40 ligand; PPARγ, peroxisome proliferator-activated receptor-γ; siRNA, small interfering RNA; SPM, specialized pro-resolving mediators; TLR, Toll-like receptor; TRAF6, tumour necrosis factor receptor-associated factor 6.

References

    1. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circulation. 2002;105:1135–1143.
    1. Arnett DK, et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;140:e596–e646.
    1. Neumann FJ, et al. 2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes. Eur. Heart J. 2020;41:407–477.
    1. Timmis A, et al. European Society of Cardiology: cardiovascular disease statistics 2019. Eur. Heart J. 2020;41:12–85.
    1. Liuzzo G, et al. The prognostic value of C-reactive protein and serum amyloid a protein in severe unstable angina. N. Engl. J. Med. 1994;331:417–424.
    1. Biasucci LM, et al. Elevated levels of interleukin-6 in unstable angina. Circulation. 1996;94:874–877.
    1. Galkina E, et al. Lymphocyte recruitment into the aortic wall before and during development of atherosclerosis is partially L-selectin dependent. J. Exp. Med. 2006;203:1273–1282.
    1. Roy P, Orecchioni M, Ley K. How the immune system shapes atherosclerosis: roles of innate and adaptive immunity. Nat. Rev. Immunol. 2021 doi: 10.1038/s41577-021-00584-1.
    1. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124:783–801.
    1. O’Neill LAJ, Golenbock D, Bowie AG. The history of Toll-like receptors–redefining innate immunity. Nat. Rev. Immunol. 2013;13:453–460.
    1. Tabas I, Lichtman AH. Monocyte-macrophages and T cells in atherosclerosis. Immunity. 2017;47:621–634.
    1. Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011;145:341–355.
    1. Tsiantoulas D, et al. APRIL limits atherosclerosis by binding to heparan sulfate proteoglycans. Nature. 2021;597:92–96.
    1. Stemme S, et al. T lymphocytes from human atherosclerotic plaques recognize oxidized low density lipoprotein. Proc. Natl Acad. Sci. USA. 1995;92:3893–3897.
    1. Stewart CR, et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat. Immunol. 2010;11:155–161.
    1. Binder CJ, Papac-Milicevic N, Witztum JL. Innate sensing of oxidation-specific epitopes in health and disease. Nat. Rev. Immunol. 2016;16:485–497.
    1. Naghavi M, et al. From vulnerable plaque to vulnerable patient: a call for new definitions and risk assessment strategies: Part I. Circulation. 2003;108:1664–1672.
    1. Zernecke A, et al. Meta-analysis of leukocyte diversity in atherosclerotic mouse aortas. Circ. Res. 2020;127:402–426.
    1. Depuydt MAC, et al. Microanatomy of the human atherosclerotic plaque by single-cell transcriptomics. Circ. Res. 2020;127:1437–1455.
    1. Lin JD, et al. Single-cell analysis of fate-mapped macrophages reveals heterogeneity, including stem-like properties, during atherosclerosis progression and regression. JCI Insight. 2019;4:e124574.
    1. Winkels H, et al. Atlas of the immune cell repertoire in mouse atherosclerosis defined by single-cell RNA-sequencing and mass cytometry. Circ. Res. 2018;122:1675–1688.
    1. Kim K, et al. Transcriptome analysis reveals nonfoamy rather than foamy plaque macrophages are proinflammatory in atherosclerotic murine models. Circ. Res. 2018;123:1127–1142.
    1. McArdle S, et al. Migratory and dancing macrophage subsets in atherosclerotic lesions. Circ. Res. 2019;125:1038–1051.
    1. Cole JE, et al. Immune cell census in murine atherosclerosis: cytometry by time of flight illuminates vascular myeloid cell diversity. Cardiovasc. Res. 2018;114:1360–1371.
    1. Fernandez DM, et al. Single-cell immune landscape of human atherosclerotic plaques. Nat. Med. 2019;25:1576–1588.
    1. Gu W, et al. Adventitial cell atlas of wt (wild type) and ApoE (apolipoprotein E)-deficient mice defined by single-cell RNA sequencing. Arterioscler. Thromb. Vasc. Biol. 2019;39:1055–1071.
    1. Cochain C, et al. Single-cell RNA-seq reveals the transcriptional landscape and heterogeneity of aortic macrophages in murine atherosclerosis. Circ. Res. 2018;122:1661–1674.
    1. McAlpine CS, et al. Sleep modulates haematopoiesis and protects against atherosclerosis. Nature. 2019;566:383–387.
    1. Robbins CS, et al. Extramedullary hematopoiesis generates Ly-6C high monocytes that infiltrate atherosclerotic lesions. Circulation. 2012;125:364–374.
    1. Fuster JJ, et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science. 2017;355:842–847.
    1. Soehnlein O, Libby P. Targeting inflammation in atherosclerosis–from experimental insights to the clinic. Nat. Rev. Drug Discov. 2021;20:589–610.
    1. Ridker PM, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 2017;377:1119–1131.
    1. Tardif J-C, et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N. Engl. J. Med. 2019;381:2497–2505.
    1. Nidorf SM, et al. Colchicine in patients with chronic coronary disease. N. Engl. J. Med. 2020;383:1838–1847.
    1. Full LE, Monaco C. Targeting inflammation as a therapeutic strategy in accelerated atherosclerosis in rheumatoid arthritis. Cardiovasc. Ther. 2011;29:231–242.
    1. Drobni ZD, et al. Association between immune checkpoint inhibitors with cardiovascular events and atherosclerotic plaque. Circulation. 2020;142:2299–2311.
    1. Poels K, et al. Immune checkpoint inhibitor therapy aggravates T cell-driven plaque inflammation in atherosclerosis. JACC Cardiovasc. Oncol. 2020;2:599–610.
    1. Ridker PM. How common is residual inflammatory risk? Circ. Res. 2017;120:617–619.
    1. Allahverdian S, Chaabane C, Boukais K, Francis GA, Bochaton-Piallat M-L. Smooth muscle cell fate and plasticity in atherosclerosis. Cardiovasc. Res. 2018;114:540–550.
    1. Doran AC, Meller N, McNamara CA. Role of smooth muscle cells in the initiation and early progression of atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2008;28:812–819.
    1. Lievens D, von Hundelshausen P. Platelets in atherosclerosis. Thromb. Haemost. 2011;106:827–838.
    1. Combadière C, et al. Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6Chi and Ly6Clo monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation. 2008;117:1649–1657.
    1. Tacke F, et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J. Clin. Invest. 2007;117:185–194.
    1. Swirski FK, et al. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J. Clin. Invest. 2007;117:195–205.
    1. Shimizu Y, et al. Radiation exposure and circulatory disease risk: Hiroshima and Nagasaki atomic bomb survivor data, 1950-2003. BMJ. 2010;340:b5349.
    1. Swirski FK, et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science. 2009;325:612–616.
    1. Rahman K, et al. Inflammatory Ly6Chi monocytes and their conversion to M2 macrophages drive atherosclerosis regression. J. Clin. Invest. 2017;127:2904–2915.
    1. Woollard KJ, Geissmann F. Monocytes in atherosclerosis: subsets and functions. Nat. Rev. Cardiol. 2010;7:77–86.
    1. Schloss MJ, Swirski FK, Nahrendorf M. Modifiable cardiovascular risk, hematopoiesis, and innate immunity. Circ. Res. 2020;126:1242–1259.
    1. Williams JW, et al. Limited proliferation capacity of aortic intima resident macrophages requires monocyte recruitment for atherosclerotic plaque progression. Nat. Immunol. 2020;21:1194–1204.
    1. Ensan S, et al. Self-renewing resident arterial macrophages arise from embryonic CX3CR1+ precursors and circulating monocytes immediately after birth. Nat. Immunol. 2016;17:159–168.
    1. Robbins CS, et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat. Med. 2013;19:1166–1172.
    1. Lim HY, et al. Hyaluronan receptor LYVE-1-expressing macrophages maintain arterial tone through hyaluronan-mediated regulation of smooth muscle cell collagen. Immunity. 2018;49:326–341.e7.
    1. Park I, et al. C-type lectin receptor CLEC4A2 promotes tissue adaptation of macrophages and protects against atherosclerosis. Nat. Commun. 2022;13:215.
    1. Weinberger T, et al. Ontogeny of arterial macrophages defines their functions in homeostasis and inflammation. Nat. Commun. 2020;11:4549.
    1. Murray PJ. Macrophage polarization. Annu. Rev. Physiol. 2017;79:541–566.
    1. Owsiany KM, Alencar GF, Owens GK. Revealing the origins of foam cells in atherosclerotic lesions. Arterioscler. Thromb. Vasc. Biol. 2019;39:836–838.
    1. Spann NJ, et al. Regulated accumulation of desmosterol integrates macrophage lipid metabolism and inflammatory responses. Cell. 2012;151:138–152.
    1. Voisin M, et al. Inhibiting LXRα phosphorylation in hematopoietic cells reduces inflammation and attenuates atherosclerosis and obesity in mice. Commun. Biol. 2021;4:420.
    1. Jaitin DA, et al. Lipid-associated macrophages control metabolic homeostasis in a Trem2-dependent manner. Cell. 2019;178:686–698.e14.
    1. Deguchi JO, et al. Inflammation in atherosclerosis: visualizing matrix metalloproteinase action in macrophages in vivo. Circulation. 2006;114:55–62.
    1. Kojima Y, Weissman IL, Leeper NJ. The role of efferocytosis in atherosclerosis. Circulation. 2017;135:476–489.
    1. Merad M, Sathe P, Helft J, Miller J, Mortha A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 2013;31:563–604.
    1. Yilmaz A, et al. Emergence of dendritic cells in rupture-prone regions of vulnerable carotid plaques. Atherosclerosis. 2004;176:101–110.
    1. Trogan E, et al. Gene expression changes in foam cells and the role of chemokine receptor CCR7 during atherosclerosis regression in ApoE-deficient mice. Proc. Natl Acad. Sci. USA. 2006;103:3781–3786.
    1. Weber C, et al. CCL17-expressing dendritic cells drive atherosclerosis by restraining regulatory T cell homeostasis in mice. J. Clin. Invest. 2011;121:2898–2910.
    1. Choi JH, et al. Flt3 signaling-dependent dendritic cells protect against atherosclerosis. Immunity. 2011;35:819–831.
    1. Subramanian M, Thorp E, Hansson GK, Tabas I. Treg-mediated suppression of atherosclerosis requires MYD88 signaling in DCs. J. Clin. Invest. 2013;123:179–188.
    1. Niessner A, et al. Pathogen-sensing plasmacytoid dendritic cells stimulate cytotoxic T-cell function in the atherosclerotic plaque through interferon-α. Circulation. 2006;114:2482–2489.
    1. MacRitchie N, et al. Plasmacytoid dendritic cells play a key role in promoting atherosclerosis in apolipoprotein e-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2012;32:2569–2579.
    1. Silvestre-Roig C, Braster Q, Ortega-Gomez A, Soehnlein O. Neutrophils as regulators of cardiovascular inflammation. Nat. Rev. Cardiol. 2020;17:327–340.
    1. Zernecke A, et al. Protective role of CXC receptor 4/CXC ligand 12 unveils the importance of neutrophils in atherosclerosis. Circ. Res. 2008;102:209–217.
    1. Wang L, et al. ROS-producing immature neutrophils in giant cell arteritis are linked to vascular pathologies. JCI Insight. 2020;5:e139163.
    1. Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science. 2015;349:316–320.
    1. Silvestre-Roig C, et al. Externalized histone H4 orchestrates chronic inflammation by inducing lytic cell death. Nature. 2019;569:236–240.
    1. Fuchs TA, et al. Extracellular DNA traps promote thrombosis. Proc. Natl Acad. Sci. USA. 2010;107:15880–15885.
    1. Soehnlein O, et al. Atherosclerosis: neutrophil-derived cathelicidin protects from neointimal hyperplasia. Sci. Transl. Med. 2011;3:103ra98.
    1. Saigusa R, Winkels H, Ley K. T cell subsets and functions in atherosclerosis. Nat. Rev. Cardiol. 2020;17:387–401.
    1. Ketelhuth DFJ, Hansson GK. Adaptive response of T and B cells in atherosclerosis. Circ. Res. 2016;118:668–678.
    1. Winkels H, Wolf D. Heterogeneity of T cells in atherosclerosis defined by single-cell RNA-sequencing and cytometry by time of flight. Arterioscler. Thromb. Vasc. Biol. 2021;41:549–563.
    1. Wolf D, et al. athogenic autoimmunity in atherosclerosis evolves from initially protective apolipoprotein B100-reactive CD4+ T-regulatory cells. Circulation. 2020;142:1279–1293.
    1. Kyaw T, et al. Cytotoxic and proinflammatory CD8+ T lymphocytes promote development of vulnerable atherosclerotic plaques in ApoE-deficient mice. Circulation. 2013;127:1028–1039.
    1. van Duijn J, Kuiper J, Slütter B. The many faces of CD8+ T cells in atherosclerosis. Curr. Opin. Lipidol. 2018;29:411–416.
    1. Hwang Y, et al. Expansion of CD8+ T cells lacking the IL-6 receptor α chain in patients with coronary artery diseases (CAD) Atherosclerosis. 2016;249:44–51.
    1. Bergström I, Backteman K, Lundberg A, Ernerudh J, Jonasson L. Persistent accumulation of interferon-γ-producing CD8+CD56+ T cells in blood from patients with coronary artery disease. Atherosclerosis. 2012;224:515–520.
    1. Getz GS, Reardon CA. Natural killer T cells in atherosclerosis. Nat. Rev. Cardiol. 2017;14:304–314.
    1. Bobryshev YV, Lord RSA. Co-accumulation of dendritic cells and natural killer T cells within rupture-prone regions in human atherosclerotic plaques. J. Histochem. Cytochem. 2005;53:781–785.
    1. Sage AP, Tsiantoulas D, Binder CJ, Mallat Z. The role of B cells in atherosclerosis. Nat. Rev. Cardiol. 2019;16:180–196.
    1. Kyaw T, et al. B1a B lymphocytes are atheroprotective by secreting natural IgM that increases IgM deposits and reduces necrotic cores in atherosclerotic lesions. Circ. Res. 2011;109:830–840.
    1. Strom AC, et al. B regulatory cells are increased in hypercholesterolaemic mice and protect from lesion development via IL-10. Thromb. Haemost. 2015;114:835–847.
    1. Nus M, et al. Marginal zone B cells control the response of follicular helper T cells to a high-cholesterol diet. Nat. Med. 2017;23:601–610.
    1. Sage AP, et al. Regulatory B cell-specific interleukin-10 is dispensable for atherosclerosis development in mice. Arterioscler. Thromb. Vasc. Biol. 2015;35:1770–1773.
    1. Rosser EC, et al. Regulatory B cells are induced by gut microbiota-driven interleukin-1β and interleukin-6 production. Nat. Med. 2014;20:1334–1339.
    1. Riggs JE, Lussier AM, Lee SK, Appel MC, Woodland RT. Differential radiosensitivity among B cell subpopulations. J. Immunol. 1988;141:1799–1807.
    1. Nidorf SM, Eikelboom JW, Budgeon CA, Thompson PL. Low-dose colchicine for secondary prevention of cardiovascular disease. J. Am. Coll. Cardiol. 2013;61:404–410.
    1. Ridker PM, et al. Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: a secondary analysis from the CANTOS randomised controlled trial. Lancet. 2018;391:319–328.
    1. Ridker PM, et al. Modulation of the interleukin-6 signalling pathway and incidence rates of atherosclerotic events and all-cause mortality: analyses from the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS) Eur. Heart J. 2018;39:3499–3507.
    1. Ridker PM, et al. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390:1833–1842.
    1. Daskalov, I. & Valova-Ilieva, T. Management of acute pericarditis: treatment and follow-up. ESC (2017).
    1. Hui M, et al. The British Society for Rheumatology guideline for the management of gout. Rheumatology. 2017;56:1056–1059.
    1. Paschke S, et al. Technical advance: inhibition of neutrophil chemotaxis by colchicine is modulated through viscoelastic properties of subcellular compartments. J. Leukoc. Biol. 2013;94:1091–1096.
    1. Aaltonen KJ, et al. Systematic review and meta-analysis of the efficacy and safety of existing TNF blocking agents in treatment of rheumatoid arthritis. PLoS ONE. 2012;7:e30275.
    1. Yamamoto-Furusho JK. Inflammatory bowel disease therapy: blockade of cytokines and cytokine signaling pathways. Curr. Opin. Gastroenterol. 2018;34:187–193.
    1. Reich K, et al. Tildrakizumab versus placebo or etanercept for chronic plaque psoriasis (reSURFACE 1 and reSURFACE 2): results from two randomised controlled, phase 3 trials. Lancet. 2017;390:276–288.
    1. Grebe A, Hoss F, Latz E. NLRP3 inflammasome and the IL-1 pathway in atherosclerosis. Circ. Res. 2018;122:1722–1740.
    1. Vromman A, et al. Stage-dependent differential effects of interleukin-1 isoforms on experimental atherosclerosis. Eur. Heart J. 2019;40:2482–2491.
    1. Gomez D, et al. Interleukin-1β has atheroprotective effects in advanced atherosclerotic lesions of mice. Nat. Med. 2018;24:1418–1429.
    1. Burzynski LC, et al. The coagulation and immune systems are directly linked through the activation of interleukin-1α by thrombin. Immunity. 2019;50:1033–1042.e6.
    1. Galea J, et al. Interleukin-1β in coronary arteries of patients with ischemic heart disease. Arterioscler. Thromb. Vasc. Biol. 1996;16:1000–1006.
    1. Abbate A, et al. Interleukin-1 blockade inhibits the acute inflammatory response in patients with ST-segment-elevation myocardial infarction. J. Am. Heart Assoc. 2020;9:e014941.
    1. Morton AC, et al. The effect of interleukin-1 receptor antagonist therapy on markers of inflammation in non-ST elevation acute coronary syndromes: The MRC-ILA heart study. Eur. Heart J. 2015;36:377–384.
    1. El Sayed H, Kerensky R, Stecher M, Mohanty P, Davies M. A randomized phase II study of Xilonix, a targeted therapy against interleukin 1α, for the prevention of superficial femoral artery restenosis after percutaneous revascularization. J. Vasc. Surg. 2016;63:133–141.e1.
    1. Ridker PM. Anticytokine agents: targeting interleukin signaling pathways for the treatment of atherothrombosis. Circ. Res. 2019;124:437–450.
    1. Zhang K, et al. Interleukin 6 destabilizes atherosclerotic plaques by downregulating prolyl-4-hydroxylase α1 via a mitogen-activated protein kinase and c-Jun pathway. Arch. Biochem. Biophys. 2012;528:127–133.
    1. Ridker PM, Rifai N, Stampfer MJ, Hennekens CH. Plasma concentration of interleukin-6 and the risk of future myocardial infarction among apparently healthy men. Circulation. 2000;101:1767–1772.
    1. Sarwar N, et al. Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies. Lancet. 2012;379:1205–1213.
    1. Swerdlow DI, et al. The interleukin-6 receptor as a target for prevention of coronary heart disease: a mendelian randomisation analysis. Lancet. 2012;379:1214–1224.
    1. Broch K, et al. Randomized trial of interleukin-6 receptor inhibition in patients with acute ST-segment elevation myocardial infarction. J. Am. Coll. Cardiol. 2021;77:1845–1855.
    1. Kleveland O, et al. Effect of a single dose of the interleukin-6 receptor antagonist tocilizumab on inflammation and troponin T release in patients with non-ST-elevation myocardial infarction: a double-blind, randomized, placebo-controlled phase 2 trial. Eur. Heart J. 2016;37:2406–2413.
    1. Ridker PM, et al. IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet. 2021;397:2060–2069.
    1. US National Library of Medicine. (2021).
    1. Tousoulis DI, Oikonomou E, Economou EK, Crea F, Kaski JC. Inflammatory cytokines in atherosclerosis: current therapeutic approaches. Eur. Heart J. 2016;37:1723–1735.
    1. Abbas A, et al. Sinterleukin 23 levels are increased in carotid atherosclerosis possible role for the interleukin 23/interleukin 17 axis. Stroke. 2015;46:793–799.
    1. Ohta H, et al. Disruption of tumor necrosis factor-α gene diminishes the development of atherosclerosis in ApoE-deficient mice. Atherosclerosis. 2005;180:11–17.
    1. Barath P, et al. Detection and localization of tumor necrosis factor in human atheroma. Am. J. Cardiol. 1990;65:297–302.
    1. Brånén L, et al. Inhibition of tumor necrosis factor-α reduces atherosclerosis in apolipoprotein E knockout mice. Arterioscler. Thromb. Vasc. Biol. 2004;24:2137–2142.
    1. Ridker PM, et al. Elevation of tumor necrosis factor-α and increased risk of recurrent coronary events after myocardial infarction. Circulation. 2000;101:2149–2153.
    1. Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT. Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-α, in patients with moderate-to-severe heart failure: results of the Anti-TNF Therapy Against Congestive Heart Failure (ATTACH) trial. Circulation. 2003;107:3133–3140.
    1. Mann DL, et al. Targeted anticytokine therapy in patients with chronic heart failure: results of the randomized etanercept worldwide evaluation (RENEWAL) Circulation. 2004;109:1594–1602.
    1. Gao Q, et al. A critical function of Th17 proinflammatory cells in the development of atherosclerotic plaque in mice. J. Immunol. 2010;185:5820–5827.
    1. Langrish CL, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J. Exp. Med. 2005;201:233–240.
    1. Ma S, et al. The immunomodulatory effect of bone marrow stromal cells (BMSCs) on interleukin (IL)-23/IL-17-mediated ischemic stroke in mice. J. Neuroimmunol. 2013;257:28–35.
    1. Tzellos T, Kyrgidis A, Zouboulis CC. Re-evaluation of the risk for major adverse cardiovascular events in patients treated with anti-IL-12/23 biological agents for chronic plaque psoriasis: a meta-analysis of randomized controlled trials. J. Eur. Acad. Dermatol. Venereol. 2013;27:622–627.
    1. Ryan C, et al. Association between biologic therapies for chronic plaque psoriasis and cardiovascular events: a meta-analysis of randomized controlled trials. JAMA. 2011;306:864–871.
    1. Ridker PM, et al. Low-dose methotrexate for the prevention of atherosclerotic events. N. Engl. J. Med. 2019;380:752–762.
    1. O’Donoghue ML, et al. Effect of losmapimod on cardiovascular outcomes in patients hospitalized with acute myocardial infarction: a randomized clinical trial. JAMA. 2016;315:1591–1599.
    1. Martin ED, Felice De Nicola G, Marber MS. New therapeutic targets in cardiology: p38 alpha mitogen-activated protein kinase for ischemic heart disease. Circulation. 2012;126:357–368.
    1. Dean JLE, Brook M, Clark AR, Saklatvala J. p38 Mitogen-activated protein kinase regulates cyclooxygenase-2 mRNA stability and transcription in lipopolysaccharide-treated human monocytes. J. Biol. Chem. 1999;274:264–269.
    1. Elkhawad M, et al. Effects of p38 mitogen-activated protein kinase inhibition on vascular and systemic inflammation in patients with atherosclerosis. Imaging. 2012;5:911–922.
    1. Newby LK, et al. Losmapimod, a novel p38 mitogen-activated protein kinase inhibitor, in non-ST-segment elevation myocardial infarction: a randomised phase 2 trial. Lancet. 2014;384:1187–1195.
    1. Lehrer Graiwer J, et al. FDG-PET imaging for oxidized LDL in stable atherosclerotic disease: a phase II study of safety, tolerability, and anti-inflammatory activity. JACC Cardiovasc. Imaging. 2015;8:493–494.
    1. Ćorović A, Wall C, Mason JC, Rudd JHF, Tarkin JM. Novel positron emission tomography tracers for imaging vascular inflammation. Curr. Cardiol. Rep. 2020;22:119.
    1. Oikonomou EK, et al. Non-invasive detection of coronary inflammation using computed tomography and prediction of residual cardiovascular risk (the CRISP CT study): a post-hoc analysis of prospective outcome data. Lancet. 2018;392:929–939.
    1. Antonopoulos AS, et al. Detecting human coronary inflammation by imaging perivascular fat. Sci. Transl. Med. 2017;9:eaal2658.
    1. Krittanawong C, et al. Machine learning prediction in cardiovascular diseases: a meta-analysis. Sci. Rep. 2020;10:16057.
    1. Padmanabhan S, Tran TQB, Dominiczak AF. Artificial intelligence in hypertension: seeing through a glass darkly. Circ. Res. 2021;128:1100–1118.
    1. Greten FR, Grivennikov SI. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity. 2019;51:27–41.
    1. Koelwyn GJ, et al. Myocardial infarction accelerates breast cancer via innate immune reprogramming. Nat. Med. 2020;26:1452–1458.
    1. Egen JG, Ouyang W, Wu LC. Human anti-tumor immunity: insights from immunotherapy clinical trials. Immunity. 2020;52:36–54.
    1. Leistner DM, et al. Differential immunological signature at the culprit site distinguishes acute coronary syndrome with intact from acute coronary syndrome with ruptured fibrous cap: results from the prospective translational OPTICO-ACS study. Eur. Heart J. 2020;41:3549–3560.
    1. Hamers AAJ, et al. Human monocyte heterogeneity as revealed by high-dimensional mass cytometry. Arterioscler. Thromb. Vasc. Biol. 2019;39:25–36.
    1. Kott KA, et al. Single-cell immune profiling in coronary artery disease: the role of state-of-the-art immunophenotyping with mass cytometry in the diagnosis of atherosclerosis. J. Am. Heart Assoc. 2020;9:e017759.
    1. Jaiswal S, et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N. Engl. J. Med. 2017;377:111–121.
    1. Svensson E, et al. TET2-driven clonal hematopoiesis predicts enhanced response to canakinumab in the CANTOS trial: an exploratory analysis [abstract] Circulation. 2019;138(Suppl. 1):15111.
    1. Pålsson-McDermott EM, O’Neill LAJ. Targeting immunometabolism as an anti-inflammatory strategy. Cell Res. 2020;30:300–314.
    1. Edgar L, et al. Hyperglycemia induces trained immunity in macrophages and their precursors and promotes atherosclerosis. Circulation. 2021;144:961–982.
    1. Seijkens T, et al. Hypercholesterolemia-induced priming of hematopoietic stem and progenitor cells aggravates atherosclerosis. FASEB J. 2014;28:2202–2213.
    1. Christ A, et al. Western diet triggers NLRP3-dependent innate immune reprogramming. Cell. 2018;172:162–175.e14.
    1. Netea MG, et al. Trained immunity: a program of innate immune memory in health and disease. Science. 2016;352:427.
    1. Bowes AJ, Khan MI, Shi Y, Robertson L, Werstuck GH. Valproate attenuates accelerated atherosclerosis in hyperglycemic ApoE-deficient mice: evidence in support of a role for endoplasmic reticulum stress and glycogen synthase kinase-3 in lesion development and hepatic steatosis. Am. J. Pathol. 2009;174:330–342.
    1. Manea SA, et al. Pharmacological inhibition of histone deacetylase reduces NADPH oxidase expression, oxidative stress and the progression of atherosclerotic lesions in hypercholesterolemic apolipoprotein E-deficient mice; potential implications for human atherosclerosis. Redox Biol. 2020;28:101338.
    1. Choi JH, et al. Trichostatin A exacerbates atherosclerosis in low density lipoprotein receptor-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2005;25:2404–2409.
    1. Hoeksema MA, et al. Targeting macrophage histone deacetylase 3 stabilizes atherosclerotic lesions. EMBO Mol. Med. 2014;6:1124–1132.
    1. Cao Q, et al. Histone deacetylase 9 represses cholesterol efflux and alternatively activated macrophages in atherosclerosis development. Arterioscler. Thromb. Vasc. Biol. 2014;34:1871–1879.
    1. Asare Y, et al. Histone deacetylase 9 activates IKK to regulate atherosclerotic plaque vulnerability. Circ. Res. 2020;127:811–823.
    1. Malhotra R, et al. HDAC9 is implicated in atherosclerotic aortic calcification and affects vascular smooth muscle cell phenotype. Nat. Genet. 2019;51:1580–1587.
    1. Bellenguez C, et al. Genome-wide association study identifies a variant in HDAC9 associated with large vessel ischemic stroke. Nat. Genet. 2012;44:328–333.
    1. Oburoglu L, et al. Glucose and glutamine metabolism regulate human hematopoietic stem cell lineage specification. Cell Stem Cell. 2014;15:169–184.
    1. Chen Z, Dudek J, Maack C, Hofmann U. Pharmacological inhibition of GLUT1 as a new immunotherapeutic approach after myocardial infarction. Biochem. Pharmacol. 2021;190:114597.
    1. Sarrazy V, et al. Disruption of Glut1 in hematopoietic stem cells prevents myelopoiesis and enhanced glucose flux in atheromatous plaques of ApoE−/− mice. Circ. Res. 2016;118:1062–1077.
    1. Klepper J, et al. Glut1 deficiency syndrome (Glut1DS): state of the art in 2020 and recommendations of the international Glut1DS study group. Epilepsia Open. 2020;5:354–365.
    1. Jaiswal S, Libby P. Clonal haematopoiesis: connecting ageing and inflammation in cardiovascular disease. Nat. Rev. Cardiol. 2020;17:137–144.
    1. Wang W, et al. Macrophage inflammation, erythrophagocytosis, and accelerated atherosclerosis in JAK2V617F mice. Circ. Res. 2018;123:E35–E47.
    1. Tang Y, et al. Inhibition of JAK2 suppresses myelopoiesis and atherosclerosis in ApoE−/− mice. Cardiovasc. Drugs Ther. 2020;34:145–152.
    1. Abplanalp WT, et al. Association of clonal hematopoiesis of indeterminate potential with inflammatory gene expression in patients with severe degenerative aortic valve stenosis or chronic postischemic heart failure. JAMA Cardiol. 2020;5:1170–1175.
    1. Sano S, et al. Tet2-mediated clonal hematopoiesis accelerates heart failure through a mechanism involving the IL-1β/NLRP3 INFLAMMASOME. J. Am. Coll. Cardiol. 2018;71:875–886.
    1. Wang Y, et al. Tet2-mediated clonal hematopoiesis in nonconditioned mice accelerates age-associated cardiac dysfunction. JCI Insight. 2020;5:e135204.
    1. Fuster JJ, et al. TET2-loss-of-function-driven clonal hematopoiesis exacerbates experimental insulin resistance in aging and obesity. Cell Rep. 2020;33:108326.
    1. Fidler TP, et al. The AIM2 inflammasome exacerbates atherosclerosis in clonal haematopoiesis. Nature. 2021;592:296–301.
    1. Schwartz DM, et al. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat. Rev. Drug Discov. 2017;16:843–862.
    1. Yang X, et al. Inhibition of JAK2/STAT3/SOCS3 signaling attenuates atherosclerosis in rabbit. BMC Cardiovasc. Disord. 2020;20:133.
    1. Hilgendorf I, Swirski FK, Robbins CS. Monocyte fate in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2015;35:272–279.
    1. Soehnlein O, et al. Distinct functions of chemokine receptor axes in the atherogenic mobilization and recruitment of classical monocytes. EMBO Mol. Med. 2013;5:471–481.
    1. Boring L, Gosling J, Cleary M, Charo IF. Decreased lesion formation in CCR2−/− mice reveals a role for chemokines in the initiation of atherosclerosis. Nature. 1998;394:894–897.
    1. Gu L, et al. Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient mice. Mol. Cell. 1998;2:275–281.
    1. Majmudar MD, et al. Monocyte-directed RNAi targeting CCR2 improves infarct healing in atherosclerosis-prone mice. Circulation. 2013;127:2038–2046.
    1. Georgakis MK, et al. Monocyte-chemoattractant protein-1 levels in human atherosclerotic lesions associate with plaque vulnerability. Arterioscler. Thromb. Vasc. Biol. 2021;41:2038–2048.
    1. Gilbert J, et al. Effect of CC chemokine receptor 2 CCR2 blockade on serum C-reactive protein in individuals at atherosclerotic risk and with a single nucleotide polymorphism of the monocyte chemoattractant protein-1 promoter region. Am. J. Cardiol. 2011;107:906–911.
    1. Cipriani S, et al. Efficacy of the CCR5 antagonist maraviroc in reducing early, ritonavir-induced atherogenesis and advanced plaque progression in mice. Circulation. 2013;127:2114–2124.
    1. Veillard NR, et al. Antagonism of RANTES receptors reduces atherosclerotic plaque formation in mice. Circ. Res. 2004;94:253–261.
    1. Maggi P, et al. Effects of therapy with maraviroc on the carotid intima media thickness in HIV-1/HCV co-infected patients. In Vivo. 2017;31:125–132.
    1. Francisci D, et al. Maraviroc intensification modulates atherosclerotic progression in HIV-suppressed patients at high cardiovascular risk. A randomized, crossover pilot study. Open Forum Infect. Dis. 2019;6:ofz112.
    1. Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat. Rev. Immunol. 2011;11:762–774.
    1. Lawrence T, Natoli G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat. Rev. Immunol. 2011;11:750–761.
    1. Smith, U. Pioglitazone: mechanism of action. Int. J. Clin. Pract. Suppl. (121), 13–18 (2001).
    1. Rigamonti E, Chinetti-Gbaguidi G, Staels B. Regulation of macrophage functions by PPAR-α, PPAR-γ, and LXRs in mice and men. Arterioscler. Thromb. Vasc. Biol. 2008;28:1050–1059.
    1. Bouhlel MA, et al. PPARγ activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab. 2007;6:137–143.
    1. Nakashiro S, et al. Pioglitazone-incorporated nanoparticles prevent plaque destabilization and rupture by regulating monocyte/macrophage differentiation in ApoE−/− mice. Arterioscler. Thromb. Vasc. Biol. 2016;36:491–500.
    1. Chang K, et al. Pioglitazone suppresses inflammation in vivo in murine carotid atherosclerosis: novel detection by dual-target fluorescence molecular imaging. Arterioscler. Thromb. Vasc. Biol. 2010;30:1933–1939.
    1. Pfützner A, et al. Improvement of cardiovascular risk markers by pioglitazone is independent from glycemic control: results from the pioneer study. J. Am. Coll. Cardiol. 2005;45:1925–1931.
    1. Erdmann E, et al. The effect of pioglitazone on recurrent myocardial infarction in 2,445 patients with type 2 diabetes and previous myocardial infarction. results from the PROactive (PROactive 05) study. J. Am. Coll. Cardiol. 2007;49:1772–1780.
    1. Langenfeld MR, et al. Pioglitazone decreases carotid intima-media thickness independently of glycemic control in patients with type 2 diabetes mellitus: results from a controlled randomized study. Circulation. 2005;111:2525–2531.
    1. de Jong M, van der Worp HB, van der Graaf Y, Visseren FLJ, Westerink J. Pioglitazone and the secondary prevention of cardiovascular disease. A meta-analysis of randomized-controlled trials. Cardiovasc. Diabetol. 2017;16:134.
    1. Seneviratne AN, et al. Interferon regulatory factor 5 controls necrotic core formation in atherosclerotic lesions by impairing efferocytosis. Circulation. 2017;136:1140–1154.
    1. Leipner J, et al. Myeloid cell-specific Irf5 deficiency stabilizes atherosclerotic plaques in Apoe−/− mice. Mol. Metab. 2021;53:101250.
    1. Courties G, et al. In vivo silencing of the transcription factor IRF5 reprograms the macrophage phenotype and improves infarct healing. J. Am. Coll. Cardiol. 2014;63:1556–1566.
    1. Krausgruber T, et al. IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat. Immunol. 2011;12:231–238.
    1. Song S, et al. Inhibition of IRF5 hyperactivation protects from lupus onset and severity. J. Clin. Invest. 2020;130:6700–6717.
    1. Ban T, et al. Genetic and chemical inhibition of IRF5 suppresses pre-existing mouse lupus-like disease. Nat. Commun. 2021;12:4379.
    1. Sharma A, et al. Specific NLRP3 inhibition protects against diabetes-associated atherosclerosis. Diabetes. 2021;70:772–787.
    1. Van Der Heijden T, et al. NLRP3 inflammasome inhibition by MCC950 reduces atherosclerotic lesion development in apolipoprotein e-deficient mice–brief report. Arterioscler. Thromb. Vasc. Biol. 2017;37:1457–1461.
    1. Mangan MSJ, et al. Targeting the NLRP3 inflammasome in inflammatory diseases. Nat. Rev. Drug Discov. 2018;17:588–606.
    1. Klück V, et al. Dapansutrile, an oral selective NLRP3 inflammasome inhibitor, for treatment of gout flares: an open-label, dose-adaptive, proof-of-concept, phase 2a trial. Lancet Rheumatol. 2020;2:e270–e280.
    1. US National Library of Medicine. (2014).
    1. Wohlford GF, et al. Phase 1B, randomized, double-blinded, dose escalation, single-center, repeat dose safety and pharmacodynamics study of the oral NLRP3 indibitor dapansutrile in subjects with NYHA II-III systolic heart failure. J. Cardiovasc. Pharmacol. 2020;77:49–60.
    1. US National Library of Medicine. (2021).
    1. Monaco C, et al. Toll-like receptor-2 mediates inflammation and matrix degradation in human atherosclerosis. Circulation. 2009;120:2462–2469.
    1. Li Y, et al. VX-765 attenuates atherosclerosis in ApoE deficient mice by modulating VSMCs pyroptosis. Exp. Cell Res. 2020;389:111847.
    1. MacKenzie SH, Schipper JL, Clark AC. The potential for caspases in drug discovery. Curr. Opin. Drug Discov. Dev. 2010;13:568–576.
    1. Nilsson J, Hansson GK. Vaccination strategies and immune modulation of atherosclerosis. Circ. Res. 2020;126:1281–1296.
    1. Chyu KY, et al. CD8+ T cells mediate the athero-protective effect of immunization with an ApoB-100 peptide. PLoS ONE. 2012;7:e30780.
    1. Dunér P, et al. Antibodies against apoB100 peptide 210 inhibit atherosclerosis in apoE−/− mice. Sci. Rep. 2021;11:9022.
    1. Herbin O, et al. Regulatory T-cell response to apolipoprotein B100-derived peptides reduces the development and progression of atherosclerosis in mice. Arterioscler. Thromb. Vasc. Biol. 2012;32:605–612.
    1. Pattarabanjird T, Li C, McNamara C. B cells in atherosclerosis: mechanisms and potential clinical applications. JACC Basic Transl. Sci. 2021;6:546–563.
    1. Ait-Oufella H, et al. B cell depletion reduces the development of atherosclerosis in mice. J. Exp. Med. 2010;207:1579–1587.
    1. Kyaw T, et al. Conventional B2 B cell depletion ameliorates whereas its adoptive transfer aggravates atherosclerosis. J. Immunol. 2010;185:4410–4419.
    1. Zhao TX, et al. Rituximab in patients with acute ST-elevation myocardial infarction: an experimental medicine safety study. Cardiovasc. Res. 2021 doi: 10.1093/cvr/cvab113.
    1. Porsch F, Binder CJ. Impact of B-cell-targeted therapies on cardiovascular disease. Arterioscler. Thromb. Vasc. Biol. 2019;39:1705–1714.
    1. Kusters PJH, Lutgens E, Seijkens TTP. Exploring immune checkpoints as potential therapeutic targets in atherosclerosis. Cardiovasc. Res. 2018;114:368–377.
    1. Foks AC, et al. Interruption of the OX40–OX40 ligand pathway in LDL receptor-deficient mice causes regression of atherosclerosis. J. Immunol. 2013;191:4573–4580.
    1. Lutgens E, et al. Requirement for CD154 in the progression of atherosclerosis. Nat. Med. 1999;5:1313–1316.
    1. Foks AC, et al. Interference of the CD30–CD30L pathway reduces atherosclerosis development. Arterioscler. Thromb. Vasc. Biol. 2012;32:2862–2868.
    1. Winkels H, et al. CD27 co-stimulation increases the abundance of regulatory T cells and reduces atherosclerosis in hyperlipidaemic mice. Eur. Heart J. 2017;38:3590–3599.
    1. Kassiteridi C, et al. CD200 limits monopoiesis and monocyte recruitment in atherosclerosis. Circ. Res. 2021;129:280–295.
    1. Poels K, et al. Antibody-mediated inhibition of CTLA4 aggravates atherosclerotic plaque inflammation and progression in hyperlipidemic mice. Cells. 2020;9:1987.
    1. Schönbeck U, Sukhova GK, Shimizu K, Mach F, Libby P. Inhibition of CD40 signaling limits evolution of established atherosclerosis in mice. Proc. Natl Acad. Sci. USA. 2000;97:7458–7463.
    1. Douna H, et al. B- and T-lymphocyte attenuator stimulation protects against atherosclerosis by regulating follicular B cells. Cardiovasc. Res. 2020;116:295–305.
    1. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat. Rev. Immunol. 2020;20:651–668.
    1. Genovese MC, et al. Abatacept for rheumatoid arthritis refractory to tumor necrosis factor α inhibition. N. Engl. J. Med. 2005;353:1114–1123.
    1. Seijkens TTP, et al. Targeting CD40-induced TRAF6 signaling in macrophages reduces atherosclerosis. J. Am. Coll. Cardiol. 2018;71:527–542.
    1. Giugliano GR, Giugliano RP, Gibson CM, Kuntz RE. Meta-analysis of corticosteroid treatment in acute myocardial infarction. Am. J. Cardiol. 2003;91:1055–1059.
    1. Torii S, et al. Drug-eluting coronary stents: insights from preclinical and pathology studies. Nat. Rev. Cardiol. 2020;17:37–51.
    1. Razavi MK, Donohoe D, D’Agostino RB, Jaff MR, Adams G. Adventitial drug delivery of dexamethasone to improve primary patency in the treatment of superficial femoral and popliteal artery disease: 12-month results from the DANCE clinical trial. JACC Cardiovasc. Interv. 2018;11:921–931.
    1. Teunissen AJP, et al. Embracing nanomaterials’ interactions with the innate immune system. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2021;13:e1719.
    1. van der Valk FM, et al. Prednisolone-containing liposomes accumulate in human atherosclerotic macrophages upon intravenous administration. Nanomed. Nanotechnol. Biol. Med. 2015;11:1039–1046.
    1. Fitzgerald K, et al. A highly durable RNAi therapeutic inhibitor of PCSK9. N. Engl. J. Med. 2017;376:41–51.
    1. Flores AM, et al. Pro-efferocytic nanoparticles are specifically taken up by lesional macrophages and prevent atherosclerosis. Nat. Nanotechnol. 2020;15:154–161.
    1. Tao W, et al. SiRNA nanoparticles targeting CaMKIIγ in lesional macrophages improve atherosclerotic plaque stability in mice. Sci. Transl. Med. 2020;12:eaay1063.
    1. Fredman G, et al. Targeted nanoparticles containing the proresolving peptide Ac2-26 protect against advanced atherosclerosis in hypercholesterolemic mice. Sci. Transl. Med. 2015;7:275ra20.
    1. Kamaly N, et al. Targeted interleukin-10 nanotherapeutics developed with a microfluidic chip enhance resolution of inflammation in advanced atherosclerosis. ACS Nano. 2016;10:5280–5292.
    1. Tsimikas S. RNA-targeted therapeutics for lipid disorders. Curr. Opin. Lipidol. 2018;29:459–466.
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2021).
    1. US National Library of Medicine. (2021).
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2021).
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2021).
    1. US National Library of Medicine. (2020).
    1. US National Library of Medicine. (2017).
    1. Jonasson L, Holm J, Skalli O, Gabbiani G, Hansson GK. Expression of class II transplantation antigen on vascular smooth muscle cells in human atherosclerosis. J. Clin. Invest. 1985;76:125–131.
    1. Hansson GK, Jonasson L, Holm J, Claesson-Welsh L. Class II MHC antigen expression in the atherosclerotic plaque: smooth muscle cells express HLA-DR, HLA-DQ and the invariant gamma chain. Clin. Exp. Immunol. 1986;64:261–268.
    1. Vedeler CA, Nyland H, Matre R. In situ characterization of the foam cells in early human atherosclerotic lesions. Acta Pathol. Microbiol. Immunol. Scand. C. 1984;92:133–137.
    1. Aqel NM, Ball RY, Waldmann H, Mitchinson MJ. Identification of macrophages and smooth muscle cells in human atherosclerosis using monoclonal antibodies. J. Pathol. 1985;146:197–204.
    1. Hansson GK, Holm J, Jonasson L. Detection of activated T lymphocytes in the human atherosclerotic plaque. Am. J. Pathol. 1989;135:169–175.
    1. Jonasson L, Holm J, Skalli O, Bondjers G, Hansson GK. Regional accumulations of T cells, macrophages, and smooth muscle cells in the human atherosclerotic plaque. Arteriosclerosis. 1986;6:131–138.
    1. Emeson EE, Robertson AL. T lymphocytes in aortic and coronary intimas: their potential role in atherogenesis. Am. J. Pathol. 1988;130:369–376.
    1. Amento EP, Ehsani N, Palmer H, Libby P. Cytokines and growth factors positively and negatively regulate interstitial collagen gene expression in human vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 1991;11:1223–1230.
    1. Warner SJC, Libby P. Human vascular smooth muscle cells. Target for and source of tumor necrosis factor. J. Immunol. 1989;142:100–109.
    1. Warner SJC, Auger KR, Libby P. Human interleukin 1 induces interleukin I gene expression in human vascular smooth muscle cells. J. Exp. Med. 1987;165:1316–1331.
    1. Geng YJ, Wu Q, Muszynski M, Hansson GK, Libby P. Apoptosis of vascular smooth muscle cells induced by in vitro stimulation with interferon-γ, tumor necrosis factor-α, and interleukin-1β. Arterioscler. Thromb. Vasc. Biol. 1996;16:19–27.
    1. De Villiers WJS, et al. Macrophage phenotype in mice deficient in both macrophage-colony- stimulating factor (Op) and apolipoprotein E. Arterioscler. Thromb. Vasc. Biol. 1998;18:631–640.
    1. Berk BC, Weintraub WS, Alexander RW. Elevation of C-reactive protein in ‘active’ coronary artery disease. Am. J. Cardiol. 1990;65:168–172.
    1. Rajavashisth T, et al. Heterozygous osteopetrotic (op) mutation reduces atherosclerosis in LDL receptor-deficient mice. J. Clin. Invest. 1998;101:2702–2710.
    1. Smith JD, et al. Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc. Natl Acad. Sci. USA. 1995;92:8264–8268.
    1. Salonen JT, et al. Autoantibody against oxidised LDL and progression of carotid atherosclerosis. Lancet. 1992;339:883–887.
    1. The Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447:661–678.
    1. Samani NJ, et al. Genomewide association analysis of coronary artery disease. N. Engl. J. Med. 2007;357:443–453.
    1. Roman MJ, et al. Prevalence and correlates of accelerated atherosclerosis in systemic lupus erythematosus. N. Engl. J. Med. 2003;349:2399–2406.
    1. Aviña-Zubieta JA, et al. Risk of cardiovascular mortality in patients with rheumatoid arthritis: a meta-analysis of observational studies. Arthritis Care Res. 2008;59:1690–1697.
    1. Ha C, Magowan S, Accortt NA, Chen J, Stone CD. Risk of arterial thrombotic events in inflammatory bowel disease. Am. J. Gastroenterol. 2009;104:1445–1451.
    1. Ridker PM, Buring JE, Shih J, Matias M, Hennekens CH. Prospective study of C-reactive protein and the risk of future cardiovascular events among apparently healthy women. Circulation. 1998;98:731–733.
    1. Ross R. Inflammation or atherogenesis. N. Engl. J. Med. 1999;340:115–126.
    1. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N. Engl. J. Med. 2005;352:1685–1695.
    1. Tomita Y, et al. Acute coronary syndrome as a possible immune-related adverse event in a lung cancer patient achieving a complete response to anti-PD-1 immune checkpoint antibody. Ann. Oncol. 2017;28:2893–2895.
    1. Bar J, et al. Acute vascular events as a possibly related adverse event of immunotherapy: a single-institute retrospective study. Eur. J. Cancer. 2019;120:122–131.
    1. Hansson GK, Hermansson A. The immune system in atherosclerosis. Nat. Immunol. 2011;12:204–212.
    1. Maeda N. Development of apolipoprotein E-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2011;31:1957–1962.
    1. Ishibashi S, et al. Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus-mediated gene delivery. J. Clin. Invest. 1993;92:883–893.
    1. Shapiro MD, Tavori H, Fazio S. PCSK9 from basic science discoveries to clinical trials. Circ. Res. 2018;122:1420–1438.
    1. Maxwell KN, Breslow JL. Adenoviral-mediated expression of Pcsk9 in mice results in a low-density lipoprotein receptor knockout phenotype. Proc. Natl Acad. Sci. USA. 2004;101:7100–7105.
    1. Mestas J, Hughes CCW. Of mice and not men: differences between mouse and human immunology. J. Immunol. 2004;172:2731–2738.
    1. Shay T, et al. Conservation and divergence in the transcriptional programs of the human and mouse immune systems. Proc. Natl Acad. Sci. USA. 2013;110:2946–2951.
    1. Von Herrath MG, Nepom GT. Lost in translation: barriers to implementing clinical immunotherapeutics for autoimmunity. J. Exp. Med. 2005;202:1159–1162.
    1. Graham AL. Naturalizing mouse models for immunology. Nat. Immunol. 2021;22:111–117.
    1. Greve JM, et al. Allometric scaling of wall shear stress from mice to humans: quantification using cine phase-contrast MRI and computational fluid dynamics. Am. J. Physiol. Hear. Circ. Physiol. 2006;291:1700–1708.
    1. Golforoush P, Yellon DM, Davidson SM. Mouse models of atherosclerosis and their suitability for the study of myocardial infarction. Basic Res. Cardiol. 2020;115:73.
    1. Schwartz SM, Galis ZS, Rosenfeld ME, Falk E. Plaque rupture in humans and mice. Arterioscler. Thromb. Vasc. Biol. 2007;27:705–713.
    1. Pasterkamp G, et al. Human validation of genes associated with a murine atherosclerotic phenotype. Arterioscler. Thromb. Vasc. Biol. 2016;36:1240–1246.
    1. Breschi A, Gingeras TR, Guigó R. Comparative transcriptomics in human and mouse. Nat. Rev. Genet. 2017;18:425–440.
    1. Sellers RS. Translating mouse models: immune variation and efficacy testing. Toxicol. Pathol. 2017;45:134–145.
    1. Mair KH, et al. The porcine innate immune system: an update. Dev. Comp. Immunol. 2014;45:321–343.
    1. Pabst R. The pig as a model for immunology research. Cell Tissue Res. 2020;380:287–304.
    1. Low LA, Mummery C, Berridge BR, Austin CP, Tagle DA. Organs-on-chips: into the next decade. Nat. Rev. Drug Discov. 2021;20:345–361.
    1. Masopust D, Sivula CP, Jameson SC. Of mice, dirty mice, and men: using mice to understand human immunology. J. Immunol. 2017;199:383–388.
    1. Abolins S, et al. The comparative immunology of wild and laboratory mice, Mus musculus domesticus. Nat. Commun. 2017;8:14811.
    1. Proto JD, et al. Hypercholesterolemia induces T cell expansion in humanized immune mice. J. Clin. Invest. 2018;128:2370–2375.
    1. Cai B, et al. MerTK receptor cleavage promotes plaque necrosis and defective resolution in atherosclerosis. J. Clin. Invest. 2017;127:564–568.
    1. Doran AC, et al. CAMKIIγ suppresses an efferocytosis pathway in macrophages and promotes atherosclerotic plaque necrosis. J. Clin. Invest. 2017;127:4075–4089.
    1. Thorp E, et al. Shedding of the Mer tyrosine kinase receptor is mediated by ADAM17 protein through a pathway involving reactive oxygen species, protein kinase Cδ, and p38 mitogen-activated protein kinase (MAPK) J. Biol. Chem. 2011;286:33335–33344.
    1. Kojima Y, et al. Cyclin-dependent kinase inhibitor 2B regulates efferocytosis and atherosclerosis. J. Clin. Invest. 2014;124:1083–1097.
    1. Kojima Y, et al. CD47-blocking antibodies restore phagocytosis and prevent atherosclerosis. Nature. 2016;536:86–90.
    1. Overton CD, Yancey PG, Major AS, Linton MF, Fazio S. Deletion of macrophage LDL receptor-related protein increases atherogenesis in the mouse. Circ. Res. 2007;100:670–677.
    1. Advani R, et al. CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin’s lymphoma. N. Engl. J. Med. 2018;379:1711–1721.
    1. Ansell SM, et al. Phase I study of the CD47 blocker TTI-621 in patients with relapsed or refractory hematologic malignancies. Clin. Cancer Res. 2021;27:2190–2199.
    1. Brown EJ, Frazier WA. Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 2001;11:130–135.
    1. Buatois V, et al. Preclinical development of a bispecific antibody that safely and effectively targets CD19 and CD47 for the treatment of B-cell lymphoma and leukemia. Mol. Cancer Ther. 2018;17:1739–1751.
    1. Moura R, et al. Thrombospondin-1 deficiency accelerates atherosclerotic plaque maturation in ApoE−/− mice. Circ. Res. 2008;103:1181–1189.
    1. Westlake SL, et al. Tumour necrosis factor antagonists and the risk of cardiovascular disease in patients with rheumatoid arthritis: a systematic literature review. Rheumatology. 2011;50:518–531.
    1. Bäck M, Yurdagul A, Tabas I, Öörni K, Kovanen PT. Inflammation and its resolution in atherosclerosis: mediators and therapeutic opportunities. Nat. Rev. Cardiol. 2019;16:389–406.
    1. Fredman G, et al. An imbalance between specialized pro-resolving lipid mediators and pro-inflammatory leukotrienes promotes instability of atherosclerotic plaques. Nat. Commun. 2016;7:12859.
    1. Thul S, Labat C, Temmar M, Benetos A, Bäck M. Low salivary resolvin D1 to leukotriene B4 ratio predicts carotid intima media thickness: a novel biomarker of non-resolving vascular inflammation. Eur. J. Prev. Cardiol. 2017;24:903–906.
    1. Laguna-Fernandez A, et al. ERV1/ChemR23 signaling protects against atherosclerosis by modifying oxidized low-density lipoprotein uptake and phagocytosis in macrophages. Circulation. 2018;138:1693–1705.
    1. Hasturk H, et al. Resolvin E1 (RvE1) attenuates atherosclerotic plaque formation in diet and inflammation-induced atherogenesis. Arterioscler. Thromb. Vasc. Biol. 2015;35:1123–1133.
    1. Salic K, et al. Resolvin E1 attenuates atherosclerosis in absence of cholesterol-lowering effects and on top of atorvastatin. Atherosclerosis. 2016;250:158–165.
    1. Petri MH, et al. Aspirin-triggered lipoxin A4 inhibits atherosclerosis progression in apolipoprotein E−/− mice. Br. J. Pharmacol. 2017;174:4043–4054.
    1. US National Library of Medicine. (2019).
    1. Schrezenmeier E, Dörner T. Mechanisms of action of hydroxychloroquine and chloroquine: implications for rheumatology. Nat. Rev. Rheumatol. 2020;16:155–166.
    1. Sharma TS, et al. Hydroxychloroquine use is associated with decreased incident cardiovascular events in rheumatoid arthritis patients. J. Am. Heart Assoc. 2016;5:e002867.
    1. Jung H, et al. The protective effect of antimalarial drugs on thrombovascular events in systemic lupus erythematosus. Arthritis Rheum. 2010;62:863–868.
    1. Graßhoff H, et al. Low-dose IL-2 therapy in autoimmune and rheumatic diseases. Front. Immunol. 2021;12:902.
    1. Von Spee-Mayer C, et al. Low-dose interleukin-2 selectively corrects regulatory T cell defects in patients with systemic lupus erythematosus. Ann. Rheum. Dis. 2016;75:1407–1415.
    1. Zhao TX, Newland SA, Mallat Z. 2019 ATVB plenary lecture: Interleukin-2 therapy in cardiovascular disease: the potential to regulate innate and adaptive immunity. Arterioscler. Thromb. Vasc. Biol. 2020;40:853–864.
    1. Zhao TX, et al. Low dose interleukin-2 in patients with stable ischaemic heart disease and acute coronary syndrome (LILACS) Eur. Heart J. 2020;41:e022452.
    1. Cole JE, et al. Unexpected protective role for Toll-like receptor 3 in the arterial wall. Proc. Natl Acad. Sci. USA. 2011;108:2372–2377.
    1. Salagianni M, et al. Toll-like receptor 7 protects from atherosclerosis by constraining inflammatory macrophage activation. Circulation. 2012;126:952–962.
    1. Cole JE, Kassiteridi C, Monaco C. Toll-like receptors in atherosclerosis: a ‘Pandora’s box’ of advances and controversies. Trends Pharmacol. Sci. 2013;34:629–636.
    1. Dinarello CA. Interleukin-1β and the autoinflammatory diseases. N. Engl. J. Med. 2009;360:2467–2470.
    1. Edfeldt K, Swedenborg J, Hansson GK, Yan ZQ. Expression of toll-like receptors in human atherosclerotic lesions: a possible pathway for plaque activation. Circulation. 2002;105:1158–1161.
    1. Methe H, et al. Expansion of circulating Toll-like receptor 4-positive monocytes in patients with acute coronary syndrome. Circulation. 2005;111:2654–2661.
    1. Mullick AE, et al. Increased endothelial expression of Toll-like receptor 2 at sites of disturbed blood flow exacerbates early atherogenic events. J. Exp. Med. 2008;205:373–383.
    1. Michelsen KS, et al. Lack of Toll-like receptor 4 or myeloid differentiation factor 88 reduces atherosclerosis and alters plaque phenotype in mice deficient in apolipoprotein E. Proc. Natl Acad. Sci. USA. 2004;101:10679–10684.
    1. Liu X, et al. Toll-like receptor 2 plays a critical role in the progression of atherosclerosis that is independent of dietary lipids. Atherosclerosis. 2008;196:146–154.
    1. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821–832.
    1. Duewell P, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464:1357–1361.
    1. Hornung V, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat. Immunol. 2008;9:847–856.
    1. Lüsebrink E, et al. AIM2 stimulation impairs reendothelialization and promotes the development of atherosclerosis in mice. Front. Cardiovasc. Med. 2020;7:223.
    1. Paulin N, et al. Double-strand DNA sensing Aim2 inflammasome regulates atherosclerotic plaque vulnerability. Circulation. 2018;138:321–323.
    1. Bauernfeind FG, et al. Cutting edge: NF-κB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J. Immunol. 2009;183:787–791.
    1. Py BF, Kim MS, Vakifahmetoglu-Norberg H, Yuan J. Deubiquitination of NLRP3 by BRCC3 critically regulates inflammasome activity. Mol. Cell. 2013;49:331–338.
    1. Kayagaki N, et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 2015;526:666–671.

Source: PubMed

3
Subscribe