Abstract
Many measures can control lipid risk factors for atherosclerosis. Yet, even with excellent control of dyslipidemia, other sources of risk remain. Hence, we must look beyond lipids to address residual risk. Lifestyle measures should form the foundation of cardiovascular risk control. Many pharmacologic interventions targeting oxidation have proven disappointing. A large program tested inhibition of a lipoprotein-associated phospholipase A2 (LpPLA2), culminating in two large-scale clinical trials that did not meet their primary endpoints. A variety of antioxidants have not shown benefit in clinical trials. Numerous laboratory and clinical studies have inculpated inflammatory pathways in the pathogenesis of atherosclerotic events. The p38 mitogen-activated protein kinase (MAPK) inhibitor losmapimod and an inhibitor of a leukocyte adhesion molecule, P-selectin, did not alter adverse events in trials. Low-dose methotrexate, despite the promising observational studies, did not lower biomarkers of inflammation or alter cardiovascular outcomes in the cardiovascular inflammation reduction trial (CIRT). Four large-scale investigations underway will determine colchicine’s ability to reduce recurrent events in secondary prevention. The Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) showed that an antibody that neutralizes IL-1β can reduce recurrent cardiovascular events in secondary prevention. The success of CANTOS points to the pathway that leads from the NLRP3 inflammasome through IL-1β to IL-6 as an attractive target for further study and clinical development beyond lipid therapies to address the unacceptable burden of risk that remains despite our best current care in secondary prevention.
Low-density lipoprotein (LDL) lies at the root of human atherosclerosis. If humans maintained for life an LDL concentration similar to that of newborns, we would probably not face a global epidemic of atherosclerosis.1,2 Cholesterol concentrations considered “normal” by far exceed that required to meet biological needs. Although LDL levels appear to be drifting down, concentrations in contemporary cohorts remain undesirably high from the perspective of atherosclerosis prevention.3, 4 Fortunately, we now possess a panel of effective tools for control of LDL. The companion article by Kastelein and colleagues presents in detail the remarkable advances in pharmacologic control of LDL that today permit driving LDL concentrations safely below those of newborn humans.
Atherosclerosis Treatment Beyond LDL
Yet, for many millions exposure to a lifetime of unnecessarily high LDL has already sown the seeds of atherosclerosis. This situation prompts the necessity of identifying targets beyond LDL to make further inroads against atherosclerotic complications. In this regard, evidence has snowballed that triglyceride-rich lipoproteins (TGRL) participate causally in atherogenesis. Rigorous clinical testing of treatments for lowering TGRL has begun. High-dose omega-3 fatty acids of pharmaceutical grade can lower cardiovascular event rates in selected individuals.5 Part, but not all of the benefit of eicosapentaenoic acid may accrue from an anti-inflammatory effect. A large-scale clinical trial has commenced that tests the ability of a novel selective PPAR-α modulator to improve outcomes in diabetic subjects with hypertriglyceridemia (> 200 mg/dL) and low HDL (< 40 mg/dL).6 RNA-based agents that lower apolipoprotein CIII and Lp(a) are likewise in development.7 Thus, we have come a long way in our ability to control effectively lipid risk factors for atherosclerosis. Yet, even with excellent control of LDL and TGRL, other sources of risk remain. Hence, we must look beyond lipids to expand the palette of interventions that we can offer to our patients who have already encountered a lifetime of exposure to hypercholesterolemia and dyslipidemia.
Lifestyle Modification: The Foundation of Management of Atherosclerosis Risk in Primary Prevention
For primary prevention, lifestyle measures should form the foundation of cardiovascular risk control. Although we lack sufficient data proving that lifestyle interventions improve cardiovascular outcomes in primary prevention, lifestyle modification confers many demonstrable benefits.8 I (P.L.) advocate placing a Pascal’s Wager on lifestyle improvements, as they entail little if any risk.9 Smoking cessation, weight control, and physical activity should thus precede or accompany pharmacologic intervention in primary prevention. Indeed, initiating pharmacologic anti-atherosclerotic therapy early in life presents vexing societal, ethical, and financial issues. As we validate biomarkers that indicate risk due to particular genetic variants or pathways, we might justify pharmacologic intervention on apparently well people in the future. This approach embodies the promise of personalized or precision medicine. Yet, few if any such biomarkers or interventions beyond LDL control have undergone sufficient validation for deployment in clinical practice today.
Secondary Prevention Beyond Lipids
Secondary prevention, however, has entered a new era regarding targeting non-lipid risk factors to reduce recurrent events.10 This review will consider the current state of this nascent field. The failure of elegant laboratory studies and animal experiments to translate into clinical practice has plagued this field for many years. A number of rational interventions predicated on substantial preclinical data have not realized their promise in rigorous large-scale clinical trials. We examine below some of these cases.
The Rise and Fall of Anti-Oxidant Interventions
Targeting oxidant pathways has proven disappointing. Almost all schemes of the pathogenesis of atherosclerosis invoke oxidatively modified LDL as a key instigating factor.11, 12 Various phospholipases can liberate lipid moieties from oxidized lipoproteins that can activate deleterious functions of vascular cells and leukocytes found in atheromata. Yet, several attempts to inhibit phospholipases to prevent the generation of these putatively pernicious mediators have not borne fruit. The largest program tested inhibition of a lipoprotein-associated phospholipase A2 (LpPLA2) in patients with acute coronary syndromes or in the stable phase of atherosclerosis.13 Despite promising signals from intravascular imaging studies in humans, reinforced by encouraging results in atherosclerotic pigs with the Lp-PLA2 inhibitor darapladib14, the two large-scale clinical trials SOLID and STABILITY did not meet their primary endpoints.15, 16 These results, and those from smaller studies targeting other phospholipases, have led to abandonment of this class of enzymes as therapeutic targets in atherosclerosis.
With respect to direct targeting of oxidative stress and lipoprotein oxidation, results have also proven disappointing. A variety of antioxidants have not shown benefit in clinical trials. Vitamins C and E, and beta carotene have failed to reduce events in appropriately conducted and powered clinical trials.17–19 The powerful antioxidant succinobucol partitions into lipoprotein particles and effectively prevents LDL oxidation in vitro. Yet, the large ARISE trial did not meet its primary endpoint.20 As in the case of phospholipases, enthusiasm has waned for the application of antioxidant vitamins and direct inhibitors of oxidation as therapeutic targets in atherosclerosis.
Anti-inflammatory Interventions: A New Day Dawns
Numerous laboratory and clinical studies have inculpated inflammatory pathways in the pathogenesis of atherosclerosis and its clinical complications.21–24 This body of evidence has inspired several trials, some completed and some underway, targeting various inflammatory pathways in secondary prevention of atherosclerotic events. The mitogen-activated protein kinases (MAPKs) mediate certain actions of stressors such as oxidant stress, pro-inflammatory cytokines, and pathogen-associated molecular patterns (PAMPs) such as bacterial lipopolysaccharide.25 Focused interest in the p38 MAPK emerged from a coherent series of preclinical and clinical studies using the inhibitor losmapimod. In patients with acute coronary syndromes, treatment with losmapimod decreased the biomarker of inflammation C-reactive protein measured with a highly sensitive assay (hsCRP).26 A study monitoring the effect of losmapimod on fluorodeoxyglucose uptake in humans failed to meet its primary endpoint.27 The preliminary phase of a large-scale clinical trial with losmapimod (LATITUDE) did not demonstrate the clinical benefit, discouraging further pursuit of this target.28, 29
A hallmark of inflammation, recruitment of leukocytes, is a process that depends on endothelial-leukocyte expression. In addition to endothelial cells, platelets contain one such adhesion molecule, P-selectin. This double source points to a pivotal role of this particular adhesion molecule in acute inflammation complicated by thrombosis. Studies of acute coronary syndrome patients treated with inclacumab, an anti-P-selectin antibody, reduced troponin release but did not alter adverse events in the SELECT-ACS trial.30, 31
Another agent that targets inflammation, methotrexate, has transformed the practice of rheumatology. Low weekly doses of this agent have modified the course of rheumatoid arthritis and furnish a mainstay of management of patients with rheumatoid arthritis and allied inflammatory conditions. Methotrexate appears to exert an anti-inflammatory effect by inhibiting the synthesis of purines and pyrimidines, the building blocks of DNA.32 Methotrexate causes the release of adenine nucleotides and adenosine from cells. The adenosine can engage G protein-coupled adenosine receptors, notably the A2A receptor implicated in downstream anti-inflammatory actions.33 Methotrexate may also inhibit the JAK/STAT protein kinase pathway, a mechanism parallel to that of losmapimod.34 The Cardiovascular Inflammation Reduction Trial (CIRT) evaluated low-dose methotrexate in patients at risk for cardiovascular events.35 Despite the promising observational studies, low-dose methotrexate did not alter cardiovascular outcomes in the individuals studied. Indeed, the intervention did not lower biomarkers of inflammation, and the baseline inflammation in the enrolled population was in a lower risk zone (hsCRP 1.6 mg/l).36 The null results of CIRT suggests that future trials of anti-inflammatory strategies should go to pains to study individuals with sufficient inflammatory burden at baseline to benefit from the intervention.
Another agent widely used to treat inflammatory conditions, colchicine, has also shown promise in cardiovascular inflammation. Colchicine has become a standard treatment for pericarditis, avoiding the corticosteroid rebound phenomenon that plagued the use of glucocorticoids in this cardiovascular inflammatory condition.37 The publication of the LoDoCo study pioneered the use of colchicine in secondary prevention of coronary artery disease events. This open label, placebo-controlled trial randomized 532 individuals with stable coronary artery disease to colchicine 0.5 mg/day vs. control.38 This study showed a highly significant reduction in recurrent cardiovascular events over a 3-year follow up. A recent study used computed tomographic coronary angiography to study individuals with acute coronary syndromes who underwent treatment with colchicine 0.5 mg/day. At a one-year follow-up they found a decrease in low-attenuation plaque volume and hsCRP.39 These promising preliminary studies have spawned four large-scale investigations of colchicine’s ability to reduce recurrent events in secondary prevention. Colchicine causes gastrointestinal distress sufficient to warrant discontinuation of the medication in over 10% of individuals. The precise molecular mechanism of colchicine’s anti-inflammatory effect remains incompletely understood.40, 41 Early work attributed colchicine’s anti-inflammatory actions to inhibition of microtubular function in inflammatory leukocytes. More recent studies have indicated that relatively high concentrations of colchicine (5 μM) can suppress activation of the NOD-like receptor family, pyrin domain-containing protein 3 (NLRP3) inflammasome, by inhibiting the assembly of this macromolecular multimer.
The NLRP3 inflammasome regulates the activity of a one of its constituent proteins, the enzyme caspase-1, originally known as IL (interleukin)-1-beta converting enzyme.42–45 The inflammasome undergoes activation by a number of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). A number of atherosclerosis-related stimuli also can co-activate the NLRP3 inflammasome. Such stimuli include cholesterol crystals, hypoxia, disturbed flow, and microbial products.46 The augmented action of caspase-1 following NLRP3 inflammasome activation processes the inactive precursors of the pro-inflammatory cytokines IL-1β and IL-18 to the mature active forms of these pro-inflammatory mediators.
CANTOS Established Inflammation as a Therapeutic Target in Atherosclerosis
The Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) assessed the ability of an antibody that neutralizes IL-1β in reducing recurrent cardiovascular events in individuals who had sustained an acute myocardial infarction at least 30 days before enrollment.47 This study included individuals who, despite treatment with guideline-directed medical therapy including highly effective doses of statins, had an hsCRP > 2.0 mg/L, the approximate population median. This study showed a 15% reduction in the primary endpoint of major adverse cardiovascular events.48 Pre-specified analyses of individuals who achieved a greater than median reduction in hsCRP after administration of the antibody, canakinumab, showed a 26% reduction in events and an over 30% reduction in cardiovascular and all-cause mortality.49 Responders to canakinumab gauged by achieving an IL-6 concentration below median showed a similar magnitude of benefit.50 CANTOS demonstrated the effectiveness of targeting inflammation in atherosclerosis. This study also validated the inflammasome pathway as a promising avenue for further anti-inflammatory interventions.
As expected from prior experience, and in keeping with interference with a host defense mechanism, canakinumab therapy associated with a small but statistically significant increase in fatal infections in CANTOS. The knowledge gained regarding the risk factors for and the types of infectious complications in the nearly 7,000 patients exposed to canakinumab in CANTOS can inform management to reduce this risk. Yet, future studies should aim to develop anti-inflammatory or pro-resolving therapies that entail less perturbation of host defenses.22, 51
Beyond CANTOS
Work published in the 1980s suggested a hierarchical scheme for a pro-inflammatory cytokine cascade whereby IL-1 would induce IL-6, the principal mediator of the acute phase response in hepatocytes (Figure).52–54 The marker of the acute-phase response, hsCRP, thus reports on this pathway. The recognition that the inflammasome regulates caspase-1 activity suggested a mechanism for triggering the IL-1 to IL-6 to CRP pathway. Targeting different elements of this pathway will provide further opportunities to build on the success of CANTOS to develop anti-inflammatory interventions for atherosclerosis. There are several programs targeting small-molecule inhibitors of the NLRP3 inflammasome.45, 55
The inflammasome–IL-1β–IL-6 pro-inflammatory pathway.
The NLRP3 inflammasome undergoes activation by a number of atherosclerosis-related stimuli including co-activation by cholesterol crystals, hypoxia, and disturbed flow. The activated inflammasome unleashes the activity of caspase-1, the converting enzyme that processes pro-IL-1β and pro-IL-18 to their mature pro-inflammatory forms. IL-1β can in turn strongly induce production of IL-6, the major mediator of the acute phase response. IL-6 signaling through the canonical receptor on hepatocytes unleashed the acute phase response. Among acute phase reactants, fibrinogen and plasminogen activator inhibitor play causal roles in atherothrombosis. Therapeutic interventions exist or are under development to inhibit this pathway at all levels as shown on the left of the figure. IL-1 alpha, the sibling cytokine to IL-1 beta shares many of its actions, but has distinct biochemical and cell biological properties, and doesn not depend on the inflammasome for activation. CRP denotes C-reactive protein.
Downstream of the inflammasome, IL-6 signaling furnished another attractive anti-inflammatory target. Strong and concordant human genetic studies support the causality of IL-6 signaling in atherothrombosis in humans.56, 57 Targeting the IL-6 pathway has complexities, however. IL-6 can signal through its canonical receptor, instigating inflammatory pathways. This receptor signals through the gp130/Jak/Stat pathway. Accordingly, in experimental atherosclerosis in mice, IL-6 promotes lesion formation. IL-6 can also signal through an alternative “trans” pathway, whereby it binds to a soluble form of the canonical receptor and activates gp130.58–60 IL-6 trans signaling may retard experimental atherosclerosis. Experimental ablation of IL-6 can augment experimental atherosclerosis. The clinically used anti-IL-6 antibody, tocilizumab, consistently increases plasma triglycerides in patients.61 Hence, this therapy may produce an atherogenic dyslipidemia that could promote atherosclerosis and balance any beneficial effect of neutralization of IL-6. In sum, the success of CANTOS points strongly to the potential benefits of targeting the NLRP3 inflammasome pathway, opening a fertile field for future investigation.
In addition to IL-1β, its sibling isoform, IL-1α, exerts many of the same biological activities.62 Yet the cell biology and biochemistry of IL-1α differ considerably from that of IL-1β. IL-1α does not require activation by the inflammasome to exert biological activity.62 IL-1α signals through contact or acts at short distances, as it associates primarily with the surface of cells. Dying cells can liberate IL-1α locally. Experimental evidence supports a causal role for IL-1α in experimental atherosclerosis.63, 64 An antibody that neutralizes human IL-1α is available for clinical use.65, 66 Thus, IL-1α, in addition to IL-1β, merits consideration as an anti-inflammatory therapy in human atherosclerosis.
Vaccinations and Other Immunotherapies for Mitigation of Atherosclerosis
The recognition of the involvement of adaptive immunity in experimental atherosclerosis has prompted several efforts to harness these insights for therapy in humans. Experimental observations showed that IgM antibodies (natural antibodies) that recognize epitopes associated with oxidized LDL or certain infectious agents such as Pneumococci can limit atherosclerosis.67 This observation has prompted the development of several strategies for stimulating endogenous antibodies that might mitigate atherogenesis in humans. Among various approaches, using immunogenic epitopes of the major apolipoprotein of LDL, apolipoprotein B, has received attention from several groups.68–70 Promising animal experiments have laid the groundwork for potential clinical investigations. Immunization experiments in mice generally use congenic strains that have a limited haplotype of the major histocompatibility complex that governs the afferent limb of adaptive immunity. Thus, extrapolation of the experimental results to humans with a much more diverse human leukocyte antigen system than inbred mice could prove challenging. Nonetheless, the hope for developing an anti-atherosclerosis vaccine strategies merits continued evaluation. Other immune therapies under clinical consideration include the use of very low-dose IL-2 to stimulate the activity of regulatory T cells (Treg), (Low-Dose Interleukin-2 in Patients with Stable Ischaemic Heart Disease and Acute Coronary Syndromes, LILACS, NCT03113773).71 These cells secrete transforming growth factor beta (TGF-β), an anti-inflammatory and pro-fibrotic cytokine. An early phase clinical study currently underway is testing the safety of administration of low-dose IL-2 to individuals with acute coronary syndromes and seeking evidence for anti-inflammatory action. This pioneering study may be the first of several to harness protective adaptive immune responses to moderate atherosclerosis.
Clonal Hematopoiesis: A Newly Recognized but Potent Cardiovascular Risk Factor
Recent observations have identified a heretofore unsuspected cardiovascular risk factor that will likely identify yet further novel approaches for targeting anti-atherosclerotic treatments and point to new pathways for therapeutic intervention. With human aging, bone marrow stem cells can acquire somatic mutations that confer a proliferative advantage. These mutated stem cells can give rise to clones of leukocytes that circulate in peripheral blood. The acquisition of further mutations by such a clone can lead to myelodysplastic syndrome and, ultimately, acute leukemia.72 The prevalence of such clones exceeds 10% in septuagenarians and continues to increase with age.73 As would be expected, individuals with these clones of mutant leukocytes have a higher incidence of acute leukemia and mortality due to hematologic malignancy. Yet, the increased mortality due to leukemia does not account for a striking increase in mortality in carriers of these clones. Cardiovascular disease makes up this gap in mortality.73, 74 CHIP associates with worsened outcomes for three major cardiovascular threats: myocardial infarction, stroke, and ischemic heart failure.74, 75 Individuals with CHIP do not have leukocytosis. Although this condition may be regarded as pre-malignant, most people with CHIP will not develop leukemia. This condition therefore carries the designation Clonal Hematopoiesis of Indeterminate Potential (CHIP).
Mutations in only a handful of genes cause CHIP. The most common mutations, TET2 and DNMT3A, encode enzymes involved in regulating the methylation of DNA. Thus, these variants likely lead to epigenetic changes that alter transcription.76 Experiments in atherosclerosis-prone mice have demonstrated that introduction of a CHIP mutation, Tet2, accelerates atherosclerosis.74, 77 These experiments support the causality of CHIP in atherosclerotic cardiovascular disease rather than this condition merely serving as a marker of aging. The augmented cardiovascular risk conferred by CHIP mutations does not depend on classical risk factors. Moreover, CHIP mutations do not appear to associate with elevations in the traditional marker of inflammation hsCRP. The leukocytes from mice engineered to harbor Tet2 lack of function show increased expression of a number of pro-inflammatory cytokines and chemokines, including IL-1β and IL-6.
The discovery that CHIP associates with cardiovascular disease, and the absence of a tight correlation with classical risk factors or hsCRP, points to the existence of leukocyte-mediated and possibly pro-inflammatory pathways that can aggravate atherothrombosis beyond those currently recognized or tracked by CRP. For example, another CHIP mutation in a Janus kinase (JAK2) associated with myelodysplastic syndrome and polycythemia vera, particularly the JAK2V617F mutation, appears to promote thrombosis by sensitizing mutant granulocytes to formation of neutrophil extracellular traps (NETs).78 Atherosclerosis-prone mice bearing the Jak2V617F mutation develop larger lipid cores within atheroma, and activate the inflammasome/IL-1β pathway. A great deal of recent work implicates NETs in venous and arterial thrombosis.79, 80 Inhibitors of JAK2, notably the JAK1/2 blocker ruxolitinib, have approval for clinical use. Targeting carriers of this CHIP mutation with this small molecule therapeutic provides an example of using a genetic variant to allocate a specific therapy, the ultimate goal of precision medicine.
A Bright Future for Taming Atherosclerosis
In conclusion, primordial and primary prevention provide a key to stemming the worldwide spread of the atherosclerosis epidemic. With the rise of obesity and insulin resistance and diabetes, we may be losing rather than gaining ground in prevention of atherosclerosis with lifestyle intervention. Nonetheless, we should redouble our efforts to implement lifestyle intervention before pharmacologic intervention in primary prevention. In secondary prevention, however, we stand on the threshold of an exciting era of innovation. Novel therapies enable us to plumb the depths of LDL lowering. Several interventions to target triglyceride-rich lipoproteins, now considered causal in cardiovascular disease, are under investigation. Intervening on inflammation has proven promising, particularly the components of the inflammasome pathway. The recent recognition of CHIP promises to reveal previously unsuspected pathways independent of classical risk factors that promote atherogenesis and that may provide new biomarkers for allocation of therapy and identify new targets for therapy. In the war against atherosclerosis, we perhaps find ourselves at the turning point that Sir Winston Churchill recognized during the conduct of the second World War when he stated, “Now this is not the end. It is not even the beginning of the end. But it is, perhaps, the end of the beginning.”
Supplementary Material
Highlights.
In addition to lipids, novel therapies that target inflammatory pathways may address risk of atherosclerotic risk.
Not all blockers of inflammation appear capable of reducing atherosclerotic events, trials targeting oxidation, phospholipases that generate pro-inflammatory lipids, p38MAP Kinase, and administration of methotrexate have not met their endpoints.
The ability of higher doses of eicosapentaenoic acid to improve cardiovascular outcomes in individuals with hypertriglyceridemia may result in part from an anti-inflammatory action.
Neutralization of interleukin (IL)-1 beta has succeeded in improving cardiovascular outcomes in statin-treated patients stable post myocardial infarction with residual inflammation, pointing to the inflammasome to the IL-1 beta to IL-6 pathway as presenting several attractive targets for further study.
Acknowledgements
Sources of Funding
PL was funded by the National Heart, Lung, and Blood Institute (R01HL080472), the American Heart Association (18CSA34080399), and the RRM Charitable Fund.
Footnotes
Disclosures
PL is an unpaid consultant to or involved in clinical trials for Amgen, AstraZeneca, Esperion Therapeutics, Ionis Pharmaceuticals, Kowa Pharmaceuticals, Novartis, Pfizer, Sanofi-Regeneron, and XBiotech, Inc. PL is a scientific advisory board member for Amgen, Corvidia Therapeutics, DalCor Pharmaceuticals, IFM Therapeutics, Kowa Pharmaceuticals, Olatec Therapeutics, Medimmune, Novartis and XBiotech, Inc. PL’s laboratory has received research funding in the last 2 years from Novartis.
References
- 1.Goldstein JL and Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell. 2015;161:161–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ference BA, Ginsberg HN, Graham I, Ray KK, Packard CJ, Bruckert E, Hegele RA, Krauss RM, Raal FJ, Schunkert H, Watts GF, Boren J, Fazio S, Horton JD, Masana L, Nicholls SJ, Nordestgaard BG, van de Sluis B, Taskinen MR, Tokgozoglu L, Landmesser U, Laufs U, Wiklund O, Stock JK, Chapman MJ and Catapano AL. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. European Heart Journal. 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Schreiner PJ, Jacobs DR Jr, Wong ND and Kiefe CI. Twenty-Five Year Secular Trends in Lipids and Modifiable Risk Factors in a Population-Based Biracial Cohort: The Coronary Artery Risk Development in Young Adults (CARDIA) Study, 1985–2011. Journal of the American Heart Association. 2016;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hopstock LA, Bonaa KH, Eggen AE, Grimsgaard S, Jacobsen BK, Lochen ML, Mathiesen EB, Njolstad I and Wilsgaard T. Longitudinal and secular trends in total cholesterol levels and impact of lipid-lowering drug use among Norwegian women and men born in 1905–1977 in the population-based Tromso Study 1979–2016. BMJ open. 2017;7:e015001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Doyle RT Jr, Juliano RA, Jiao L, Granowitz C, Tardif JC, Ballantyne CM and Investigators R-I. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. New England Journal of Medicine. 2018. [Google Scholar]
- 6.Pradhan AD, Paynter NP, Everett BM, Glynn RJ, Amarenco P, Elam M, Ginsberg H, Hiatt WR, Ishibashi S, Koenig W, Nordestgaard BG, Fruchart JC, Libby P and Ridker PM. Rationale and design of the Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes (PROMINENT) study. Am Heart J. 2018;206:80–93. [DOI] [PubMed] [Google Scholar]
- 7.Tsimikas S, Viney NJ, Hughes SG, Singleton W, Graham MJ, Baker BF, Burkey JL, Yang Q, Marcovina SM, Geary RS, Crooke RM and Witztum JL. Antisense therapy targeting apolipoprotein(a): a randomised, double-blind, placebo-controlled phase 1 study. Lancet. 2015;386:1472–83. [DOI] [PubMed] [Google Scholar]
- 8.Gregg EW, Chen H, Wagenknecht LE, Clark JM, Delahanty LM, Bantle J, Pownall HJ, Johnson KC, Safford MM, Kitabchi AE, Pi-Sunyer FX, Wing RR and Bertoni AG. For the Look AHEAD Research Group. Association of an intensive lifestyle intervention with remission of type 2 diabetes. JAMA. 2012;308:2489–2496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Pascal B Pascal’s Pensées: New York: : E.P. Dutton, [1958]; 1958. [Google Scholar]
- 10.Gallone G, Baldetti L, Pagnesi M, Latib A, Colombo A, Libby P and Giannini F. Medical Therapy for Long-Term Prevention of Atherothrombosis Following an Acute Coronary Syndrome: JACC State-of-the-Art Review. Journal of the American College of Cardiology. 2018;72:2886–2903. [DOI] [PubMed] [Google Scholar]
- 11.Barter PJ, Nicholls S, Rye KA, Anantharamaiah GM, Navab M and Fogelman AM. Antiinflammatory properties of HDL. Circ Res. 2004;95:764–72. [DOI] [PubMed] [Google Scholar]
- 12.Steinberg D The LDL modification hypothesis of atherogenesis: an update. J Lipid Res. 2009;50 Suppl:S376–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wilensky RL and Macphee CH. Lipoprotein-associated phospholipase A(2) and atherosclerosis. Curr Opin Lipidol. 2009;20:415–20. [DOI] [PubMed] [Google Scholar]
- 14.Wilensky RL, Shi Y, Mohler ER 3rd, Hamamdzic D, Burgert ME, Li J, Postle A, Fenning RS, Bollinger JG, Hoffman BE, Pelchovitz DJ, Yang J, Mirabile RC, Webb CL, Zhang L, Zhang P, Gelb MH, Walker MC, Zalewski A and Macphee CH. Inhibition of lipoprotein-associated phospholipase A2 reduces complex coronary atherosclerotic plaque development. Nat Med. 2008;14:1059–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.White HD, Held C, Stewart R, Tarka E, Brown R, Davies RY, Budaj A, Harrington RA, Steg PG, Ardissino D, Armstrong PW, Avezum A, Aylward PE, Bryce A, Chen H, Chen MF, Corbalan R, Dalby AJ, Danchin N, De Winter RJ, Denchev S, Diaz R, Elisaf M, Flather MD, Goudev AR, Granger CB, Grinfeld L, Hochman JS, Husted S, Kim HS, Koenig W, Linhart A, Lonn E, Lopez-Sendon J, Manolis AJ, Mohler ER 3rd, Nicolau JC, Pais P, Parkhomenko A, Pedersen TR, Pella D, Ramos-Corrales MA, Ruda M, Sereg M, Siddique S, Sinnaeve P, Smith P, Sritara P, Swart HP, Sy RG, Teramoto T, Tse HF, Watson D, Weaver WD, Weiss R, Viigimaa M, Vinereanu D, Zhu J, Cannon CP and Wallentin L. Darapladib for preventing ischemic events in stable coronary heart disease. New England Journal of Medicine. 2014;370:1702–11. [DOI] [PubMed] [Google Scholar]
- 16.O’Donoghue ML, Braunwald E, White HD, Lukas MA, Tarka E, Steg PG, Hochman JS, Bode C, Maggioni AP, Im K, Shannon JB, Davies RY, Murphy SA, Crugnale SE, Wiviott SD, Bonaca MP, Watson DF, Weaver WD, Serruys PW, Cannon CP and Steen DL. Effect of darapladib on major coronary events after an acute coronary syndrome: the SOLID-TIMI 52 randomized clinical trial. Jama. 2014;312:1006–15. [DOI] [PubMed] [Google Scholar]
- 17.Bjelakovic G, Nikolova D and Gluud C. Antioxidant supplements to prevent mortality. Jama. 2013;310:1178–9. [DOI] [PubMed] [Google Scholar]
- 18.Bjelakovic G, Nikolova D and Gluud C. Meta-regression analyses, meta-analyses, and trial sequential analyses of the effects of supplementation with beta-carotene, vitamin A, and vitamin E singly or in different combinations on all-cause mortality: do we have evidence for lack of harm? PLoS One. 2013;8:e74558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Manson JE, Cook NR, Lee IM, Christen W, Bassuk SS, Mora S, Gibson H, Gordon D, Copeland T, D’Agostino D, Friedenberg G, Ridge C, Bubes V, Giovannucci EL, Willett WC, Buring JE and Group VR. Vitamin D Supplements and Prevention of Cancer and Cardiovascular Disease. New England Journal of Medicine. 2018. [DOI] [PubMed] [Google Scholar]
- 20.Tardif JC, McMurray JJ, Klug E, Small R, Schumi J, Choi J, Cooper J, Scott R, Lewis EF, L’Allier PL and Pfeffer MA. Effects of succinobucol (AGI-1067) after an acute coronary syndrome: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;371:1761–8. [DOI] [PubMed] [Google Scholar]
- 21.Libby P, Ridker PM and Hansson GK. Inflammation in atherosclerosis: from pathophysiology to practice. J Am Coll Cardiol. 2009;54:2129–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Tabas I and Glass CK. Anti-inflammatory therapy in chronic disease: challenges and opportunities. Science. 2013;339:166–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hansson GK. Inflammation and Atherosclerosis - The End of a Controversy. Circulation. 2017. [DOI] [PubMed] [Google Scholar]
- 24.Raggi P, Genest J, Giles JT, Rayner KJ, Dwivedi G, Beanlands RS and Gupta M. Role of inflammation in the pathogenesis of atherosclerosis and therapeutic interventions. Atherosclerosis. 2018;276:98–108. [DOI] [PubMed] [Google Scholar]
- 25.Fisk M, Gajendragadkar PR, Maki-Petaja KM, Wilkinson IB and Cheriyan J. Therapeutic potential of p38 MAP kinase inhibition in the management of cardiovascular disease. American journal of cardiovascular drugs : drugs, devices, and other interventions. 2014;14:155–65. [DOI] [PubMed] [Google Scholar]
- 26.Newby LK, Marber MS, Melloni C, Sarov-Blat L, Aberle LH, Aylward PE, Cai G, de Winter RJ, Hamm CW, Heitner JF, Kim R, Lerman A, Patel MR, Tanguay J-F, Lepore JJ, Al-Khalidi HR, Sprecher DL and Granger CB. Losmapimod, a novel p38 mitogen-activated protein kinase inhibitor, in non-ST-segment elevation myocardial infarction: a randomised phase 2 trial. The Lancet. 2014;384:1187–1195. [DOI] [PubMed] [Google Scholar]
- 27.Elkhawad M, Rudd JH, Sarov-Blat L, Cai G, Wells R, Davies LC, Collier DJ, Marber MS, Choudhury RP, Fayad ZA, Tawakol A, Gleeson FV, Lepore JJ, Davis B, Willette RN, Wilkinson IB, Sprecher DL and Cheriyan J. Effects of p38 mitogen-activated protein kinase inhibition on vascular and systemic inflammation in patients with atherosclerosis. JACC Cardiovasc Imaging. 2012;5:911–22. [DOI] [PubMed] [Google Scholar]
- 28.O’Donoghue ML, Glaser R, Aylward PE, Cavender MA, Crisp A, Fox KA, Laws I, Lopez-Sendon JL, Steg PG, Theroux P, Sabatine MS and Morrow DA. Rationale and design of the LosmApimod To Inhibit p38 MAP kinase as a TherapeUtic target and moDify outcomes after an acute coronary syndromE trial. Am Heart J. 2015;169:622–630 e6. [DOI] [PubMed] [Google Scholar]
- 29.O’Donoghue ML, Glaser R, Cavender MA, Aylward PE, Bonaca MP, Budaj A, Davies RY, Dellborg M, Fox KA, Gutierrez JA, Hamm C, Kiss RG, Kovar F, Kuder JF, Im KA, Lepore JJ, Lopez-Sendon JL, Ophuis TO, Parkhomenko A, Shannon JB, Spinar J, Tanguay JF, Ruda M, Steg PG, Theroux P, Wiviott SD, Laws I, Sabatine MS, Morrow DA and Investigators L-T. Effect of Losmapimod on Cardiovascular Outcomes in Patients Hospitalized With Acute Myocardial Infarction: A Randomized Clinical Trial. JAMA. 2016. [DOI] [PubMed] [Google Scholar]
- 30.Stahli BE, Gebhard C, Duchatelle V, Cournoyer D, Petroni T, Tanguay JF, Robb S, Mann J, Guertin MC, Wright RS, LLA P and Tardif JC. Effects of the P-Selectin Antagonist Inclacumab on Myocardial Damage After Percutaneous Coronary Intervention According to Timing of Infusion: Insights From the SELECT-ACS Trial. Journal of the American Heart Association. 2016;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Tardif JC, Tanguay JF, Wright SR, Duchatelle V, Petroni T, Gregoire JC, Ibrahim R, Heinonen TM, Robb S, Bertrand OF, Cournoyer D, Johnson D, Mann J, Guertin MC and L’Allier PL. Effects of the P-selectin antagonist inclacumab on myocardial damage after percutaneous coronary intervention for non-ST-segment elevation myocardial infarction: results of the SELECT-ACS trial. J Am Coll Cardiol. 2013;61:2048–55. [DOI] [PubMed] [Google Scholar]
- 32.Brown PM, Pratt AG and Isaacs JD. Mechanism of action of methotrexate in rheumatoid arthritis, and the search for biomarkers. Nature reviews Rheumatology. 2016;12:731–742. [DOI] [PubMed] [Google Scholar]
- 33.Chan ES and Cronstein BN. Mechanisms of action of methotrexate. Bull Hosp Jt Dis (2013). 2013;71 Suppl 1:S5–8. [PubMed] [Google Scholar]
- 34.Thomas S, Fisher KH, Snowden JA, Danson SJ, Brown S and Zeidler MP. Methotrexate Is a JAK/STAT Pathway Inhibitor. PLoS One. 2015;10:e0130078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ridker PM. Testing the inflammatory hypothesis of atherothrombosis: scientific rationale for the cardiovascular inflammation reduction trial (CIRT). J Thromb Haemost. 2009;7 Suppl 1:332–9. [DOI] [PubMed] [Google Scholar]
- 36.Ridker PM. Clinician’s Guide to Reducing Inflammation to Reduce Atherothrombotic Risk: JACC Review Topic of the Week. J Am Coll Cardiol. 2018. [DOI] [PubMed] [Google Scholar]
- 37.Imazio M, Brucato A, Cemin R, Ferrua S, Maggiolini S, Beqaraj F, Demarie D, Forno D, Ferro S, Maestroni S, Belli R, Trinchero R, Spodick DH, Adler Y and Investigators I. A randomized trial of colchicine for acute pericarditis. New England Journal of Medicine. 2013;369:1522–8. [DOI] [PubMed] [Google Scholar]
- 38.Nidorf SM, Eikelboom JW, Budgeon CA and Thompson PL. Low-dose colchicine for secondary prevention of cardiovascular disease. J Am Coll Cardiol. 2013;61:404–10. [DOI] [PubMed] [Google Scholar]
- 39.Vaidya K, Arnott C, Martinez GJ, Ng B, McCormack S, Sullivan DR, Celermajer DS and Patel S. Colchicine Therapy and Plaque Stabilization in Patients With Acute Coronary Syndrome: A CT Coronary Angiography Study. JACC Cardiovasc Imaging. 2017. [DOI] [PubMed] [Google Scholar]
- 40.Leung YY, Yao Hui LL and Kraus VB. Colchicine--Update on mechanisms of action and therapeutic uses. Semin Arthritis Rheum. 2015;45:341–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Martinez GJ, Robertson S, Barraclough J, Xia Q, Mallat Z, Bursill C, Celermajer DS and Patel S. Colchicine Acutely Suppresses Local Cardiac Production of Inflammatory Cytokines in Patients With an Acute Coronary Syndrome. Journal of the American Heart Association. 2015;4:e002128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, Miller DK, Molineaux SM, Weidner JR, Aunins J and et al. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature. 1992;356:768–74. [DOI] [PubMed] [Google Scholar]
- 43.Latz E, Xiao TS and Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13:397–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Baldrighi M, Mallat Z and Li X. NLRP3 inflammasome pathways in atherosclerosis. Atherosclerosis. [DOI] [PubMed] [Google Scholar]
- 45.Mangan MSJ, Olhava EJ, Roush WR, Seidel HM, Glick GD and Latz E. Targeting the NLRP3 inflammasome in inflammatory diseases. Nat Rev Drug Discov. 2018. [DOI] [PubMed] [Google Scholar]
- 46.Libby P Interleukin-1 Beta as a Target for Atherosclerosis Therapy: Biological Basis of CANTOS and Beyond. J Am Coll Cardiol. 2017;70:2278–2289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ridker PM, Thuren T, Zalewski A and Libby P. Interleukin-1β inhibition and the prevention of recurrent cardiovascular events: rationale and design of the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). Am Heart J. 2011;162:597–605. [DOI] [PubMed] [Google Scholar]
- 48.Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, Kastelein JJP, Cornel JH, Pais P, Pella D, Genest J, Cifkova R, Lorenzatti A, Forster T, Kobalava Z, Vida-Simiti L, Flather M, Shimokawa H, Ogawa H, Dellborg M, Rossi PRF, Troquay RPT, Libby P and Glynn RJ. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. New England Journal of Medicine. 2017;377:1119–1131. [DOI] [PubMed] [Google Scholar]
- 49.Ridker PM, MacFadyen JG, Everett BM, Libby P, Thuren T, Glynn RJ and Group CT. 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. [DOI] [PubMed] [Google Scholar]
- 50.Ridker P, Ballantyne C, Fohnseca F, Koenig W, Shimokawa H and Everett B. Relationships of Interleukin-6 Reduction to Atherosclerotic Events and All-cause Mortality: Analyses from the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) European Heart Journal. 2018;in press. [DOI] [PubMed] [Google Scholar]
- 51.Fredman G and Tabas I. Boosting Inflammation Resolution in Atherosclerosis: The Next Frontier for Therapy. Am J Pathol. 2017;187:1211–1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Libby P, Salomon RN, Payne DD, Schoen FJ and Pober JS. Functions of vascular wall cells related to the development of transplantation-associated coronary arteriosclerosis. Transplant Proc. 1989;21:3677–3684. [PubMed] [Google Scholar]
- 53.Libby P and Ridker PM. Novel inflammatory markers of coronary risk: theory versus practice. Circulation. 1999;100:1148–50. [DOI] [PubMed] [Google Scholar]
- 54.Libby P History of discovery: inflammation in atherosclerosis. Arteriosclerosis and Thrombosis: a journal of vascular biology. 2012;32:2045–2051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Coll RC, Robertson AA, Chae JJ, Higgins SC, Munoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, Croker DE, Butler MS, Haneklaus M, Sutton CE, Nunez G, Latz E, Kastner DL, Mills KH, Masters SL, Schroder K, Cooper MA and O’Neill LA. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med. 2015;21:248–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Hingorani AD and Casas JP. Interleukin-6 Receptor Mendelian Randomisation Analysis (IL6R MR) Consortium. The interleukin-6 receptor as a target for prevention of coronary heart disease: a mendelian randomisation analysis. Lancet. 2012;379:1214–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sarwar N, Butterworth AS, Freitag DF, Gregson J, Willeit P, Gorman DN, Gao P, Saleheen D, Rendon A, Nelson CP, Braund PS, Hall AS, Chasman DI, Tybjaerg-Hansen A, Chambers JC, Benjamin EJ, Franks PW, Clarke R, Wilde AA, Trip MD, Steri M, Witteman JC, Qi L, van der Schoot CE, de Faire U, Erdmann J, Stringham HM, Koenig W, Rader DJ, Melzer D, Reich D, Psaty BM, Kleber ME, Panagiotakos DB, Willeit J, Wennberg P, Woodward M, Adamovic S, Rimm EB, Meade TW, Gillum RF, Shaffer JA, Hofman A, Onat A, Sundstrom J, Wassertheil-Smoller S, Mellstrom D, Gallacher J, Cushman M, Tracy RP, Kauhanen J, Karlsson M, Salonen JT, Wilhelmsen L, Amouyel P, Cantin B, Best LG, Ben-Shlomo Y, Manson JE, Davey-Smith G, de Bakker PI, O’Donnell CJ, Wilson JF, Wilson AG, Assimes TL, Jansson JO, Ohlsson C, Tivesten A, Ljunggren O, Reilly MP, Hamsten A, Ingelsson E, Cambien F, Hung J, Thomas GN, Boehnke M, Schunkert H, Asselbergs FW, Kastelein JJ, Gudnason V, Salomaa V, Harris TB, Kooner JS, Allin KH, Nordestgaard BG, Hopewell JC, Goodall AH, Ridker PM, Holm H, Watkins H, Ouwehand WH, Samani NJ, Kaptoge S, Di Angelantonio E, Harari O and Danesh J. IL6R Genetics Consortium Emerging Risk Factors Collaboration. Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies. Lancet. 2012;379:1205–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Schuett H, Oestreich R, Waetzig GH, Annema W, Luchtefeld M, Hillmer A, Bavendiek U, von Felden J, Divchev D, Kempf T, Wollert KC, Seegert D, Rose-John S, Tietge UJ, Schieffer B and Grote K. Transsignaling of interleukin-6 crucially contributes to atherosclerosis in mice. Arteriosclerosis and Thrombosis: a journal of vascular biology. 2012;32:281–90. [DOI] [PubMed] [Google Scholar]
- 59.Rose-John S IL-6 trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL-6. Int J Biol Sci. 2012;8:1237–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Morieri ML, Passaro A and Zuliani G. Interleukin-6 “Trans-Signaling” and Ischemic Vascular Disease: The Important Role of Soluble gp130. Mediators Inflamm. 2017;2017:1396398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Jones SA, Scheller J and Rose-John S. Therapeutic strategies for the clinical blockade of IL-6/gp130 signaling. J Clin Invest. 2011;121:3375–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Garlanda C, Dinarello CA and Mantovani A. The interleukin-1 family: back to the future. Immunity. 2013;39:1003–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Freigang S, Ampenberger F, Weiss A, Kanneganti TD, Iwakura Y, Hersberger M and Kopf M. Fatty acid-induced mitochondrial uncoupling elicits inflammasome-independent IL-1alpha and sterile vascular inflammation in atherosclerosis. Nature immunology. 2013;14:1045–53. [DOI] [PubMed] [Google Scholar]
- 64.Sheedy FJ and Moore KJ. IL-1 signaling in atherosclerosis: sibling rivalry. Nature immunology. 2013;14:1030–2. [DOI] [PubMed] [Google Scholar]
- 65.Hong DS, Janku F, Naing A, Falchook GS, Piha-Paul S, Wheler JJ, Fu S, Tsimberidou AM, Stecher M, Mohanty P, Simard J and Kurzrock R. Xilonix, a novel true human antibody targeting the inflammatory cytokine interleukin-1 alpha, in non-small cell lung cancer. Invest New Drugs. 2015;33:621–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.El Sayed H, Kerensky R, Stecher M, Mohanty P and Davies M. A randomized phase II study of Xilonix, a targeted therapy against interleukin 1alpha, for the prevention of superficial femoral artery restenosis after percutaneous revascularization. Journal Of Vascular Surgery. 2016;63:133–41 e1. [DOI] [PubMed] [Google Scholar]
- 67.Tsiantoulas D, Diehl CJ, Witztum JL and Binder CJ. B Cells and humoral immunity in atherosclerosis. Circ Res. 2014;114:1743–1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Chyu K-Y, Dimayuga PC and Shah PK. Vaccine against arteriosclerosis: an update. Therapeutic Advances in Vaccines. 2017:205101361769375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Gistera A, Hermansson A, Strodthoff D, Klement ML, Hedin U, Fredrikson GN, Nilsson J, Hansson GK and Ketelhuth DF. Vaccination against T-cell epitopes of native ApoB100 reduces vascular inflammation and disease in a humanized mouse model of atherosclerosis. J Intern Med. 2017;281:383–397. [DOI] [PubMed] [Google Scholar]
- 70.Wolf D, Gerhardt T and Ley K. Vaccination to Prevent Cardiovascular Disease In: Zirlik A, Bode C and Gawaz M, eds. Platelets, Haemostasis and Inflammation Cham: Springer International Publishing; 2017: 29–52. [Google Scholar]
- 71.Nus M and Mallat Z. Immune-mediated mechanisms of atherosclerosis and implications for the clinic. Expert Rev Clin Immunol. 2016;12:1217–1237. [DOI] [PubMed] [Google Scholar]
- 72.Gibson CJ and Steensma DP. New Insights from Studies of Clonal Hematopoiesis. Clinical Cancer Research. 2018. [DOI] [PubMed] [Google Scholar]
- 73.Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, Lindsley RC, Mermel CH, Burtt N, Chavez A, Higgins JM, Moltchanov V, Kuo FC, Kluk MJ, Henderson B, Kinnunen L, Koistinen HA, Ladenvall C, Getz G, Correa A, Banahan BF, Gabriel S, Kathiresan S, Stringham HM, McCarthy MI, Boehnke M, Tuomilehto J, Haiman C, Groop L, Atzmon G, Wilson JG, Neuberg D, Altshuler D and Ebert BL. Age-related clonal hematopoiesis associated with adverse outcomes. New England Journal of Medicine. 2014;371:2488–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, McConkey M, Gupta N, Gabriel S, Ardissino D, Baber U, Mehran R, Fuster V, Danesh J, Frossard P, Saleheen D, Melander O, Sukhova GK, Neuberg D, Libby P, Kathiresan S and Ebert BL. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. New England Journal of Medicine. 2017;377:111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Dorsheimer L, Assmus B, Rasper T, Ortmann CA, Ecke A, Abou-El-Ardat K, Schmid T, Brüne B, Wagner S, Serve H, Hoffmann J, Seeger F, Dimmeler S, Zeiher AM and Rieger MA. Mutations contributing to clonal hematopoiesis are associated with poor prognosis in chronic ischemic heart failure JAMA Cardiol. 2018;in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Costantino S, Libby P, Kishore R, Tardif J-C, El-Osta A and Paneni F. Epigenetics and precision medicine in cardiovascular patients: from basic concepts to the clinical arena. European Heart Journal. 2017:ehx568–ehx568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Fuster JJ, MacLauchlan S, Zuriaga MA, Polackal MN, Ostriker AC, Chakraborty R, Wu C-L, Sano S, Muralidharan S, Rius C, Vuong J, Jacob S, Muralidhar V, Robertson AAB, Cooper MA, Andrés V, Hirschi KK, Martin KA and Walsh K. Clonal hematopoiesis associated with Tet2 deficiency accelerates atherosclerosis development in mice. Science. 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wolach O, Sellar RS, Martinod K, Cherpokova D, McConkey M, Chappell RJ, Silver AJ, Adams D, Castellano CA, Schneider RK, Padera RF, DeAngelo DJ, Wadleigh M, Steensma DP, Galinsky I, Stone RM, Genovese G, McCarroll SA, Iliadou B, Hultman C, Neuberg D, Mullally A, Wagner DD and Ebert BL. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci Transl Med. 2018;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Martinod K and Wagner DD. Thrombosis: tangled up in NETs. Blood. 2014;123:2768–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Savchenko AS, Martinod K, Seidman MA, Wong SL, Borissoff JI, Piazza G, Libby P, Goldhaber SZ, Mitchell RN and Wagner DD. Neutrophil extracellular traps form predominantly during the organizing stage of human venous thromboembolism development. J Thromb Haemost. 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.

