Skip to main content
Top
Gepubliceerd in: Netherlands Heart Journal 1/2022

Open Access 17-08-2021 | Review Article

Targeting residual inflammatory risk in coronary disease: to catch a monkey by its tail

Auteurs: A. T. L. Fiolet, T. S. J. Opstal, M. J. M. Silvis, J. H. Cornel, A. Mosterd

Gepubliceerd in: Netherlands Heart Journal | Uitgave 1/2022

Abstract

Patients with coronary disease remain at high risk for future cardiovascular events, even with optimal risk factor modification, lipid-lowering drugs and antithrombotic regimens. A myriad of inflammatory pathways contribute to progression of the atherosclerotic burden in these patients. Only in the last few years has the inflammatory biology of atherosclerosis translated into clinical therapeutic options. Low-dose colchicine can provide a clinically relevant reduction in the risk for composite and individual major cardiovascular outcomes in patients with acute and chronic coronary syndromes. Among others, its anti-inflammatory effects in atherosclerosis seem to be related to neutrophil recruitment and adhesion, inflammasome inhibition, and morphological changes in platelets and platelet aggregation. Future research is aimed at further elucidating its particular mechanism of action, as well as identifying patients with the highest expected benefit and evaluating efficacy in other vascular beds. These data will help to formulate the role of colchicine and other anti-inflammatory drugs in patients with coronary disease and atherosclerosis in general in the near future.

Introduction

Atherosclerosis is a slowly progressive disease. By focusing on lifestyle changes, management of dyslipidaemia and intensive treatment with antithrombotic and anticoagulant agents atherosclerosis-associated mortality rates were lowered by 25–50% in the last two decades [13]. Lipid-lowering drugs have had the most notable role in this process, with an estimated 20% relative risk reduction for a major adverse cardiovascular event following every millimole per litre (mmol/l) decrease in low-density lipoprotein (LDL)-cholesterol [4]. Nevertheless, the atherothrombotic risk for patients with coronary disease remains high. Depending on co-morbidities and the extent of the atherosclerotic burden, a given patient will have a 5-year risk of up to 20% for myocardial infarction, ischaemic stroke or death [5, 6].
An inter-individual varying proportion of this risk is explained by the myriad of inflammatory pathways that contribute to progression of the atherosclerotic burden and plaque destabilisation, often referred to as the residual inflammatory risk [7]. Only in the last few years has the inflammatory biology of atherosclerosis translated into several clinical therapeutic options that we will discuss below.

The magnitude of inflammation as residual risk

Contemporary studies on LDL-cholesterol have provided insight into the magnitude of the remaining risk after optimal treatment of dyslipidaemia. In the Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk (FOURIER) trial, intensive lipid lowering was attained with statins and the proprotein convertase subtilisin/kexin type 9‑inhibiting molecular antibody evolocumab. Patients achieved a median LDL-cholesterol of 0.78 mmol/l. Even with this state-of-the-art lipid-lowering strategy, 9.8% of patients developed a major adverse cardiovascular event after a median follow-up of just over 2 years [8]. The most important independent contributors to such risk are hypertriglyceridaemia, residual thrombotic risk, diabetes-associated morbidity and low-grade inflammation [9]. When assessing the relevance of the last-mentioned factor, high-sensitivity C‑Reactive Protein (hsCRP) proved a robust prognostic risk marker. Patient-level meta-analyses demonstrated that each standard deviation increment in log-normalised hsCRP was associated with a 37% increase in the relative risk for coronary heart disease. This risk is similar to that associated with an increase in systolic blood pressure (35% relative risk increase per standard deviation increment) and twice as high as the risk associated with an increase in total cholesterol (16% relative risk increase per standard deviation increment) [10].
The pleiotropic effects of statins can be used to estimate the proportional contribution of the residual inflammatory risk. In the Pravastatin or Atorvastatin Evaluation and Infection Therapy-Thrombolysis in Myocardial Infarction 22 (PROVE IT-TIMI 22) trial and in The Improved Reduction of Outcomes: Vytorin Efficacy International Trial (IMPROVE-IT), approximately one third of patients had an hsCRP equal to or above 2 mg/l after achieving an LDL-cholesterol below 1.8 mmol/l. Depending on treatment intensity, statin treatment could reduce median hsCRP by a third, with a greater hsCRP reduction associated with a greater relative risk reduction for cardiovascular events [11, 12]. In addition, the risk reduction following the lowering of hsCRP levels in these patients occurred irrespective of the change in LDL-cholesterol, emphasising the independence of the inflammatory pathway [13].
Mendelian randomisation studies later proved that hsCRP was not causally related to cardiovascular disease [14]. HsCRP, however, is a downstream derivate of the interleukin‑1 and interleukin‑6 inflammatory cascade, which does have a causal role in atherothrombosis [15, 16].
The above-mentioned findings shaped the ‘inflammation hypothesis’ for atherosclerotic disease. Subsequently, multiple both broad-acting as well as targeted agents were introduced to investigate whether anti-inflammatory treatment would truly translate into beneficial effects for patients with atherosclerosis.

Efficacy of anti-inflammatory drugs in atherosclerosis and coronary disease

Anti-inflammatory agents with unfavourable effects on cardiovascular events

Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used anti-inflammatory drugs. Their anti-inflammatory properties are not suitable for addressing inflammation in atherosclerosis, as they are consistently associated with an increased risk for major coronary events, with the exception of low-dose acetylsalicylic acid [17]. This is a consequence of the dose-dependent pharmacodynamic properties of the drug. Acetylsalicylic acid inhibits cyclooxygenase (COX)-1 and COX‑2. COX‑1 inhibition reduces thromboxane A2-induced platelet aggregation at low doses, accounting for its atherothrombotic protective effects. At higher doses, the anti-inflammatory properties driven by COX‑2 inhibition arise [18]. Many novel NSAIDs are designed to selectively inhibit only COX‑2. COX‑2 inhibition also leads to undesired cardiovascular effects, such as prostaglandin E2-mediated sodium and water retention, vascular endothelium prostacyclin-mediated platelet activation and vasoconstriction, increasing the risk for cardiovascular events [19].
Glucocorticoids play an integral role in the management of multiple inflammatory conditions. However, many unfavourable side effects such as hypertension, impaired glucose tolerance and obesity following long-term treatment with corticosteroids render them unsuitable for dampening the inflammatory component of atherosclerosis [20].

Anti-inflammatory agents without effects on cardiovascular events

One of the first attempts to specifically target inflammation in coronary disease was by inhibition of lipoprotein-associated phospholipase A2 (Lp-PLA2). Lp-PLA2 is bound to LDL-cholesterol and plays a role in oxidative modification within the vascular wall. It increases vascular inflammation in atherosclerosis. The Lp-PLA2 inhibitor darapladib was tested in two major trials in almost 20,000 patients with chronic coronary disease and recent acute coronary syndrome. Neither trial demonstrated a reduction in the risk for major cardiovascular events [21, 22]. A variant of the compound inhibiting the secretory form of PLA2, varespladib, was associated with a higher rate of recurrent myocardial infarction [23].
Parallel to these efforts, inhibition of the broad p38 mitogen-activated protein (MAP) kinases system was proposed. The p38 MAP kinase system participates in various intracellular signalling routes and is active in endothelial cells, smooth muscle cells and leucocytes. The p38 MAP kinase system inhibitor losmapimod, however, did not show any clinical effect [24].
Methotrexate is a broad-acting immunomodulating drug that inhibits DNA synthesis by competing with folate synthesis and by inhibiting T‑cell adhesion molecules and T‑cell activity. The drug is used in a wide array of auto-immune and oncological conditions. The ability of methotrexate to dampen atherosclerotic inflammation was studied in over 4500 patients with chronic coronary disease and type 2 diabetes or metabolic syndrome. The trial was ceased prematurely for reasons of futility, being unable to demonstrate any sign of clinical benefit [25].
One of the common denominators in these trials was the absence of any evident biochemical response to the treatment. These data strengthened the hypothesis that effective inhibition of the inflammatory pathways in atherosclerosis should comprise targeted and detectable interleukin‑1 and interleukin‑6 inhibition.

Anti-inflammatory agents with beneficial effects on cardiovascular events

To date, two anti-inflammatory compounds have shown to be efficacious in reducing the risk for major cardiovascular outcomes in patients with coronary disease. The Canakinumab Antiinflammatory Thrombosis Outcome Study (CANTOS) was the first to prove that modulating the inflammatory pathway in atherosclerosis reduces major adverse cardiovascular events in patients with a recent history of myocardial infarction and hsCRP equal to or above 2 mg/l. The CANTOS trial randomised 10,061 patients to three subcutaneous doses of canakinumab, the selective interleukin-1β inhibitor, or placebo. Canakinumab 150 mg reduced the risk for the composite end point of non-fatal myocardial infarction, non-fatal stroke or cardiovascular death by 15% and for the composite end point of non-fatal myocardial infarction, non-fatal stroke, cardiovascular death or urgent hospitalisation for angina leading to urgent revascularisation by 17% [16]. Although these results were ground breaking, canakinumab is a costly drug and the findings of the trial have not led to registration of the drug for secondary prevention in cardiovascular disease.
Shortly after CANTOS had demonstrated that selective cytokine inhibition is effective after recent myocardial infarction, two major clinical trials reported on the efficacy of the broad-acting anti-inflammatory drug colchicine in both acute and chronic coronary disease.
The Colchicine Cardiovascular Outcomes Trial (COLCOT) randomised 4745 patients within 30 days of myocardial infarction to colchicine 0.5 mg or placebo once daily. The trial showed a 23% relative risk reduction for the composite end point of death from cardiovascular causes, resuscitated cardiac arrest, myocardial infarction, stroke or urgent hospitalisation for angina leading to coronary revascularisation as compared to placebo [26]. The Low-dose Colchicine 2 (LoDoCo2) trial randomised 5522 patients with chronic coronary disease to 0.5 mg colchicine or placebo once daily and demonstrated a 31% relative risk reduction for the composite end point of cardiovascular death, myocardial infarction, ischaemic stroke or ischaemia-driven revascularisation as compared to placebo [27].
In COLCOT as well as in the LoDoCo2 trial, treatment benefit appeared soon after initiation and continued to accrue over time. Both trials recruited participants treated with optimal medical therapy and irrespective of inflammatory status. The trials excluded patients with severe heart or renal failure, since colchicine is partially eliminated via renal excretion [28]. Exploratory analyses revealed no interaction of treatment with relevant clinical subgroups or concomitant drugs. In the 30-day open-label run-in period used in the LoDoCo2 trial, 9.3% of patients were intolerant to the drug, most often due to benign gastro-intestinal upset. Per the pragmatic design of the trials, major safety parameters were only investigated in an explorative manner. An increased occurrence of pneumonia, seen both with canakinumab in CANTOS and with colchicine in COLCOT, was not observed in the LoDoCo2 trial. The majority of patients in the trials were treated with intensive lipid-lowering agents and many with high-dose statins. No increased occurrence of severe myotoxicity was observed.

Mechanism of action of colchicine

Colchicine, originally extracted from the autumn crocus (Colchicum autumnale), is a widely available drug. It is used chronically to prevent gout and treat familial Mediterranean fever and intermittently for the treatment of pericarditis [29, 30].
Low-dose colchicine is associated with a 30–40% reduction in median hsCRP levels and a 16% reduction in median interleukin‑6 levels in patients with chronic coronary disease [31, 32]. These effects occur early after treatment initiation and are less pronounced than the anti-inflammatory response seen with canakinumab following myocardial infarction [16].
The mechanism of action of colchicine in atherosclerosis relates to the inhibition of microtubule self-assembly. Microtubules are structural components found in various static and dynamic processes of the cell. They form the cytoskeleton, and contribute to the shape and movement of cells. They are also the structures that are used for moving components within the cell. This facilitates cell division (mitosis), intracellular trafficking and secretion of cytokine and chemokines, as well as regulation of ion channels [28]. Microtubules are thus essential in cell movement and activation, the principal part of any inflammatory response. Among others, the anti-inflammatory effects of colchicine are related to the following established concepts in the pathogenesis of atherosclerosis: (1) neutrophil recruitment and adhesion, (2) inflammasome inhibition and (3) morphological changes in platelets and platelet aggregation (Fig. 1).

Neutrophil chemotaxis, activation and adhesion

Atherosclerotic plaque inflammation is driven by pathological stimuli such as oxidised LDL-cholesterol, blood-pressure-related shear stress and reduced bioavailability of nitric oxide caused by tobacco smoking. The inflammatory response cascades an increasing influx of inflammatory cells such as neutrophils and monocytes that contribute to development of a thin capped fibroatheroma. Structural disturbances of these fibroatheromatas, classically occurring at the cap and shoulders, may lead to erosion or rupture, precipitating atherothrombotic events [33].
Colchicine accumulates with a three- to four-fold higher concentration in neutrophils as compared to mononuclear leucocytes [34]. It reduces the mobility and deformability of neutrophils, thereby inhibiting cell movement and endothelial extravasation. In addition, endothelial adhesion is reduced by means of L‑selectin shedding [35, 36]. Protein expression related to neutrophil degranulation is attenuated by colchicine in patients with coronary disease [32]. The decreased neutrophil mobility and adhesion limits neutrophils entering the plaque. Computed tomography analyses accordingly show fewer high-risk features in patients treated with colchicine than in controls [37].

Inflammasome inhibition

The microtubule inhibitory properties of colchicine specifically affect inflammasome activity, localised in many immune cells, and macrophages in particular. Activation of the nucleotide-binding oligomerisation domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome leads to caspase‑1 activation and subsequent interleukin-1β and interleukin-18 expression [38]. These cytokines have a flywheel effect in the inflammatory response and increase cell activation and recruitment [39]. Crystalloid structures, among others, contribute to NLRP3 inflammasome activation. The role of this crystal-induced inflammation in gouty arthritis is a result of monosodium urate crystals [38]. In the atherosclerotic plaque, crystallisation of cholesterol can initiate the NLRP3 inflammasome activation and interleukin-1β expression [40]. The attenuating effects of colchicine on NLRP3 inflammasome activity and interleukin-1β may explain its clinical effects in atherosclerosis.

Platelet morphological changes and aggregation

Microtubules support the shape of non-activated platelets and contribute to the internal reorganisation for shape change after activation. Microtubules and filaments are also structural parts of the pseudopodia of the platelet, used for motility and aggregation. Historical in vitro evidence shows that moderate doses of colchicine impair internal transformation and late aggregation of platelets. High-dose colchicine leads to complete microtubule dissolution and fully halts platelet aggregation [41]. More recently, colchicine was shown to reduce maximal platelet aggregation even after prior COX‑1 and P2Y12 adenosine diphosphate receptor inhibition with acetylsalicylic acid and clopidogrel [42]. Leucocyte-platelet aggregation, rather than platelet-platelet aggregation, seems to be preferentially affected [43]. In vivo evidence of such mechanisms is not yet available. There are no clinical studies that have specially addressed haemostasis or bleeding events associated with colchicine.

Clinical implementation, unresolved issues and future perspectives

Now multiple large cardiovascular outcome trials have unequivocally demonstrated the clinical efficacy of modulating inflammatory pathways in coronary disease, solid proof of principle of the inflammatory hypothesis in atherosclerotic disease has been provided. When pooled, the results of the low-dose colchicine studies show a relative risk reduction of 25% (relative risk 0.75, 95% confidence interval 0.61–0.91) for major cardiovascular events, with consistent effects on the individual components myocardial infarction, ischaemia-driven coronary revascularisation and stroke [44]. This additional risk reduction, achieved in patients on optimal medical therapy, is a clinically relevant effect that is equal to the risk reduction that can be reached with intensive lipid-lowering therapy and antithrombotic therapy [4, 45]. When trying to translate such effect sizes into daily practice, consider a typical out-patient clinic example of a cigarette smoking 65-year-old man with coronary disease. If treated with acetylsalicylic acid, anti-hypertensive drugs lowering his systolic blood pressure to below 140 mm Hg, and lipid-lowering drugs to achieve an LDL-cholesterol of 1.8 mmol/l, he still has a 10-year risk of 36% for a major cardiovascular event and his lifetime risk will approach 65%. The addition of anti-inflammatory treatment with low-dose colchicine could provide an 8.4% absolute risk reduction to his 10 -year risk for a cardiovascular event and a 9.4% absolute risk reduction for his lifetime risk. This translates into almost 2 years free of major cardiovascular events. (Fig. 2; [46]).
We are only at the beginning of translating the findings from trials on anti-inflammatory therapy into clinical practice. When evaluating the role of such drugs as adjuvant in coronary disease, several questions remain to be answered, among which are the following:
1.
How can we identify patients that will benefit most from anti-inflammatory therapy?
 
2.
Will long-term anti-inflammatory treatment translate into a reduction in all-cause mortality?
 
3.
Do patients with atherosclerotic disease of other vascular beds benefit equally?
 
4.
What is the role of alternative highly targeted anti-inflammatory drugs acting on similar pathways?
 
First, when identifying the ‘right’ patient, the Reynolds risk score and the Secondary Manifestations of ARTerial disease (SMART) risk score demonstrated additional prognostic value of using hsCRP for risk stratification, but have not yet led to clinical recommendation in European guidelines [47, 48]. Clinical risk calculators such as U‑Prevent.com may contribute to estimating the benefit for individual patients based on clinical parameters. The clinical evidence for colchicine was accrued in an ‘all-comer’ population of coronary disease without pre-selection based on inflammatory biomarkers. Multiple ongoing clinical studies are focusing on particular subpopulations (Tab. 1): The ongoing Colchicine Cardiovascular Outcomes in Acute Coronary Syndrome trial (COLCARDIO-ACS; ACTRN12616000400460) aims to recruit patients with recent acute coronary syndrome with increased hsCRP levels and hereby determine whether baseline inflammatory status modifies treatment effect. Results are expected in 2025. A substudy of the Colchicine and Spironolactone in Patients with Myocardial Infarction/SYNERGY Stent Registry (CLEAR SYNERGY; NCT03048825 and NCT03874338) aims to assess the effect of colchicine on neutrophil activation in response to ST-elevated myocardial infarction, and thus to identify clinical and genetic factors contributing to treatment response.
Table 1
Ongoing and planned clinical outcome trials of colchicine in atherosclerosis
Principle investigator
Acronym;
Trial Registration
Study name
Patients
Population
Regimen
Maximal follow-up
Primary end point
Estimated completion date
Javed N
No acronym;
NCT04218786
Effect of Colchicine in Patients with Myocardial Infarction
 800
Myocardial infarction in previous 30 days
Colchicine 0.5 mg once daily vs placebo
3 months
Composite of cardiovascular death, non-fatal myocardial infarction, resuscitated cardiac arrest or hospitalisation for unstable angina
Mid-2021
Kelly P
CONVINCE; NCT02898610
Colchicine for Prevention of Vascular Inflammation in Non-cardio Embolic Stroke
2623
Prior ischaemic stroke or transient ischaemic attack
Colchicine 0.5 mg once daily vs usual care
5 years
Any recurrence of non-fatal ischaemic stroke, non-fatal hospitalisation for unstable angina, myocardial infarction, cardiac arrest or vascular death
End of 2021
Patel S
COLCARDIO-ACS; ACTRN12616000400460
Colchicine Cardiovascular Outcomes in Acute Coronary Syndrome
3000
Acute coronary syndrome in previous 30–45 days and high-sensitivity
C‑reactive protein ≥ 2 mg/l
Colchicine 0.5 mg once daily vs placebo
3 years
Composite of cardiovascular death, acute coronary syndrome, urgent revascularisation or non-fatal stroke
2025
Jolly S
CLEAR SYNERGY; NCT03048825
Colchicine and Spironolactone in Patients with Myocardial Infarction/SYNERGY Stent Registry
7000
Acute myocardial infarction with percutaneous coronary revascularisation
Colchicine 0.5 mg once daily vs spironolactone 25 mg once daily vs colchicine 0.5 mg and spironolactone 25 mg vs placebo
3 years
Composite of death, recurrent target vessel myocardial infarction, stroke or ischaemia-driven target vessel revascularisation
2025
Second, when evaluating the net clinical efficacy of the drug, it should be noted that although the combined colchicine trials recruited over 10,000 patients, all-cause mortality in the trials was low. The majority of deaths were non-cardiovascular and the decreased number of cardiovascular deaths was counterbalanced by an increase in non-cardiovascular deaths. The low incidence of fatalities and varying study durations restrict interpretation of these findings. If an intervention delays cardiovascular death substantially, other causes of death may gain the upper hand. Additional studies and long-term follow-up studies will give way to methodological approaches such as competing risk analyses to evaluate long-term effects and evaluate the overall benefit of the drug [49].
Third, when considering other vascular beds, particular interest is given to peripheral artery disease and cerebrovascular disease. None of the trials have yet reported on the incidence or progression of peripheral artery disease. Patients with symptomatic peripheral artery disease are at high risk for adverse cardiovascular events and still have an impressive 10% annual risk for death after revascularisation [50]. Based on the common pathophysiological grounds of coronary disease and peripheral artery disease, a particular protective effect is expected in this population. When considering the effect in cerebrovascular disease, the pooled relative risk reduction of 46% for the risk of strokes was a notable finding in the colchicine trials [44]. The Colchicine for Prevention of Vascular Inflammation in Non-cardio Embolic Stroke (CONVINCE; NCT02898610) trial is designed to investigate the effect of colchicine 0.5 mg once daily on the risk for major adverse cerebrovascular and cardiac events in patients with prior cerebrovascular disease in particular. The trial is estimated to complete recruitment at the end of 2021.
Lastly, drugs targeting similar pathways with different pharmacokinetic properties may become increasingly relevant. The interleukin‑1 receptor antagonist anakinra was never able to progress to phase 3 studies in coronary disease. However, the novel interleukin-6 ligand monoclonal antibody ziltivekimab seems effective at reducing multiple inflammatory and thrombotic biomarkers relevant to atherosclerosis. These data have led to the initiation of a large-scale cardiovascular outcomes trial of ziltivekimab compared with placebo in patients with chronic kidney disease who have increased hsCRP and established cardiovascular disease [51]. Selective NLRP3 inflammasome inhibitors are being developed, but no clinical studies have been registered yet.

Conclusion

In conclusion, in this era of optimal risk factor modification, intensive lipid-lowering and antithrombotic therapy, patients with coronary disease remain at a significant risk for major cardiovascular events. This residual risk partially emanates from the inflammatory drivers of atherosclerosis. The anti-inflammatory drug colchicine can provide a clinically relevant reduction for the risk of major cardiovascular events. Many of its mechanisms of action as well as its efficacy in other atherosclerotic conditions remain to be demonstrated in the near future.

Conflict of interest

A.T.L. Fiolet, T.S.J. Opstal, M.J.M. Silvis, J.H. Cornel and A. Mosterd declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Onze productaanbevelingen

Netherlands Heart Journal

Het Netherlands Heart Journal wordt uitgegeven in samenwerking met de Nederlandse Vereniging voor Cardiologie en de Nederlandse Hartstichting. Het tijdschrift is Engelstalig en wordt gratis beschikbaa ...

Literatuur
1.
go back to reference Roth GA, Huffman MD, Moran AE, et al. Global and regional patterns in cardiovascular mortality from 1990 to 2013. Circulation. 2015;132:1667–78.PubMedCrossRef Roth GA, Huffman MD, Moran AE, et al. Global and regional patterns in cardiovascular mortality from 1990 to 2013. Circulation. 2015;132:1667–78.PubMedCrossRef
2.
go back to reference Shah NS, Molsberry R, Rana JS, et al. Heterogeneous trends in burden of heart disease mortality by subtypes in the United States, 1999–2018: observational analysis of vital statistics. BMJ. 2020;370:m2688.PubMedPubMedCentralCrossRef Shah NS, Molsberry R, Rana JS, et al. Heterogeneous trends in burden of heart disease mortality by subtypes in the United States, 1999–2018: observational analysis of vital statistics. BMJ. 2020;370:m2688.PubMedPubMedCentralCrossRef
3.
go back to reference Ezzati M, Obermeyer Z, Tzoulaki I, Mayosi BM, Elliott P, Leon DA. Contributions of risk factors and medical care to cardiovascular mortality trends. Nat Rev Cardiol. 2015;12:508–30.PubMedPubMedCentralCrossRef Ezzati M, Obermeyer Z, Tzoulaki I, Mayosi BM, Elliott P, Leon DA. Contributions of risk factors and medical care to cardiovascular mortality trends. Nat Rev Cardiol. 2015;12:508–30.PubMedPubMedCentralCrossRef
4.
go back to reference Cholesterol Treatment Trialists’ (CTT) Collaboration. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170 000 participants in 26 randomised trials. Lancet. 2010;376:1670–81.CrossRef Cholesterol Treatment Trialists’ (CTT) Collaboration. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170 000 participants in 26 randomised trials. Lancet. 2010;376:1670–81.CrossRef
5.
go back to reference Murchie P, Campbell NC, Ritchie LD, Simpson JA, Thain J. Secondary prevention clinics for coronary heart disease: four year follow up of a randomised controlled trial in primary care. BMJ. 2003;326:84.PubMedPubMedCentralCrossRef Murchie P, Campbell NC, Ritchie LD, Simpson JA, Thain J. Secondary prevention clinics for coronary heart disease: four year follow up of a randomised controlled trial in primary care. BMJ. 2003;326:84.PubMedPubMedCentralCrossRef
6.
go back to reference Schwartz GG, Steg PG, Szarek M, et al. Alirocumab and cardiovascular outcomes after acute coronary syndrome. N Engl J Med. 2018;379:2097–107.PubMedCrossRef Schwartz GG, Steg PG, Szarek M, et al. Alirocumab and cardiovascular outcomes after acute coronary syndrome. N Engl J Med. 2018;379:2097–107.PubMedCrossRef
8.
go back to reference Sabatine MS, Giugliano RP, Keech AC, et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017;376:1713–22.PubMedCrossRef Sabatine MS, Giugliano RP, Keech AC, et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017;376:1713–22.PubMedCrossRef
9.
go back to reference Lawler PR, Bhatt DL, Godoy LC, et al. Targeting cardiovascular inflammation: next steps in clinical translation. Eur Heart J. 2021;42:113–31.PubMedCrossRef Lawler PR, Bhatt DL, Godoy LC, et al. Targeting cardiovascular inflammation: next steps in clinical translation. Eur Heart J. 2021;42:113–31.PubMedCrossRef
10.
go back to reference Emerging Risk Factors Collaboration. C‑reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis. Lancet. 2010;375:132–40.CrossRef Emerging Risk Factors Collaboration. C‑reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis. Lancet. 2010;375:132–40.CrossRef
11.
go back to reference Ridker PM, Cannon CP, Morrow D, et al. C‑reactive protein levels and outcomes after statin therapy. N Engl J Med. 2005;352:20–8.PubMedCrossRef Ridker PM, Cannon CP, Morrow D, et al. C‑reactive protein levels and outcomes after statin therapy. N Engl J Med. 2005;352:20–8.PubMedCrossRef
12.
go back to reference Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015;372:2387–97.PubMedCrossRef Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015;372:2387–97.PubMedCrossRef
13.
go back to reference Ridker PM, Danielson E, Fonseca FAH, et al. Reduction in C‑reactive protein and LDL cholesterol and cardiovascular event rates after initiation of rosuvastatin: a prospective study of the JUPITER trial. Lancet. 2009;373:1175–82.PubMedCrossRef Ridker PM, Danielson E, Fonseca FAH, et al. Reduction in C‑reactive protein and LDL cholesterol and cardiovascular event rates after initiation of rosuvastatin: a prospective study of the JUPITER trial. Lancet. 2009;373:1175–82.PubMedCrossRef
14.
go back to reference C Reactive Protein Coronary Heart Disease Genetics Collaboration (CCGC). Association between C reactive protein and coronary heart disease: mendelian randomisation analysis based on individual participant data. BMJ. 2011;342:d548.CrossRef C Reactive Protein Coronary Heart Disease Genetics Collaboration (CCGC). Association between C reactive protein and coronary heart disease: mendelian randomisation analysis based on individual participant data. BMJ. 2011;342:d548.CrossRef
15.
go back to reference 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.CrossRef 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.CrossRef
16.
go back to reference Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377:1119–31.PubMedCrossRef Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377:1119–31.PubMedCrossRef
17.
go back to reference Bally M, Dendukuri N, Rich B, et al. Risk of acute myocardial infarction with NSAIDs in real world use: bayesian meta-analysis of individual patient data. BMJ. 2017;357:j1909.PubMedPubMedCentralCrossRef Bally M, Dendukuri N, Rich B, et al. Risk of acute myocardial infarction with NSAIDs in real world use: bayesian meta-analysis of individual patient data. BMJ. 2017;357:j1909.PubMedPubMedCentralCrossRef
18.
go back to reference Patrono C, García Rodríguez LA, Landolfi R, Baigent C. Low-dose aspirin for the prevention of atherothrombosis. N Engl J Med. 2005;353:2373–83.PubMedCrossRef Patrono C, García Rodríguez LA, Landolfi R, Baigent C. Low-dose aspirin for the prevention of atherothrombosis. N Engl J Med. 2005;353:2373–83.PubMedCrossRef
19.
go back to reference Mitchell JA, Warner TD. COX isoforms in the cardiovascular system: understanding the activities of non-steroidal anti-inflammatory drugs. Nat Rev Drug Discov. 2006;5:75–86.PubMedCrossRef Mitchell JA, Warner TD. COX isoforms in the cardiovascular system: understanding the activities of non-steroidal anti-inflammatory drugs. Nat Rev Drug Discov. 2006;5:75–86.PubMedCrossRef
20.
go back to reference Wei L, MacDonald TM, Walker BR. Taking glucocorticoids by prescription is associated with subsequent cardiovascular disease. Ann Intern Med. 2004;141:764–70.PubMedCrossRef Wei L, MacDonald TM, Walker BR. Taking glucocorticoids by prescription is associated with subsequent cardiovascular disease. Ann Intern Med. 2004;141:764–70.PubMedCrossRef
21.
go back to reference White HD, Held C, Stewart R, et al. Darapladib for preventing ischemic events in stable coronary heart disease. N Engl J Med. 2014;370:1702–11.PubMedCrossRef White HD, Held C, Stewart R, et al. Darapladib for preventing ischemic events in stable coronary heart disease. N Engl J Med. 2014;370:1702–11.PubMedCrossRef
22.
go back to reference O’Donoghue ML, Braunwald E, White HD, et al. Effect of darapladib on major coronary events after an acute coronary syndrome: the SOLID-TIMI 52 randomized clinical trial. JAMA. 2014;312:1006–15.PubMedCrossRef O’Donoghue ML, Braunwald E, White HD, et al. Effect of darapladib on major coronary events after an acute coronary syndrome: the SOLID-TIMI 52 randomized clinical trial. JAMA. 2014;312:1006–15.PubMedCrossRef
23.
go back to reference Nicholls SJ, Kastelein JJP, Schwartz GG, et al. Varespladib and cardiovascular events in patients with an acute coronary syndrome: the VISTA-16 randomized clinical trial. JAMA. 2014;311:252–62.PubMedCrossRef Nicholls SJ, Kastelein JJP, Schwartz GG, et al. Varespladib and cardiovascular events in patients with an acute coronary syndrome: the VISTA-16 randomized clinical trial. JAMA. 2014;311:252–62.PubMedCrossRef
24.
go back to reference O’Donoghue ML, Glaser R, Cavender MA, et al. Effect of losmapimod on cardiovascular outcomes in patients hospitalized with acute myocardial infarction: a randomized clinical trial. JAMA. 2016;315:1591–9.PubMedCrossRef O’Donoghue ML, Glaser R, Cavender MA, et al. Effect of losmapimod on cardiovascular outcomes in patients hospitalized with acute myocardial infarction: a randomized clinical trial. JAMA. 2016;315:1591–9.PubMedCrossRef
25.
go back to reference Ridker PM, Everett BM, Pradhan A, et al. Low-dose methotrexate for the prevention of atherosclerotic events. N Engl J Med. 2018;380:752–62.PubMedPubMedCentralCrossRef Ridker PM, Everett BM, Pradhan A, et al. Low-dose methotrexate for the prevention of atherosclerotic events. N Engl J Med. 2018;380:752–62.PubMedPubMedCentralCrossRef
26.
go back to reference Tardif J‑C, Kouz S, Waters DD, et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N Engl J Med. 2019;381:2497–505.PubMedCrossRef Tardif J‑C, Kouz S, Waters DD, et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N Engl J Med. 2019;381:2497–505.PubMedCrossRef
27.
go back to reference Nidorf SM, Fiolet ATL, Mosterd A, et al. Colchicine in patients with chronic coronary disease. N Engl J Med. 2020;383:1838–47.PubMedCrossRef Nidorf SM, Fiolet ATL, Mosterd A, et al. Colchicine in patients with chronic coronary disease. N Engl J Med. 2020;383:1838–47.PubMedCrossRef
28.
29.
go back to reference Imazio M, Bobbio M, Cecchi E, et al. Colchicine in addition to conventional therapy for acute pericarditis: results of the COlchicine for acute PEricarditis (COPE) trial. Circulation. 2005;112:2012–6.PubMedCrossRef Imazio M, Bobbio M, Cecchi E, et al. Colchicine in addition to conventional therapy for acute pericarditis: results of the COlchicine for acute PEricarditis (COPE) trial. Circulation. 2005;112:2012–6.PubMedCrossRef
30.
go back to reference Imazio M, Brucato A, Cemin R, et al. A randomized trial of colchicine for acute pericarditis. N Engl J Med. 2013;369:1522–8.PubMedCrossRef Imazio M, Brucato A, Cemin R, et al. A randomized trial of colchicine for acute pericarditis. N Engl J Med. 2013;369:1522–8.PubMedCrossRef
31.
go back to reference Fiolet ATL, Silvis MJM, Opstal TSJ, et al. Short-term effect of low-dose colchicine on inflammatory biomarkers, lipids, blood count and renal function in chronic coronary artery disease and elevated high-sensitivity C‑reactive protein. PLoS ONE. 2020;15:e237665.PubMedPubMedCentralCrossRef Fiolet ATL, Silvis MJM, Opstal TSJ, et al. Short-term effect of low-dose colchicine on inflammatory biomarkers, lipids, blood count and renal function in chronic coronary artery disease and elevated high-sensitivity C‑reactive protein. PLoS ONE. 2020;15:e237665.PubMedPubMedCentralCrossRef
32.
go back to reference Opstal TSJ, Hoogeveen RM, Fiolet ATL, et al. Colchicine attenuates inflammation beyond the inflammasome in chronic coronary artery disease: a LoDoCo2 proteomic substudy. Circulation. 2020;142:1996–8.PubMedCrossRef Opstal TSJ, Hoogeveen RM, Fiolet ATL, et al. Colchicine attenuates inflammation beyond the inflammasome in chronic coronary artery disease: a LoDoCo2 proteomic substudy. Circulation. 2020;142:1996–8.PubMedCrossRef
33.
34.
go back to reference Ben-Chetrit E, Levy M. Does the lack of the P‑glycoprotein efflux pump in neutrophils explain the efficacy of colchicine in familial Mediterranean fever and other inflammatory diseases? Med Hypotheses. 1998;51:377–80.PubMedCrossRef Ben-Chetrit E, Levy M. Does the lack of the P‑glycoprotein efflux pump in neutrophils explain the efficacy of colchicine in familial Mediterranean fever and other inflammatory diseases? Med Hypotheses. 1998;51:377–80.PubMedCrossRef
35.
go back to reference Paschke S, Weidner AF, Paust T, Marti O, Beil M, Ben-Chetrit E. Technical advance: inhibition of neutrophil chemotaxis by colchicine is modulated through viscoelastic properties of subcellular compartments. J Leukoc Biol. 2013;94:1091–6.PubMedCrossRef Paschke S, Weidner AF, Paust T, Marti O, Beil M, Ben-Chetrit E. Technical advance: inhibition of neutrophil chemotaxis by colchicine is modulated through viscoelastic properties of subcellular compartments. J Leukoc Biol. 2013;94:1091–6.PubMedCrossRef
36.
go back to reference Cronstein BN, Molad Y, Reibman J, Balakhane E, Levin RI, Weissmann G. Colchicine alters the quantitative and qualitative display of selectins on endothelial cells and neutrophils. J Clin Invest. 1995;96:994–1002.PubMedPubMedCentralCrossRef Cronstein BN, Molad Y, Reibman J, Balakhane E, Levin RI, Weissmann G. Colchicine alters the quantitative and qualitative display of selectins on endothelial cells and neutrophils. J Clin Invest. 1995;96:994–1002.PubMedPubMedCentralCrossRef
37.
go back to reference Vaidya K, Arnott C, Martínez GJ, et al. Colchicine therapy and plaque stabilization in patients with acute coronary syndrome: a CT coronary angiography study. JACC Cardiovasc Imaging. 2018;11:305–16.PubMedCrossRef Vaidya K, Arnott C, Martínez GJ, et al. Colchicine therapy and plaque stabilization in patients with acute coronary syndrome: a CT coronary angiography study. JACC Cardiovasc Imaging. 2018;11:305–16.PubMedCrossRef
38.
go back to reference Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–41.PubMedCrossRef Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–41.PubMedCrossRef
40.
go back to reference Duewell P, Kono H, Rayner KJ, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464:1357–61.PubMedPubMedCentralCrossRef Duewell P, Kono H, Rayner KJ, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464:1357–61.PubMedPubMedCentralCrossRef
41.
go back to reference White JG. Effects of colchicine and vinca alkaloids on human platelets. 3. Influence on primary internal contraction and secondary aggregation. Am J Pathol. 1969;54:467–78.PubMedPubMedCentral White JG. Effects of colchicine and vinca alkaloids on human platelets. 3. Influence on primary internal contraction and secondary aggregation. Am J Pathol. 1969;54:467–78.PubMedPubMedCentral
42.
go back to reference Cirillo P, Taglialatela V, Pellegrino G, et al. Effects of colchicine on platelet aggregation in patients on dual antiplatelet therapy with aspirin and clopidogrel. J Thromb Thrombolysis. 2020;50:468–72.PubMedPubMedCentralCrossRef Cirillo P, Taglialatela V, Pellegrino G, et al. Effects of colchicine on platelet aggregation in patients on dual antiplatelet therapy with aspirin and clopidogrel. J Thromb Thrombolysis. 2020;50:468–72.PubMedPubMedCentralCrossRef
43.
go back to reference Shah B, Allen N, Harchandani B, et al. Effect of colchicine on platelet-platelet and platelet-leukocyte interactions: a pilot study in healthy subjects. Inflammation. 2016;39:182–9.PubMedCrossRef Shah B, Allen N, Harchandani B, et al. Effect of colchicine on platelet-platelet and platelet-leukocyte interactions: a pilot study in healthy subjects. Inflammation. 2016;39:182–9.PubMedCrossRef
44.
go back to reference Fiolet ATL, Opstal TSJ, Mosterd A, et al. Efficacy and safety of low-dose colchicine in patients with coronary disease: a systematic review and meta-analysis of randomized trials. Eur Heart J. 2021;45:2765–75.CrossRef Fiolet ATL, Opstal TSJ, Mosterd A, et al. Efficacy and safety of low-dose colchicine in patients with coronary disease: a systematic review and meta-analysis of randomized trials. Eur Heart J. 2021;45:2765–75.CrossRef
45.
go back to reference Baigent C, Blackwell L, Collins R, et al. Aspirin in the primary and secondary prevention of vascular disease: collaborative meta-analysis of individual participant data from randomised trials. Lancet. 2009;373:1849–60.PubMedCrossRef Baigent C, Blackwell L, Collins R, et al. Aspirin in the primary and secondary prevention of vascular disease: collaborative meta-analysis of individual participant data from randomised trials. Lancet. 2009;373:1849–60.PubMedCrossRef
47.
go back to reference Ridker PM, Paynter NP, Rifai N, Gaziano JM, Cook NR. C‑reactive protein and parental history improve global cardiovascular risk prediction: the Reynolds risk score for men. Circulation. 2008;118:2243–51.PubMedPubMedCentralCrossRef Ridker PM, Paynter NP, Rifai N, Gaziano JM, Cook NR. C‑reactive protein and parental history improve global cardiovascular risk prediction: the Reynolds risk score for men. Circulation. 2008;118:2243–51.PubMedPubMedCentralCrossRef
48.
go back to reference van’t Klooster CC, Bhatt DL, Steg PG, et al. Predicting 10-year risk of recurrent cardiovascular events and cardiovascular interventions in patients with established cardiovascular disease: results from UCC-SMART and REACH. Int J Cardiol. 2021;325:140–8.CrossRef van’t Klooster CC, Bhatt DL, Steg PG, et al. Predicting 10-year risk of recurrent cardiovascular events and cardiovascular interventions in patients with established cardiovascular disease: results from UCC-SMART and REACH. Int J Cardiol. 2021;325:140–8.CrossRef
49.
go back to reference Pradhan AD. Time to commence or time out for colchicine in secondary prevention of cardiovascular disease? Eur Heart J. 2021;42:2776–9.PubMedCrossRef Pradhan AD. Time to commence or time out for colchicine in secondary prevention of cardiovascular disease? Eur Heart J. 2021;42:2776–9.PubMedCrossRef
50.
go back to reference Nordanstig J, James S, Andersson M, et al. Mortality with paclitaxel-coated devices in peripheral artery disease. N Engl J Med. 2020;383:2538–46.PubMedCrossRef Nordanstig J, James S, Andersson M, et al. Mortality with paclitaxel-coated devices in peripheral artery disease. N Engl J Med. 2020;383:2538–46.PubMedCrossRef
51.
go back to reference Ridker PM, Devalaraja M, Baeres FMM, 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;397:2060–9.PubMedCrossRef Ridker PM, Devalaraja M, Baeres FMM, 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;397:2060–9.PubMedCrossRef
Metagegevens
Titel
Targeting residual inflammatory risk in coronary disease: to catch a monkey by its tail
Auteurs
A. T. L. Fiolet
T. S. J. Opstal
M. J. M. Silvis
J. H. Cornel
A. Mosterd
Publicatiedatum
17-08-2021
Uitgeverij
Bohn Stafleu van Loghum
Gepubliceerd in
Netherlands Heart Journal / Uitgave 1/2022
Print ISSN: 1568-5888
Elektronisch ISSN: 1876-6250
DOI
https://doi.org/10.1007/s12471-021-01605-3

Andere artikelen Uitgave 1/2022

Netherlands Heart Journal 1/2022 Naar de uitgave