Arterial Effects of Canakinumab in Patients With Atherosclerosis and Type 2 Diabetes or Glucose Intolerance

Robin P Choudhury, Jacqueline S Birks, Venkatesh Mani, Luca Biasiolli, Matthew D Robson, Philippe L L'Allier, Marc-Alexandre Gingras, Nadia Alie, Mary Ann McLaughlin, Craig T Basson, Alison D Schecter, Eric C Svensson, Yiming Zhang, Denise Yates, Jean-Claude Tardif, Zahi A Fayad, Robin P Choudhury, Jacqueline S Birks, Venkatesh Mani, Luca Biasiolli, Matthew D Robson, Philippe L L'Allier, Marc-Alexandre Gingras, Nadia Alie, Mary Ann McLaughlin, Craig T Basson, Alison D Schecter, Eric C Svensson, Yiming Zhang, Denise Yates, Jean-Claude Tardif, Zahi A Fayad

Abstract

Background: Evidence suggests that interleukin (IL)-1β is important in the pathogenesis of atherosclerosis and its complications and that inhibiting IL-1β may favorably affect vascular disease progression.

Objectives: The goal of this study was to evaluate the effects of IL-1β inhibition with canakinumab versus placebo on arterial structure and function, determined by magnetic resonance imaging.

Methods: Patients (N = 189) with atherosclerotic disease and either type 2 diabetes mellitus or impaired glucose tolerance were randomized to receive placebo (n = 94) or canakinumab 150 mg monthly (n = 95) for 12 months. They underwent magnetic resonance imaging of the carotid arteries and aorta.

Results: There were no statistically significant differences between canakinumab compared with placebo in the primary efficacy and safety endpoints. There was no statistically significant change in mean carotid wall area and no effect on aortic distensibility, measured at 3 separate anatomic sites. The change in mean carotid artery wall area was -3.37 mm2 after 12 months with canakinumab versus placebo. High-sensitivity C-reactive protein was significantly reduced by canakinumab compared with placebo at 3 months (geometric mean ratio [GMR]: 0.568; 95% confidence interval [CI]: 0.436 to 0.740; p < 0.0001) and 12 months (GMR: 0.56; 95% CI: 0.414 to 0.758; p = 0.0002). Lipoprotein(a) levels were reduced by canakinumab compared with placebo (-4.30 mg/dl [range: -8.5 to -0.55 mg/dl]; p = 0.025] at 12 months), but triglyceride levels increased (GMR: 1.20; 95% CI: 1.046 to 1.380; p = 0.01). In these patients with type 2 diabetes mellitus or impaired glucose tolerance, canakinumab had no effect compared with placebo on any of the measures assessed by using a standard oral glucose tolerance test.

Conclusions: There were no statistically significant effects of canakinumab on measures of vascular structure or function. Canakinumab reduced markers of inflammation (high-sensitivity C-reactive protein and interleukin-6), and there were modest increases in levels of total cholesterol and triglycerides. (Safety & Effectiveness on Vascular Structure and Function of ACZ885 in Atherosclerosis and Either T2DM or IGT Patients; NCT00995930).

Keywords: C-reactive protein; homeostasis model assessment; inflammation; interleukin-1.

Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
Changes in Carotid Wall Area We evaluated 12-month change from baseline in wall area as an indicator of atherosclerosis progression. (A) In both left and right carotid arteries, canakinumab retarded progression of wall area compared with placebo. Consistent in magnitude and direction, the changes did not reach statistical significance in the mean carotid wall area (pre-stated endpoint) or in either carotid artery analyzed separately. Bars = means with 95% confidence intervals. (B) Within the same patient, there was a striking concordance of change in wall area between left- and right-sided arteries, which was maintained in both treatment groups (p < 0.0001 for each). Upper right quadrant = patients with progression in both carotid arteries.
Figure 2
Figure 2
Changes in Lipids and C-Reactive Protein Absolute level or change from baseline at 3 and 12 months are shown for (A) C-reactive protein; (B) lipoprotein(a) (Lp[a]); (C) total cholesterol; (D) high-density lipoprotein (HDL) cholesterol; and (E) triglycerides.
Central Illustration
Central Illustration
Effects of IL-1β Inhibition Interleukin (IL)-1β seems important in the pathogenesis of atherosclerosis. In this placebo-controlled trial in patients with evidence of clinical atherosclerosis and either type 2 diabetes mellitus or impaired glucose tolerance, the IL-1β inhibitor canakinumab reduced measures of inflammation but did not significantly affect measures of vascular structure or function. LDL = low-density lipoprotein.

References

    1. Libby P. Inflammation in atherosclerosis. Nature. 2002;420:868–874.
    1. Libby P., Ridker P.M., Maseri A. Inflammation and atherosclerosis. Circulation. 2002;105:1135–1143.
    1. Ross R. Atherosclerosis—an inflammatory disease. N Engl J Med. 1999;340:115–126.
    1. Ridker P.M., Hennekens C.H., Buring J.E., Rifai N. C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. N Engl J Med. 2000;342:836–843.
    1. Ridker P.M., Rifai N., Pfeffer M.A. Inflammation, pravastatin, and the risk of coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and Recurrent Events (CARE) Investigators. Circulation. 1998;98:839–844.
    1. Ridker P.M., Cushman M., Stampfer M.J. Inflammation, aspirin, and the risk of cardiovascular disease in apparently healthy men. N Engl J Med. 1997;336:973–979.
    1. Ridker P.M., Rifai N., Pfeffer M.A. Long-term effects of pravastatin on plasma concentration of C-reactive protein. The Cholesterol and Recurrent Events (CARE) Investigators. Circulation. 1999;100:230–235.
    1. Tipping P.G., Hancock W.W. Production of tumor necrosis factor and interleukin-1 by macrophages from human atheromatous plaques. Am J Pathol. 1993;142:1721–1728.
    1. Duewell P., Kono H., Rayner K.J. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464:1357–1361.
    1. Kirii H., Niwa T., Yamada Y. Lack of interleukin-1beta decreases the severity of atherosclerosis in apoE-deficient mice. Arterioscler Thromb Vasc Biol. 2003;23:656–660.
    1. Elhage R., Maret A., Pieraggi M.T. Differential effects of interleukin-1 receptor antagonist and tumor necrosis factor binding protein on fatty-streak formation in apolipoprotein E-deficient mice. Circulation. 1998;97:242–244.
    1. Isoda K., Sawada S., Ishigami N. Lack of interleukin-1 receptor antagonist modulates plaque composition in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2004;24:1068–1073.
    1. Merhi-Soussi F., Kwak B.R., Magne D. Interleukin-1 plays a major role in vascular inflammation and atherosclerosis in male apolipoprotein E-knockout mice. Cardiovasc Res. 2005;66:583–593.
    1. Ikonomidis I., Lekakis J.P., Nikolaou M. Inhibition of interleukin-1 by anakinra improves vascular and left ventricular function in patients with rheumatoid arthritis. Circulation. 2008;117:2662–2669.
    1. Maedler K., Sergeev P., Ris F. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest. 2002;110:851–860.
    1. Larsen C.M., Faulenbach M., Vaag A. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356:1517–1526.
    1. Stratton I.M., Adler A.I., Neil H.A. Association of glycaemia with macrovascular and microvascular complications of type 2 diabetes (UKPDS 35): prospective observational study. BMJ. 2000;321:405–412.
    1. Moreno P.R., Murcia A.M., Palacios I.F. Coronary composition and macrophage infiltration in atherectomy specimens from patients with diabetes mellitus. Circulation. 2000;102:2180–2184.
    1. Lee J.M., Shirodaria C., Jackson C.E. Multi-modal magnetic resonance imaging quantifies atherosclerosis and vascular dysfunction in patients with type 2 diabetes mellitus. Diab Vasc Dis Res. 2007;4:44–48.
    1. Ridker P.M., Howard C.P., Walter V. Effects of interleukin-1beta inhibition with canakinumab on hemoglobin A1c, lipids, C-reactive protein, interleukin-6, and fibrinogen: a phase IIb randomized, placebo-controlled trial. Circulation. 2012;126:2739–2748.
    1. Ridker P.M., Thuren T., Zalewski A., Libby P. Interleukin-1beta 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.
    1. Duivenvoorden R., de Groot E., Elsen B.M. In vivo quantification of carotid artery wall dimensions: 3.0-Tesla MRI versus B-mode ultrasound imaging. Circ Cardiovasc Imaging. 2009;2:235–242.
    1. Fayad Z.A., Mani V., Woodward M. Safety and efficacy of dalcetrapib on atherosclerotic disease using novel non-invasive multimodality imaging (dal-PLAQUE): a randomised clinical trial. Lancet. 2011;378:1547–1559.
    1. Lee J.M., Robson M.D., Yu L.M. Effects of high-dose modified-release nicotinic acid on atherosclerosis and vascular function: a randomized, placebo-controlled, magnetic resonance imaging study. J Am Coll Cardiol. 2009;54:1787–1794.
    1. Wiesmann F., Petersen S.E., Leeson P.M. Global impairment of brachial, carotid, and aortic vascular function in young smokers: direct quantification by high-resolution magnetic resonance imaging. J Am Coll Cardiol. 2004;44:2056–2064.
    1. Cruickshank K., Riste L., Anderson S.G. Aortic pulse-wave velocity and its relationship to mortality in diabetes and glucose intolerance: an integrated index of vascular function? Circulation. 2002;106:2085–2090.
    1. Biasiolli L., Lindsay A.C., Chai J.T. In-vivo quantitative T2 mapping of carotid arteries in atherosclerotic patients: segmentation and T2 measurement of plaque components. J Cardiovasc Magn Reson. 2013;15:69.
    1. Elkhawad M., Rudd J.H., Sarov-Blat L. Effects of p38 mitogen-activated protein kinase inhibition on vascular and systemic inflammation in patients with atherosclerosis. J Am Coll Cardiol Img. 2012;5:911–922.
    1. Tawakol A., Migrino R.Q., Bashian G.G. In vivo 18F-fluorodeoxyglucose positron emission tomography imaging provides a noninvasive measure of carotid plaque inflammation in patients. J Am Coll Cardiol. 2006;48:1818–1824.
    1. El Kasmi K.C., Stenmark K.R. Contribution of metabolic reprogramming to macrophage plasticity and function. Semin Immunol. 2015;27:267–275.
    1. Sica A., Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122:787–795.
    1. Dutta P., Courties G., Wei Y. Myocardial infarction accelerates atherosclerosis. Nature. 2012;487:325–329.
    1. Sager H.B., Heidt T., Hulsmans M. Targeting interleukin-1beta reduces leukocyte production after acute myocardial infarction. Circulation. 2015;132:1880–1890.
    1. Alexander M.R., Moehle C.W., Johnson J.L. Genetic inactivation of IL-1 signaling enhances atherosclerotic plaque instability and reduces outward vessel remodeling in advanced atherosclerosis in mice. J Clin Invest. 2012;122:70–79.
    1. Saiki O., Takao R., Naruse Y. Infliximab but not methotrexate induces extra-high levels of VLDL-triglyceride in patients with rheumatoid arthritis. J Rheumatol. 2007;34:1997–2004.
    1. Lindsay A.C., Choudhury R.P. Form to function: current and future roles for atherosclerosis imaging in drug development. Nat Rev Drug Discov. 2008;7:517–529.

Source: PubMed

3
Subscribe