Failure to Launch: Targeting Inflammation in Acute Coronary Syndromes

Jennifer A Rymer, L Kristin Newby, Jennifer A Rymer, L Kristin Newby

Abstract

The importance of inflammation and inflammatory pathways in atherosclerotic disease and acute coronary syndromes (ACS) is well established. The success of statin therapy rests not only on potently reducing levels of low-density lipoprotein cholesterol, but also on the many beneficial, pleiotropic effects statin therapy has on various inflammatory mechanisms in atherosclerotic disease, from reducing endothelial dysfunction to attenuating levels of serum C-reactive protein. Due to the growing awareness of the importance of inflammation in ACS, investigators have attempted to develop novel therapies against known markers of inflammation for several decades. Targeted pathways have ranged from inhibiting C5 cleavage with a high-affinity monoclonal antibody against C5 to inhibiting the activation of the p38 mitogen-activated protein kinase signaling cascades. In each of these instances, despite promising early preclinical and mechanistic studies and phase 2 trials suggesting a potential benefit in reducing post-MI complications or restenosis, these novel therapies have failed to show benefits during large, phase 3 clinical outcomes trials. This review discusses several examples of novel anti-inflammatory therapies that failed to show significant improvement on clinical outcomes when tested in large, randomized trials and highlights potential explanations for why targeted therapies against known markers of inflammation in ACS have failed to launch.

Keywords: ACS, acute coronary syndromes; CABG, coronary artery bypass graft; CAD, coronary artery disease; HDL-C, high-density lipoprotein cholesterol; IL, interleukin; LDL-C, low-density lipoprotein cholesterol; Lp-PLA2, lipoprotein-associated phospholipase A2; MAPK, mitogen-activated protein kinase; MI, myocardial infarction; NSTEMI, non–ST-segment myocardial infarction; PCI, percutaneous coronary intervention; PSGL, P-selectin glycoprotein ligand; STEMI, ST-segment elevation myocardial infarction; SVG, saphenous vein grafts; TBR, tissue-to-background ratio; acute coronary syndrome; anti-inflammatory; drug targets; hsCRP, high-sensitivity C-reactive protein; sPLA2, secretory phospholipase A2.

Figures

Graphical abstract
Graphical abstract
Central Illustration
Central Illustration
Mechanisms of Anti-Inflammatory Agents in ACS ACS = acute coronary syndromes; IL = interleukin; Lp-PLA2 = lipoprotein-associated phospholipase A2; MAPK = mitogen-activated protein kinase; PSGL-1 = P-selectin glycoprotein ligand-1; sPLA2 = secretory phospholipase A2; TNF = tumor necrosis factor.
Figure 1
Figure 1
6-Month Kaplan-Meier Curves for Mortality by Randomized Treatment Assignment (COMMA, Phase 2) COMMA = COMplement inhibition in Myocardial infarction treated with Angioplasty.
Figure 2
Figure 2
30-Day Mortality and Composite of Death, Shock, or CHF by Treatment With Pexelizumab Versus Placebo (APEX-AMI, Phase 3) (A) 30-day mortality; (B) composite of death, shock, or CHF. APEX AMI = Assessment of Pexelizumab in Acute Myocardial Infarction; CHF = congestive heart failure; CI = confidence interval; HR = hazard ratio.
Figure 3
Figure 3
Kaplan-Meier Survival Curves for the Primary Composite Outcome in Patients Treated With Placebo Versus Varespladib (VISTA-16, Phase 3) VISTA-16 = Vascular Inflammation Suppression to Treat Acute Coronary Syndrome for 16 weeks; other abbreviations as in Figure 2.
Figure 4
Figure 4
Cumulative Incidence Curves for CHD Death, MI, or Urgent Coronary Revascularization for Myocardial Ischemia for Treatment With Darapladib Versus Placebo (SOLID-TIMI 52, Phase 3) CHD = coronary heart disease; MI = myocardial infarction; SOLID-TIMI 52 = Stabilization of pLaques usIng Darapladib-Thrombolysis In Myocardial Infarction 52; other abbreviations as in Figure 2.
Figure 5
Figure 5
Cumulative Incidence Curves for Cardiovascular Death, MI, or Severe Recurrent Ischemia Leading to Urgent Revascularization Through 12 Weeks With Losmapimod Versus Placebo (LATITUDE-TIMI 60, Phase 3) CI = confidence interval; LATITUDE-TIMI 60 = Losmapimod to Inhibit p38 MAP Kinase as a Therapeutic Target and Modify Outcomes After an Acute Coronary Syndrome-Thrombolysis In Myocardial Infarction 60; MI = myocardial infarction.
Figure 6
Figure 6
Potential Reasons for Surrogate Endpoint Failure (A) The surrogate endpoint is not in the causal pathway of the disease state. (B) There are several causal pathways of the disease state, but the intervention only impacts the pathway mediated by the surrogate endpoint. (C) There are several causal pathways of the disease state, but the pathway mediated by the surrogate endpoint is not impacted by the intervention. (D) The intervention acts on a pathway separate from the disease process.

References

    1. Go A.S., Mozaffarian D., Roger V.L. Heart disease and stroke statistics—2014 update: a report from the American Heart Association. Circulation. 2014;129:e28–e292.
    1. Zhao Z., Winget M. Economic burden of illness of acute coronary syndromes: medical and productivity costs. BMC Health Services Research. 2011;11:2–9.
    1. Ghushchyan V., Nair K.V., Page R.L. Indirect and direct costs of acute coronary syndromes with comorbid atrial fibrillation, heart failure, or both. Vasc Health Risk Manag. 2015;11:25–34.
    1. Yeh R.W., Sidney S., Chandra M., Sorel M., Selby J.V., Go A.S. Population trends in the incidence and outcomes of acute myocardial infarction. N Engl J Med. 2010;362:2155–2165.
    1. Tobbia P., Brodie B.R., Witzenbichler B. Adverse event rates following primary PCI for STEMI at US and non-US hospitals: three-year analysis from the HORIZONS-AMI trial. EuroIntervention. 2013;8:1134–1142.
    1. Brieger D., Fox K.A., Fitzgerald G. Predicting freedom from clinical events in non-ST-elevation acute coronary syndromes: the Global Registry of Acute Coronary Events. Heart. 2009;95:888.
    1. Suleiman M., Khatib R., Agmon Y. Early inflammation and risk of long-term development of heart failure and mortality in survivors of acute myocardial infarction predictive role of C-reactive protein. J Am Coll Cardiol. 2006;47:962–968.
    1. Kragholm K., Newby L.K., Melloni C. Emerging treatment options to improve cardiovascular outcomes in patients with acute coronary syndrome: focus on losmapimod. Drug Des Devel Ther. 2015;9:4279–4286.
    1. Thomas T.C., Rollins S.A., Rother R.P. Inhibition of complement activity by humanized anti-C5 antibody and single-chain Fv. Mol Immunol. 1996;33:1389–1401.
    1. Pinckard R.N., Olson M.S., Giclas P.C., Terry R., Boyer J.T., O'Rourke R.A. Consumption of classical complement components by heart subcellular membranes in vitro and in patients after acute myocardial infarction. J Clin Invest. 1975;56:740–750.
    1. Mathey D., Schofer J., Schafer H.J. Early accumulation of the terminal complement-complex in the ischaemic myocardium after reperfusion. Eur Heart J. 1994;15:418–423.
    1. Vakeva A.P., Agah A., Rollins S.A. Myocardial infarction and apoptosis after myocardial ischemia and reperfusion: role of the terminal complement components and inhibition by anti-C5 therapy. Circulation. 1998;97:2259–2267.
    1. Tang K., Cheng Y., Wu S., Liu L., Cheng L. Protective effect of C5 shRNA on myocardial ischemia-reperfusion injury in rats. Can J Physiol Pharmacol. 2012;90:1394–1402.
    1. Amsterdam E.A., Stahl G.L., Pan H.L., Rendig S.V., Fletcher M.P., Longhurst J.C. Limitation of reperfusion injury by a monoclonal antibody to c5a during myocardial infarction in pigs. Am J Physiol. 1995;268:H448–H457.
    1. Mahaffey K.W., Granger C.B., Nicolau J.C. Effect of pexelizumab, an anti-C5 complement antibody, as adjunctive therapy to fibrinolysis in acute myocardial infarction: The COMPlement inhibition in myocardial infarction treated with thromboLYtics (COMPLY) trial. Circulation. 2003;108:1176–1183.
    1. Granger C.B., Mahaffey K.W., Weaver W.D. Effect of pexelizumab, an anti-C5 complement antibody, as adjunctive therapy to primary percutaneous intervention acute myocardial infarction: the COMplement inhibition in Myocardial infarction treated with Angioplasty (COMMA) trial. Circulation. 2003;108:1184–1190.
    1. Armstrong P.W., Mahaffey K.W., Chang W.C. Concerning the mechanism of pexelizumab’s benefit in acute myocardial infarction. Am Heart J. 2006;151:787–790.
    1. Stahl G.L., Amsterdam E.A., Symons J.D. Role of thromboxane A2 in the cardiovascular response to intracoronary C5a. Circ Res. 1990;66:1103–1111.
    1. Topham P.S., Haydar S.A., Kuphal R. Complement-mediated injury reversibly disrupts glomerular epithelial cell actin microfilaments and focal adhesions. Kidney Int. 1999;55:1763–1775.
    1. Armstrong P.W., Granger C.B., Adams P.X., for the APEX AMI Investigators Pexelizumab for acute ST-elevation myocardial infarction in patients undergoing primary percutaneous coronary intervention: a randomized controlled trial. JAMA. 2007;297:43–51.
    1. Fitch J.C., Rollins S., Matis L. Pharmacology and biological efficacy of a recombinant, humanized, single-chain antibody C5 complement inhibitor in patients undergoing coronary artery bypass graft surgery with cardiopulmonary bypass. Circulation. 1999;100:2499–2506.
    1. Royston D. Systemic inflammatory responses to surgery with CPB. Perfusion. 1996;11:177–189.
    1. Wan S., LeClerc J.-L., Vincent J.-L. Inflammatory response to CPB. Chest. 1997;112:676–692.
    1. Hall R.I., Smith M.S., Rocker G. The systemic inflammatory response to CPB. Anesth Analg. 1997;85:766–782.
    1. Verrier E.D., Shernan S.K., Taylor K.M. Terminal complement blockade with pexelizumab during coronary artery bypass graft surgery requiring cardiopulmonary bypass: a randomized trial. JAMA. 2004;291:2319–2327.
    1. Haverich A., Shernan S.K., Levy J.H. Inhibition of complement activation by pexelizumab reduced death and myocardial infarction in higher risk cardiac surgical patients. Ann Thorac Surg. 2006;82:486–492.
    1. Smith P.K., Shernan S.K., Chen J.C. Effects of C5 complement inhibitor pexelizumab on outcome in high-risk coronary artery bypass grafting: combined results from the PRIMO-CABG I and II trials. J Thorac Cardiovasc Surg. 2011;142:89–98.
    1. Testa L., Van Gaal W.J., Bhindi R. Pexelizumab in ischemic heart disease: a systematic review and meta-analysis on 15,196 patients. J Thorac Cardiovasc Surg. 2008;136:884–893.
    1. Nicholls S.J., Cavender M.A., Kastelein J.P. Inhibition of secretory phospholipase A(2) in patients with acute coronary syndromes: rationale and design of the Vascular Inflammation Suppression to Treat Acute Coronary Syndrome for 16 Weeks (VISTA-16) trial. Cardiovasc Drugs Ther. 2012;26:71–75.
    1. Karabina S.A., Brocheriou S., Naour G.L. Atherogenic properties of LDL particles modified by human group X secreted phospholipase A2 on human endothelial cell function. FASEB. 2006;20:2547–2549.
    1. Gora S., Maouche S., Atout R. Phospholipolyzed LDL induces an inflammatory response in endothelial cells through endoplasmic reticulum stress signaling. FASEB. 2010;24:3284–3297.
    1. Fuentes L., Hernandez M., Fernandez-Aviles F.J. Cooperation between secretory phospholipase A2 and TNF-receptor superfamily signaling: implications for the inflammatory response in atherogenesis. Circ Res. 2002;91:681–688.
    1. Liu P.Y., Li Y.H., Wei-Chuan T. Prognostic value and the changes of plasma levels of secretory type II phospholipase A2 in patients with coronary artery disease undergoing percutaneous coronary intervention. Eur Heart J. 2003;24:1824–1832.
    1. Kugiyama K., Ota Y., Sugiyama S. Prognostic value of plasma levels of secretory type II phospholipase A2 in patients with unstable angina pectoris. Am J Cardiol. 2000;86:718–722.
    1. Mallat Z., Steg G., Benessiano J. Circulating secretory phospholipase A2 activity predicts recurrent events in patients with severe acute coronary syndromes. J Am Coll Cardiol. 2005;46:1249–1257.
    1. Rosenson R.S., Hislop C., McConnell D. Effects of 1-H-indole-3-glyoxamide (A-002) on concentration of secretory phospholipase A2 (PLASMA study): a phase II double-blind, randomised, placebo-controlled trial. Lancet. 2009;373:649–658.
    1. Rosenson R.S., Hislop C., Elliott M. Effects of varespladib methyl on biomarkers and major cardiovascular events in acute coronary syndrome patients. J Am Coll Cardiol. 2010;56:1079–1088.
    1. Rosenson R.S., Elliott M., Stasiv Y. Randomized trial of an inhibitor of secretory phospholipase A2 on atherogenic lipoprotein subclasses in statin-treated patients with coronary heart disease. Eur Heart J. 2011;32:999–1005.
    1. Nicholls S.J., Kastelein J., Schwartz G.G. Varespladib and cardiovascular events in patients with an acute coronary syndrome: the VISTA-16 randomized clinical trial. JAMA. 2014;311:252–262.
    1. MacPhee C.H., Moores K.E., Boyd H.F. Lipoprotein-associated phospholipase A2, platelet-activating factor acetylhydrolase, generates two bioactive products during the oxidation of low-density lipoprotein: use of a novel inhibitor. Biochem J. 1999;338:479–487.
    1. Kolodgie F.D., Burke A.P., Skorija K.S. Lipoprotein-associated phospholipase A2 protein expression in the natural progression of human coronary atherosclerosis. Arterioscler Thromb Vasc Biol. 2006;26:2523–2529.
    1. Häkkinen T., Luoma J.S., Hiltunen M.O. Lipoprotein-associated phospholipase A(2), platelet-activating factor acetylhydrolase, is expressed by macrophages in human and rabbit atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 1999;19:2909–2917.
    1. Jang Y., Waterworth D., Lee J.-E. Carriage of the V279F null allele within the gene encoding Lp-PLA2 is protective from coronary artery disease in South Korean males. PloS One. 2011;6:e18208.
    1. The Lp-PLA2 Studies Collaboration Lipoprotein-associated phospholipase A(2) and risk of coronary disease, stroke, and mortality: collaborative analysis of 32 prospective studies. Lancet. 2010;375:1536–1544.
    1. Di Angelantonio E., Gao P., Pennells L. Lipid-related markers and cardiovascular disease prediction. JAMA. 2012;307:2499–2506.
    1. Serruys P.W., Garcia-Garcia H.M., Buszman P. Effects of the direct-lipoprotein associated phospholipase A2 inhibitor darapladib on human coronary atherosclerotic plaque. Circulation. 2008;118:1172–1182.
    1. Wilensky R.L., Shi Y., Mohler E.R. Inhibition of lipoprotein-associated Phospholipase A2 reduces complex coronary atherosclerotic plaque development. Nature Medicine. 2008;14:1059–1066.
    1. White H.D., Held C., Stewart R. Darapladib for preventing ischemic events in stable coronary disease. N Eng J Med. 2014;370:1702–1711.
    1. O’Donoghue M.L., Braunwald E., White H.D. Effect of darapladib on major coronary events after an acute coronary syndrome: the SOLID-TIMI 52 randomized clinical trial. JAMA. 2014;312:1006–1015.
    1. Wallentin L., Held C., Armstrong P.W. Lipoprotein-associated phospholipase A2 activity is a marker of risk but not a useful target for treatment in patients with stable coronary heart disease. J Am Heart Assoc. 2016;5:e003407.
    1. Ridker P.M., Thuren T., Zalewski A. 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.
    1. Bevilacqua M.P., Pober J.S., Wheeler M.E. Interleukin 1 acts on cultured human vascular endothelium to increase the adhesion of polymorphonuclear leukocytes, monocytes, and related leukocyte cell lines. J Clin Invest. 1985;76:2003–2011.
    1. Dinarello C.A. Interleukin-1beta and the autoinflammatory diseases. N Eng J Med. 2009;360:2467–2470.
    1. Fearon W.F., Fearon D.T. Inflammation and cardiovascular disease: role of the interleukin-1 receptor antagonist. Circulation. 2008;117:2577–2579.
    1. Patti G., D’Ambrosio A., Dobrina A. Interleukin-1 receptor antagonist: a sensitive marker of instability in patients with coronary artery disease. J Thromb Thrombolysis. 2002;14:139–143.
    1. Galea J., Armstrong J., Gadsdon P. Interleukin-1 beta in coronary arteries of patients with ischemic heart disease. Arterioscler Thromb Vasc Biol. 1996;16:1000–1006.
    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. Casini-Raggi V., Kam L., Chong Y.J. Mucosal imbalance of IL-1 and IL-1 receptor antagonist in inflammatory bowel disease. A novel mechanism of chronic intestinal inflammation. J Immunol. 1995;154:2434–2440.
    1. Martinon F., Petrilli V., Mayor A. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–241.
    1. Abbate A., Van Tassell B.W., Seropian I.M. Interleukin-1β modulation using a genetically engineered antibody prevents adverse cardiac remodeling following acute myocardial infarction in the mouse. Eur J Heart Fail. 2010;12:319–322.
    1. Ridker P.M., Howard C.P., Walter V. Effects of interleukin-1[beta] 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. Blann A.D., Nadar S.K., Lip G.Y.H. The adhesion molecule P-selectin and cardiovascular disease. Eur Heart J. 2003;24:2166–2179.
    1. Tenaglia A.N., Buda J.A., Wilkins G.R. Levels of expression of P-selectin, E-selectin, and intercellular adhesion molecule-1 in coronary atherectomy specimens from patients with stable and unstable angina pectoris. Am J Cardiol. 1997;79:742–747.
    1. Scialla J.J., Plantinga L.C., Kao W.H.L. Soluble P-selectin levels are associated with cardiovascular mortality and sudden cardiac death in male dialysis patients. Am J Nephrol. 2011;33:224–230.
    1. Wagner D.D., Burger P.C. Platelets in inflammation and thrombosis. Arterioscler Thromb Vasc Biol. 2003;23:2131–2137.
    1. Burger P.C., Wagner D.D. Platelet P-selectin facilitates atherosclerotic lesion development. Blood. 2003;101:2661–2666.
    1. Phillips W.J., Barringhaus K.G., Sanders J.M. Single injection of P-selectin or P-selectin glycoprotein ligand-1 monoclonal antibody blocks neointima formation after arterial injury in apolipoprotein E-deficient mice. Circulation. 2003;107:2244–2249.
    1. Wang K., Zhou Z., Zhou X. Prevention of intimal hyperplasia with recombinant soluble P-selectin glycoprotein ligand-immunoglobulin in the porcine coronary artery balloon injury model. J Am Coll Cardiol. 2001;38:577–582.
    1. Tanguay J.F., Geoffrey P., Sirois M.G. Prevention of in-stent restenosis via reduction of thrombo-inflammatory reactions with recombinant P-selectin glycoprotein ligand-1. Thromb Haemost. 2004;91:1186–1193.
    1. Tardif J.C., Tanguay J.F., Wright S.R. 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;61:2048–2055.
    1. Stahli B.E., Tardif J.C., Carrier M. Effects of P-selectin antagonist inclacumab in patients undergoing coronary artery bypass graft surgery: SELECT-CABG trial. J Am Coll Cardiol. 2016;67:344–346.
    1. Martin E.D., Bassi R., Marber M.S. p38 MAPK in cardioprotection—are we there yet? Br J Pharmacol. 2015;172:2101–2113.
    1. Fisk M., Gajendragadkar P.R., Mäki-Petäjä K.M., Wilkinson I.B., Cheriyan J. Therapeutic potential of p38 MAP kinase inhibition in the management of cardiovascular disease. Am J Cardiovasc Drugs. 2014;14:155–165.
    1. Cheriyan J., Webb A.J., Sarov-Blat L. Inhibition of p38 mitogen-activated protein kinase improves nitric oxide-mediated vasodilatation and reduces inflammation in hypercholesterolemia. Circulation. 2011;123:515–523.
    1. Denise Martin E., De Nicola G.F., Marber M.S. New therapeutic targets in cardiology: p38 alpha mitogen-activated protein kinase for ischemic heart disease. Circulation. 2012;126:357–368.
    1. Ma X.L., Kumar S., Gao F. Inhibition of p38 mitogen-activated protein kinase decreases cardiomyocyte apoptosis and improves cardiac function after myocardial ischemia and reperfusion. Circulation. 1999;13:1685–1691.
    1. Surinkaew S., Kumphune S., Chattipakorn S. Inhibition of p38 MAPK during ischemia, but not reperfusion, effectively attenuates fatal arrhythmia in ischemia/reperfusion heart. J Cardiovasc Pharmacol. 2013;61:133–141.
    1. See F., Thomas W., Way K. p38 mitogen-activated protein kinase inhibition improves cardiac function and attenuates left ventricular remodeling following myocardial infarction in the rat. J Am Coll Cardiol. 2004;44:1679–1689.
    1. Seeger F.H., Sedding D., Langheinrich A.C., Haendeler J., Zeiher A.M., Dimmeler S. Inhibition of the p38 MAP kinase in vivo improves number and functional activity of vasculogenic cells and reduces atherosclerotic disease progression. Basic Res Cardiol. 2010;105:389–397.
    1. Almagor M., Keren A., Banai S. Increased C-reactive protein level after coronary stent implantation in patients with stable coronary artery disease. Am Heart J. 2003;145:248–253.
    1. Sarov-Blat L., Morgan J.M., Fernandez P. Inhibition of p38 mitogen-activated protein kinase reduces inflammation after coronary vascular injury in humans. Arterioscler Thromb Vasc Biol. 2010;30:2256–2263.
    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. Newby L.K., Marber M.S., Melloni C. Losmapimod, a novel p38 mitogen-activated protein kinase inhibitor, in non-ST-segment elevation myocardial infarction: results of a randomised phase 2 trial. Lancet. 2014;384:1187–1195.
    1. O’Donoghue M.L., Glaser R., Cavender M.A. Effect of losmapimod on cardiovascular outcomes in patients hospitalized with acute myocardial infarction: a randomized clinical trial. JAMA. 2016;315:1591–1599.
    1. Fleming T. Surrogate endpoints and FDA’s accelerated approval process. Health Aff (Millwood) 2005;24:67–78.
    1. Fleming T.R., DeMets D.L. Surrogate end points in clinical trials: are we being misled? Ann Intern Med. 1996;125:605–613.
    1. Davignon J. Beneficial cardiovascular pleiotropic effects of statins. Circulation. 2004;109:39–43.
    1. Eichstadt H.W., Eskotter H., Hoffman I. Improvement of myocardial perfusion by short-term fluvastatin therapy in coronary artery disease. Am J Cardiol. 1995;76:122A–125A.
    1. Crisby M., Nordin-Fredriksson G., Shah P.K. Pravastatin treatment increases collagen content and decreases lipid content, inflammation, metalloproteinases, and cell death in human carotid plaques: implications for plaque stabilization. Circulation. 2001;103:926–933.
    1. Hackman A., Abe Y., Insull W., Jr. Levels of soluble cell adhesion molecules in patients with dyslipidemia. Circulation. 1996;93:1334–1338.
    1. Seljeflot I., Tonstad S., Hjermann I. Reduced expression of endothelial cell markers after 1 year treatment with simvastatin and atorvastatin in patients with coronary heart disease. Atherosclerosis. 2002;162:179–185.
    1. Ridker P.M., Rifai N., Pfeffer M.A., for the Cholesterol and Recurrent Events (CARE) Investigators Long-term effects of pravastatin on plasma concentration of C-reactive protein. Circulation. 1999;100:230–235.
    1. Conn P.M. Elsevier; London: 2013. Animal Models for the Study of Human Disease.
    1. Libby P. Murine “model” monotheism: an iconoclast at the altar of mouse. Circ Res. 2015;117:921–925.
    1. Schwartz R.S., Murphy J.G., Edwards W.D. Restenosis after balloon angioplasty: a practical proliferative model in porcine coronary arteries. Circulation. 1990;82:2190–2200.
    1. Bloor C., White F., Roth D. The pig as a model of myocardial ischemia and gradual coronary artery occlusion. In: Swindle M.M., editor. Swine as Models in Biomedical Research. Iowa State University Press; Ames, Iowa: 1992. pp. 163–175.
    1. Buchwald A.B., Unterberg C., Nebendahl K. Low-molecular-weight heparin reduces neointimal proliferation after coronary stent implantation in hypercholesterolemic minipigs. Circulation. 1992;86:531–537.
    1. Clopath P. The effect of acetylsalicylic acid (ASA) on the development of atherosclerotic lesions in miniature swine. Br J Exp Pathol. 1980;61:440–443.
    1. Legendre C.M., Licht C., Muus P. Terminal complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med. 2013;368:2169–2181.
    1. Kelly R.J., Höchsmann B., Szer J. Eculizumab in Pregnant Patients with Paroxysmal Nocturnal Hemoglobinuria. N Engl J Med. 2015;373:1032–1039.
    1. Yehoshua Z., de Amorim Garcia Filho C.A. Systemic complement inhibition with eculizumab for geographic atrophy in age-related macular degeneration: the COMPLETE study. Ophthalmology. 2014;121:693–701.
    1. Shaddinger B.C., Xu Y., Roger J.H. Platelet aggregation unchanged by lipoprotein-associated phospholipase A2 inhibition: results from an in vitro study and two randomized phase I trials. PLoS One. 2014;9:e83094.
    1. Brucato A., Imazio M., Gattorno M. Effect of anakinra on recurrent pericarditis among patients with colchicine resistance and corticosteroid dependence: the AIRTRIP randomized clinical trial. JAMA. 2016;316:1906–1912.
    1. Scott I.C., Ibrahim F., Simpson G. A randomised trial evaluating anakinra in early active rheumatoid arthritis. Clin Exp Rheumatol. 2016;34:88–93.
    1. Tzanetakou V., Kanni T., Giatrakou S. Safety and efficacy of anakinra in severe hidradenitis suppurativa: a randomized clinical trial. JAMA Dermatol. 2016;152:52–59.
    1. Ataga K.I., Kutlar A., Kanter J. Crizanlizumab for the prevention of pain crises in sickle cell disease. N Engl J Med. 2017;376:429–439.
    1. Schreiber S., Feagan B., D'Haens G. Oral p38 mitogen-activated protein kinase inhibition with BIRB 796 for active Crohn's disease: a randomized, double-blind, placebo-controlled trial. Clin Gastroenterol Hepatol. 2006;4:325–334.

Source: PubMed

3
Subscribe