Phase 1B, Randomized, Double-Blinded, Dose Escalation, Single-Center, Repeat Dose Safety and Pharmacodynamics Study of the Oral NLRP3 Inhibitor Dapansutrile in Subjects With NYHA II-III Systolic Heart Failure

George F Wohlford, Benjamin W Van Tassell, Hayley E Billingsley, Dinesh Kadariya, Justin M Canada, Salvatore Carbone, Virginia L Mihalick, Aldo Bonaventura, Alessandra Vecchié, Juan Guido Chiabrando, Edoardo Bressi, Georgia Thomas, Ai-Chen Ho, Amr A Marawan, Megan Dell, Cory R Trankle, Jeremy Turlington, Roshanak Markley, Antonio Abbate, George F Wohlford, Benjamin W Van Tassell, Hayley E Billingsley, Dinesh Kadariya, Justin M Canada, Salvatore Carbone, Virginia L Mihalick, Aldo Bonaventura, Alessandra Vecchié, Juan Guido Chiabrando, Edoardo Bressi, Georgia Thomas, Ai-Chen Ho, Amr A Marawan, Megan Dell, Cory R Trankle, Jeremy Turlington, Roshanak Markley, Antonio Abbate

Abstract

The NLRP3 inflammasome has been implicated in the development and progression of heart failure. The aim of this study was to determine the safety of an oral inhibitor of the NLRP3 inflammasome, dapansutrile (OLT1177), in patients with heart failure and reduced ejection fraction (HFrEF). This was a phase 1B, randomized, double-blind, dose escalation, single-center, repeat dose safety and pharmacodynamics study of dapansutrile in stable patients with HFrEF (New York Heart Association Class II-III). Subjects were randomized to treatment with dapansutrile for up to 14 days at a ratio of 4:1 into 1 of 3 sequential ascending dose cohorts (500, 1000, or 2000 mg) each including 10 patients. Subjects underwent clinical assessment, biomarker determination, transthoracic echocardiogram, and maximal cardiopulmonary exercise testing at baseline, day 14, and day 28 to ascertain changes in clinical status. Placebo cases (N = 2 per cohort) were used as a decoy to reduce bias and not for statistical comparisons. Thirty participants (20 men) were treated for 13 (12-14) days. No serious adverse events during the study were recorded. All clinical or laboratory parameters at day 14 compared with baseline suggested clinical stability without significant within-group differences in the dapansutrile-pooled group or the 3 dapansutrile cohorts. Improvements in left ventricular EF [from 31.5% (27.5-39) to 36.5% (27.5-45), P = 0.039] and in exercise time [from 570 (399.5-627) to 616 (446.5-688) seconds, P = 0.039] were seen in the dapansutrile 2000 mg cohort. Treatment with dapansutrile for 14 days was safe and well tolerated in patients with stable HFrEF.

Trial registration: ClinicalTrials.gov NCT03534297.

Conflict of interest statement

B. W. Van Tassell has served as a consultant for Novartis and Serpin Pharma. A. Abbate has served as a consultant for AstraZeneca, Janssen, Kiniksa Pharmaceuticals Ltd, Merck, Novartis, Olatec, and Serpin Pharma. A. Bonaventura and A. Vecchié received a travel grant from Kiniksa Pharmaceuticals Ltd to attend the 2019 American Heart Association Scientific Sessions. S. Carbone is supported by a Career Development Award 19CDA34660318 from the American Heart Association. The remaining authors report no conflicts of interest.

Copyright © 2020 The Author(s). Published by Wolters Kluwer Health, Inc.

Figures

FIGURE 1.
FIGURE 1.
CONSORT diagram of the flow of study participants.
FIGURE 2.
FIGURE 2.
Dapansutrile levels during the study period. A dose-dependent increase in the dapansutrile level was observed across the 3 cohorts, whereas no detectable levels were recorded in the placebo cohort.
FIGURE 3.
FIGURE 3.
LVEF modifications across the study period. LVEF significantly increased from baseline at day 12 ± 2 in the dapansutrile-pooled cohort. This was mainly because of an improvement in LVEF in the dapansutrile 2000 mg daily cohort, whereas no significant changes in the 500 mg daily cohort, 1000 mg daily cohort, or in the placebo-pooled group were observed. *P < 0.05 for Wilcoxon signed-rank.
FIGURE 4.
FIGURE 4.
Exercise time changes across the study period. A significant increase in the exercise time was found in the dapansutrile 2000 mg daily cohort, but not in the dapansutrile-pooled, the 500 mg daily, the 1000 mg daily, or the placebo-pooled groups. *P < 0.05 for Wilcoxon signed-rank.

References

    1. Murphy SP, Kakkar R, McCarthy CP, et al. Inflammation in heart failure: JACC state-of-the-art Review. J Am Coll Cardiol. 2020;75:1324–1340.
    1. Buckley LF, Abbate A. Interleukin-1 blockade in cardiovascular diseases: a clinical update. Eur Heart J. 2018;39:2063–2069.
    1. Page RL, II, O'Bryant CL, Cheng D, et al. American heart association clinical P, heart F, transplantation committees of the council on clinical C, council on cardiovascular S, anesthesia, council on C, stroke N, council on quality of C, outcomes R. Drugs that may cause or exacerbate heart failure: a scientific statement from the American heart association. Circulation. 2016;134:e32–69.
    1. Pepine CJ, Gurbel PA. Cardiovascular safety of NSAIDs: additional insights after PRECISION and point of view. Clin Cardiol. 2017;40:1352–1356.
    1. Sholter DE, Armstrong PW. Adverse effects of corticosteroids on the cardiovascular system. Can J Cardiol. 2000;16:505–511.
    1. Toldo S, Abbate A. The NLRP3 inflammasome in acute myocardial infarction. Nat Rev Cardiol. 2018;15:203–214.
    1. Mauro AG, Bonaventura A, Mezzaroma E, et al. NLRP3 inflammasome in acute myocardial infarction. J Cardiovasc Pharmacol. 2019;74:175–187.
    1. Van Tassell BW, Seropian IM, Toldo S, et al. Interleukin-1beta induces a reversible cardiomyopathy in the mouse. Inflamm Res. 2013;62:637–640.
    1. Van Tassell BW, Arena R, Biondi-Zoccai G, et al. Effects of interleukin-1 blockade with anakinra on aerobic exercise capacity in patients with heart failure and preserved ejection fraction (from the D-HART pilot study). Am J Cardiol. 2014;113:321–327.
    1. Abbate A, Kontos MC, Grizzard JD, et al. Interleukin-1 blockade with anakinra to prevent adverse cardiac remodeling after acute myocardial infarction (Virginia Commonwealth University Anakinra Remodeling Trial [VCU-ART] Pilot study). Am J Cardiol. 2010;105:1371–1377 e1.
    1. Van Tassell BW, Canada J, Carbone S, et al. Interleukin-1 blockade in recently decompensated systolic heart failure: results from REDHART (recently decompensated heart failure anakinra response trial). Circ Heart Fail. 2017;10:e004373.
    1. Buckley LF, Carbone S, Trankle CR, et al. Effect of interleukin-1 blockade on left ventricular systolic performance and work: a post hoc pooled analysis of 2 clinical trials. J Cardiovasc Pharmacol. 2018;72:68–70.
    1. Toldo S, Mauro AG, Cutter Z, et al. Inflammasome, pyroptosis, and cytokines in myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2018;315:H1553–H68.
    1. Marchetti C, Swartzwelter B, Gamboni F, et al. OLT1177, a beta-sulfonyl nitrile compound, safe in humans, inhibits the NLRP3 inflammasome and reverses the metabolic cost of inflammation. Proc Natl Acad Sci U S A. 2018;115:E1530–E9.
    1. Marchetti C, Swartzwelter B, Koenders MI, et al. NLRP3 inflammasome inhibitor OLT1177 suppresses joint inflammation in murine models of acute arthritis. Arthritis Res Ther. 2018;20:169.
    1. Toldo S, Mauro AG, Cutter Z, et al. The NLRP3 inflammasome inhibitor, OLT1177 (dapansutrile), reduces infarct size and preserves contractile function after ischemia reperfusion injury in the mouse. J Cardiovasc Pharmacol. 2019;73:215–222.
    1. Klück V, Jansen TLTA, Janssen M, et al. Dapansutrile, an oral selective NLRP3 inflammasome inhibitor, for treatment of gout flares: an open-label, dose-adaptive, proof-of-concept, phase 2a trial. Lancet Rheumatol. 2020;2:e270–e280.
    1. Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16:31–41.
    1. Lang RM, Badano LP, Mor-Avi V, et al. Recommendations for cardiac chamber quantification by echocardiography in adults: an update from the American society of echocardiography and the European association of, cardiovascular imaging. Eur Heart J Cardiovasc Imag. 2015;16:233–270.
    1. Fletcher GF, Ades PA, Kligfield P, et al. American heart association exercise CR, prevention committee of the council on clinical Cardiology CoNPA, metabolism CoC, stroke N, council on E, prevention. Exercise standards for testing and training: a scientific statement from the American heart association. Circulation. 2013;128:873–934.
    1. Green CP, Porter CB, Bresnahan DR, et al. Development and evaluation of the Kansas City Cardiomyopathy Questionnaire: a new health status measure for heart failure. J Am Coll Cardiol. 2000;35:1245–1255.
    1. Hlatky MA, Boineau RE, Higginbotham MB, et al. A brief self-administered questionnaire to determine functional capacity (the Duke Activity Status Index). Am J Cardiol. 1989;64:651–654.
    1. Daut RL, Cleeland CS, Flanery RC. Development of the Wisconsin Brief Pain Questionnaire to assess pain in cancer and other diseases. Pain. 1983;17:197–210.
    1. Shirazi LF, Bissett J, Romeo F, et al. Role of inflammation in heart failure. Curr Atheroscler Rep. 2017;19:27.
    1. Wintrich J, Kindermann I, Ukena C, et al. Therapeutic approaches in heart failure with preserved ejection fraction: past, present, and future. Clin Res Cardiol. 2020;109:1079–1098.
    1. Montecucco F, Liberale L, Bonaventura A, et al. The role of inflammation in cardiovascular outcome. Curr Atheroscler Rep. 2017;19:11.
    1. Ayoub KF, Pothineni NVK, Rutland J, et al. Immunity, inflammation, and oxidative stress in heart failure: emerging molecular targets. Cardiovasc Drugs Ther. 2017;31:593–608.
    1. Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377:1119–1131.
    1. Everett BM, Cornel JH, Lainscak M, et al. Anti-inflammatory therapy with canakinumab for the prevention of hospitalization for heart failure. Circulation. 2019;139:1289–1299.
    1. Trankle CR, Canada JM, Cei L, et al. Usefulness of canakinumab to improve exercise capacity in patients with long-term systolic heart failure and elevated C-reactive protein. Am J Cardiol. 2018;122:1366–1370.
    1. Abbate A, Trankle CR, Buckley LF, et al. Interleukin-1 blockade inhibits the acute inflammatory response in patients with ST-segment-elevation myocardial infarction. J Am Heart Assoc. 2020;9:e014941.
    1. Trankle CR, Canada JM, Kadariya D, et al. IL-1 blockade reduces inflammation in pulmonary arterial hypertension and right ventricular failure: a single-arm, open-label, phase IB/II pilot study. Am J Respir Crit Care Med. 2019;199:381–384.
    1. Abbate A, Kontos MC, Abouzaki NA, et al. Comparative safety of interleukin-1 blockade with anakinra in patients with ST-segment elevation acute myocardial infarction (from the VCU-ART and VCU-ART2 pilot studies). Am J Cardiol. 2015;115:288–292.
    1. Van Tassell BW, Abouzaki NA, Oddi Erdle C, et al. Interleukin-1 blockade in acute decompensated heart failure: a randomized, double-blinded, placebo-controlled pilot study. J Cardiovasc Pharmacol. 2016;67:544–551.
    1. Van Tassell BW, Trankle CR, Canada JM, et al. IL-1 blockade in patients with heart failure with preserved ejection fraction. Circ Heart Fail. 2018;11:e005036.
    1. Abbate A, Van Tassell BW, Biondi-Zoccai G, et al. Effects of interleukin-1 blockade with anakinra on adverse cardiac remodeling and heart failure after acute myocardial infarction [from the Virginia Commonwealth University-Anakinra Remodeling Trial (2) (VCU-ART2) pilot study]. Am J Cardiol. 2013;111:1394–1400.
    1. Mechanick JI, Farkouh ME, Newman JD, et al. Cardiometabolic-based chronic disease, adiposity and dysglycemia drivers: JACC state-of-the-art Review. J Am Coll Cardiol. 2020;75:525–538.
    1. Canada JM, Van Tassell BW, Christopher S, et al. Clinical predictors of response to anakinra in patients with heart failure. Int J Cardiol. 2014;173:537–539.

Source: PubMed

3
Iratkozz fel