Pharmacokinetics and Safety of CSL112 (Apolipoprotein A-I [Human]) in Adults With Moderate Renal Impairment and Normal Renal Function

Michael A Tortorici, Danielle Duffy, Rebecca Evans, John Feaster, Andreas Gille, Timothy G K Mant, Samuel D Wright, Denise D'Andrea, Michael A Tortorici, Danielle Duffy, Rebecca Evans, John Feaster, Andreas Gille, Timothy G K Mant, Samuel D Wright, Denise D'Andrea

Abstract

CSL112 (Apolipoprotein A-I [human]) is an intravenous preparation of apolipoprotein A-I (apoA-I), formulated with phosphatidylcholine (PC) and stabilized with sucrose, in development to prevent early recurrent cardiovascular events following acute myocardial infarction (AMI). This phase 1 study was designed to determine if moderate renal impairment (RI) influenced the pharmacokinetics (PK) and safety of CSL112. Thirty-two subjects, 16 with moderate RI (estimated glomerular filtration rate [eGFR] ≥ 30 and < 60 mL/min/1.73 m2 ) and 16 age-, sex-, and weight-matched subjects with normal renal function (eGFR ≥ 90 mL/min/1.73 m2 ) were randomized 3:1 to receive a single infusion of CSL112 2 g (n = 6) or placebo (n = 2), or CSL112 6 g (n = 6) or placebo (n = 2). PK sampling was at prespecified times from 48 hours prior to 144 hours following infusions, with final safety assessments at 90 days. Renal and hepatic safety, and adverse events (AEs) were monitored throughout the study. Plasma apoA-I and PC PK profiles were similar between renal function cohorts at both doses. For CSL112 6 g mean ± SD apoA-I AUC0-last was 7670 ± 1900 and 9170 ± 2910 mg·h/dL in normal renal function and moderate RI subjects, respectively. Renal apoA-I clearance was <1% of CSL112 dose. In moderate RI, sucrose clearance was slower; however, approximately 70% was excreted within 48 hours in both renal function cohorts. No CSL112-related serious AEs or clinically significant renal or hepatic safety changes were observed. Dose adjustment of CSL112 is not required in subjects with moderate RI, supporting its further investigation in AMI patients with moderate RI.

Trial registration: ClinicalTrials.gov NCT02427035.

Keywords: atherosclerosis; chronic kidney disease; lipids; pharmacokinetics; randomized controlled trial.

© 2018 The Authors. Clinical Pharmacology in Drug Development Published by Wiley Periodicals, Inc. on behalf of The American College of Clinical Pharmacology.

Figures

Figure 1
Figure 1
Study design (A) and schedule of visits and assessments (B). Dose escalation to 6 g CSL112 was recommended following Data Safety and Monitoring Board review of data for the 2 g CSL112 dose. DSMB, Data and Safety Monitoring Board; Mod RI, moderate renal impairment; NRF, normal renal function; PD, pharmacodynamics; PK, pharmacokinetics.
Figure 2
Figure 2
Plasma pharmacokinetic profiles of apolipoprotein A‐I and phosphatidylcholine following CSL112 infusion. Shown are mean + standard deviation unadjusted (A) apolipoprotein A‐I (apoA‐I) and (B) phosphatidylcholine (PC) plasma concentration‐time profiles by renal function group, moderate renal impairment (RI), and normal renal function following infusion of CSL112 2 or 6 g. Please note that the small differences in the 2 g curves are attributable to differences in values at baseline rather than differences in response to CSL112.
Figure 3
Figure 3
Relationship between plasma clearance and baseline eGFR for apolipoprotein A‐I. Slope (90% confidence band), −0.000417 (−0.0015 to 0.000314). CL, systematic plasma clearance; eGFR, estimated glomerular filtration rate.
Figure 4
Figure 4
Pharmacokinetic profile of sucrose in plasma following CSL112 infusion. Shown are mean + standard deviation unadjusted sucrose plasma concentration‐time profiles for each renal function group, moderate renal impairment (RI), and normal renal function following infusion of CSL112 2 or 6 g.

References

    1. Diditchenko S, Gille A, Pragst I, et al. Novel formulation of a reconstituted high‐density lipoprotein (CSL112) dramatically enhances ABCA1‐dependent cholesterol efflux. Arterioscler Thromb Vasc Biol. 2013;33(9):2202–2211.
    1. Dharmarajan K, Hsieh AF, Kulkarni VT, et al. Trajectories of risk after hospitalization for heart failure, acute myocardial infarction, or pneumonia: retrospective cohort study. BMJ. 2015;350:h411.
    1. Jernberg T, Hasvold P, Henriksson M, Hjelm H, Thuresson M, Janzon M. Cardiovascular risk in post‐myocardial infarction patients: nationwide real world data demonstrate the importance of a long‐term perspective. Eur Heart J. 2015;36(19):1163–1170.
    1. Smolina K, Wright FL, Rayner M, Goldacre MJ. Long‐term survival and recurrence after acute myocardial infarction in England, 2004 to 2010. Circ Cardiovasc Qual Outcomes. 2012;5(4):532–540.
    1. Anavekar NS, McMurray JJ, Velazquez EJ, et al. Relation between renal dysfunction and cardiovascular outcomes after myocardial infarction. N Engl J Med. 2004;351(13):1285–1295.
    1. Easton R, Gille A, D'Andrea D, Davis R, Wright SD, Shear C. A multiple ascending dose study of CSL112, an infused formulation of ApoA‐I. J Clin Pharmacol. 2014;54(3):301–310.
    1. Steinitz K. The renal excretion of sucrose in normal man; comparison with inulin. Am J Physiol 1940;129(2):252–258.
    1. Tricoci P, D'Andrea DM, Gurbel PA, et al. Infusion of reconstituted high‐density lipoprotein, CSL112, in patients with atherosclerosis: safety and pharmacokinetic results from a phase 2a randomized clinical trial. J Am Heart Assoc. 2015;4(8):e002171.
    1. Tortorici MA, Gille A, Liss C, et al. Direct augmentation of cholesterol efflux capacity in AMI patients: a PKPD substudy of AEGIS‐I. Eur Heart J. 2017;38(suppl 1):P1106.
    1. Gibson CM, Korjian S, Tricoci P, et al. Safety and tolerability of CSL112, a reconstituted, infusible, plasma‐derived apolipoprotein A‐I, after acute myocardial infarction: the AEGIS‐I Trial (ApoA‐I Event Reducing in Ischemic Syndromes I). Circulation. 2016;134(24):1918–1930.
    1. Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604–612.
    1. Gille A, Easton R, D'Andrea D, Wright SD, Shear CL. CSL112 enhances biomarkers of reverse cholesterol transport after single and multiple infusions in healthy subjects. Arterioscler Thromb Vasc Biol. 2014;34(9):2106–2114.
    1. Yao Z, Pfister M, Comisar C, et al. ApoA‐I enhancement, by CSL112 administration, leads to a linear increase in total cholesterol efflux capacity. Poster presented at the American Conference of Pharmacometrics (ACoP). 2014.
    1. Glass C, Pittman RC, Weinstein DB, Steinberg D. Dissociation of tissue uptake of cholesterol ester from that of apoprotein A‐I of rat plasma high density lipoprotein: selective delivery of cholesterol ester to liver, adrenal, and gonad. Proc Natl Acad Sci U S A. 1983;80(17):5435–5439.
    1. Aseem O, Smith BT, Cooley MA, et al. Cubilin maintains blood levels of HDL and albumin. J Am Soc Nephrol. 2014;25(5):1028–1036.
    1. Storm T, Emma F, Verroust PJ, Hertz JM, Nielsen R, Christensen EI. A patient with cubilin deficiency. N Engl J Med. 2011;364(1):89–91.

Source: PubMed

3
購読する