Pharmacometric analyses to characterize the effect of CSL112 on apolipoprotein A-I and cholesterol efflux capacity in acute myocardial infarction patients

Bo Zheng, Danielle Duffy, Pierluigi Tricoci, Helen Kastrissios, Marc Pfister, Samuel D Wright, Andreas Gille, Michael A Tortorici, Bo Zheng, Danielle Duffy, Pierluigi Tricoci, Helen Kastrissios, Marc Pfister, Samuel D Wright, Andreas Gille, Michael A Tortorici

Abstract

Aims: To characterize relationships between apolipoprotein A-I (apoA-I) exposure and cholesterol efflux capacity (CEC) and covariate effects following CSL112 (apoA-I [human]) administration in an integrated population including acute myocardial infarction (AMI) patients.

Methods: A pharmacometric analysis utilized data from seven clinical trials, including patients with AMI, subjects with renal impairment and healthy subjects. A population pharmacokinetic (PK) analysis was performed to relate CSL112 doses to changes in apoA-I plasma concentrations. Covariate analysis was conducted to identify sources of variability in apoA-I exposure. Exposure-response modeling was conducted to describe the relationship between apoA-I exposure and total or ATP binding cassette transporter A1-(ABCA1)-dependent CEC and to identify clinical predictors of CEC.

Results: A two-compartment model described apoA-I PK. ApoA-I clearance was slightly lower in subjects with AMI, whereas baseline apoA-I was marginally higher in female and Japanese subjects. Covariate effects on apoA-I exposure were in the order of 10% and thus not clinically relevant. The relationships between apoA-I exposure and CECs were described by nonlinear models. Simulations showed CEC elevation resulting from apoA-I exposure increment was comparable in AMI and non-AMI subjects; no covariate had clinically meaningful effects on CEC. Simulations also demonstrated that CEC in patients with AMI post 6 g CSL112 dosing was substantially elevated compared to placebo and lower dose levels.

Conclusions: The model-based exposure-response analysis demonstrated, irrespective of body weight, sex and race, that fixed 6 g CSL112 dosing causes a desired CEC elevation, which may benefit AMI patients by potentially reducing early recurrent cardiovascular event risk.

Trial registration: ClinicalTrials.gov NCT01129661 NCT01281774 NCT02427035 NCT01499420 NCT02742103 NCT02108262.

Keywords: atherosclerosis; clinical trials; modelling and simulation; pharmacokinetics-pharmacodynamics; pharmacometrics.

Conflict of interest statement

B.Z., D.D., P.T., B.K., S.W., A.G. and M.T. are employees of CSL Behring. H.K. and M.P. are employees of Certara.

© 2020 CSL Behring. British Journal of Clinical Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.

Figures

FIGURE 1
FIGURE 1
Prediction‐corrected visual predictive check (VPC) obtained through n = 1000 simulations from the final population PK model by regimen: placebo, single doses and multiple doses (QW × 4) of CSL112, and by all subjects, healthy subjects and patients. The blue circles represent the prediction‐corrected observed apoA‐I concentrations. The solid and dashed red lines represent the observed median and 10th‐90th percentile range, respectively. The pink and purple bands represent the 95% confidence interval (CI) of the median prediction, and the 10th and 90th prediction interval, respectively. ApoA‐I, apolipoprotein A‐I; QW × 4, weekly dose for 4 weeks
FIGURE 2
FIGURE 2
Box plots of model‐estimated individual apoA‐I exposures (AUC0‐48) following the fourth infusion by dose and race in healthy subjects (A) and by dose and sex in post‐AMI subjects (B), and the median value of AUC0‐48 from each group of post‐AMI subjects (B). Circles denote outlying data. AMI, acute myocardial infarction; AUC0‐48, area under the curve from 0 to 48 hours; Jp, Japanese; M, male; F, female
FIGURE 3
FIGURE 3
Scatter plots of observed apoA‐I concentrations vs observed total and ABCA1‐dependent CEC by race (Japanese and non‐Japanese) (A, B), AMI status (C, D), sex (E, F) and weight (G, H). ApoA‐I, apolipoprotein A‐I; ABCA1, ATP‐binding cassette transporter‐1; AMI, acute myocardial infarction; CEC, cholesterol efflux capacity
FIGURE 4
FIGURE 4
Prediction‐corrected visual predictive check (VPC) of the exposure‐response models for total CEC (A) and ABCA1‐dependent CEC (B). The blue circles represent the prediction‐corrected observed CECs. The solid and dashed red lines represent the observed median and 10th‐90th percentile range, respectively. The pink and purple bands represent the 95% confidence intervals of the 90th, median and 10th percentiles of the simulated data. ABCA1, ATP‐binding cassette transporter 1; CEC, cholesterol efflux capacity
FIGURE 5
FIGURE 5
Forest plots of the model‐predicted covariate (AMI, sex and body weight) effects on total (A)‐(D) and ABCA1‐dependent CEC (E)‐(H) in response to different increment in apoA‐I concentration. Typical subjects were defined as non‐Japanese, healthy males with apoA‐I baseline concentration of 121.9 mg dL−1. Baseline‐corrected CECs in response to increment in apoA‐I concentration, which is equal to 50% (A, E), 75% (B, F), 100% (C, G) and 125% (D, H) of a typical subject's baseline, respectively, were simulated and summarized. The covariate effects were displayed as fold change from median predicted CEC in typical subject. For each covariate value, the horizontal line represents the 90% prediction interval for the respective ratio. The vertical dashed lines indicate the predefined clinical relevance criteria (0.80‐1.25, bioequivalence range). ApoA‐I, apolipoprotein A‐I; AMI, acute myocardial infarction; ABCA1, ATP‐binding cassette transporter‐1; CEC, cholesterol efflux capacity
FIGURE 6
FIGURE 6
Model‐predicted individual time‐averaged total (A) and ABCA1‐dependent (B) CEC (AUC0‐48/48 h) vs baseline‐corrected apoA‐I exposure (AUC0‐48). The black lines and yellow shaded area represent median and 90% prediction interval, respectively, of power‐model predictions for 1000 virtual subjects. Blue, green and red circles represent individual model estimations for subjects assigned to placebo, 2 g and 6 g, respectively, in the AEGIS‐I and CSL112_1002 studies. Blue horizontal dash lines indicate the efflux capacity threshold, which is the median of predicted CEC in the placebo group. Horizontal bars and black star represent 90% CI and median of individual apoA‐I exposures for placebo and the CSL112 2 g and 6 g doses for subjects in both the AEGIS‐I and CSL112_1002 studies. ApoA‐I, apolipoprotein A‐I; ABCA1, ATP‐binding cassette transporter‐1; AUC0‐48, area under the curve from 0‐48 hours; CEC, cholesterol efflux capacity

References

    1. Tall AR. An overview of reverse cholesterol transport. Eur Heart J. 1998;19(Suppl A):A31‐A35.
    1. Tabas I. Consequences of cellular cholesterol accumulation: basic concepts and physiological implications. J Clin Invest. 2002;110(7):905‐911.
    1. Du XM, Kim MJ, Hou L, et al. HDL particle size is a critical determinant of ABCA1‐mediated macrophage cellular cholesterol export. Circ Res. 2015;116(7):1133‐1142.
    1. Ray KK, Ditmarsch M, Kallend D, et al. The effect of cholesteryl ester transfer protein inhibition on lipids, lipoproteins, and markers of HDL function after an acute coronary syndrome: the dal‐ACUTE randomized trial. Eur Heart J. 2014;35(27):1792‐1800.
    1. Soares AAS, Tavoni TM, de Faria EC, Remalay AT, Maranhao RC, Sposito AC. HDL acceptor capacities for cholesterol efflux from macrophages and lipid transfer are both acutely reduced after myocardial infarction. Clin Chim Acta. 2018;478:51‐56.
    1. Annema W, Willemsen HM, de Boer JF, et al. HDL function is impaired in acute myocardial infarction independent of plasma HDL cholesterol levels. J Clin Lipidol. 2016;10(6):1318‐1328.
    1. Rohatgi A, Khera A, Berry JD, et al. HDL cholesterol efflux capacity and incident cardiovascular events. N Engl J Med. 2014;371(25):2383‐2393.
    1. Khera AV, Cuchel M, de la Llera‐Moya M, et al. Cholesterol efflux capacity, high‐density lipoprotein function, and atherosclerosis. N Engl J Med. 2011;364(2):127‐135.
    1. Liu C, Zhang Y, Ding D, et al. Cholesterol efflux capacity is an independent predictor of all‐cause and cardiovascular mortality in patients with coronary artery disease: a prospective cohort study. Atherosclerosis. 2016;249:116‐124.
    1. Guerin M, Silvain J, Gall J, et al. Association of serum cholesterol efflux capacity with mortality in patients with ST‐segment elevation myocardial infarction. J Am Coll Cardiol. 2018;72:3259‐3269.
    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;33:2202‐2211.
    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:e002171.
    1. Gibson MC, 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;134:1918‐1930.
    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;34:2106‐2114.
    1. Silvain J, Kerneis M, Guerin M, Montalescot G. Modulation of cholesterol efflux capacity in patients with myocardial infarction. Curr Opin Cardiol. 2019;34(6):714‐720.
    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;54:301‐310.
    1. Australian New Zealand Clinical Trials Registry (ANZCTR) . A phase 1, randomized, double‐blind, placebo‐controlled study to evaluate the pharmacokinetics, safety, and tolerability of CSL112 in healthy Japanese and Caucasian subjects. Available at: . Date last updated: November 22 2018. Date last accessed: 08 May 2019.
    1. Gille A, Duffy D, Tortorici MA, Wright SD, Deckelbaum LI, D'Andrea DM. Moderate renal impairment does not impact the ability of CSL112 (apolipoprotein A‐I [human]) to enhance cholesterol efflux capacity. J Clin Pharmacol. 2019;59:427‐436.
    1. Tortorici MA, Duffy D, Evans R, et al. Pharmacokinetics and safety of CSL112 (apolipoprotein A‐I [human]) in adults with moderate renal impairment and normal renal function. Clin Pharmacol Drug Dev. 2018;8(5):628‐636. 10.1002/cpdd.618
    1. Gibson CM, Kerneis M, Yee MK, et al. The CSL112‐2001 trial: safety and tolerability of multiple doses of CSL112 (apolipoprotein A‐I [human]), an intravenous formulation of plasma‐derived apolipoprotein A‐I, among subjects with moderate renal impairment after acute myocardial infarction. Am Heart J. 2019;208:81‐90.
    1. Gille A, D'Andrea D, Tortorici MA, Hartel G, Wright SD. CSL112 (Apolipoprotein A‐I [human]) enhances cholesterol efflux similarly in healthy individuals and stable atherosclerotic disease patients. Arterioscler Thromb Vasc Biol. 2018;38:953‐963.
    1. Beal SL, Sheiner LB, Boeckmann AJ, Bauer RJ. NONMEM users guides. Ellicott City, Maryland, USA: Icon Development Solutions; 1989. ‐2011.
    1. Lindbom L, Pihlgren P, Jonsson EN. PsN‐toolkit‐‐a collection of computer intensive statistical methods for non‐linear mixed effect modeling using NONMEM. Comput Methods Programs Biomed. 2005;79(3):241‐257.
    1. Mendivil CO, Furtado J, Morton AM, Wang L, Sacks FM. Novel pathways of apolipoprotein A‐I metabolism in high‐density lipoprotein of different sizes in humans. Arterioscler Thromb Vasc Biol. 2016;36:156‐165.
    1. Velez‐Carrasco W, Lichtenstein AH, Li Z, et al. Apolipoprotein A‐I and A‐II kinetic parameters as assessed by endogenous labeling with [(2)H(3)]leucine in middle‐aged and elderly men and women. Arterioscler Thromb Vasc Biol. 2000;20:801‐806.
    1. Whyte MB, Shojaee‐Moradie F, Sharaf SE, et al. HDL‐apoA‐I kinetics in response to 16 wk of exercise training in men with nonalcoholic fatty liver disease. Am J Physiol Endocrinol Metab. 2020;318(6):E839‐E847.
    1. Capodanno D, Mehran R, Gibson CM, Angiolillo DJ. CSL112, a reconstituted, infusible, plasma‐derived apolipoprotein A‐I: safety and tolerability profiles and implications for management in patients with myocardial infarction. Expert Opin Investig Drugs. 2018;27(12):997‐1005.
    1. Tortorici MA, Gille A, Liss C, et al. Direct enhancement of cholesterol efflux in AMI patients: a PKPD substudy of AEGIS‐I. Eur Heart J. 2017;38(Suppl 1):1106.
    1. Alexander SPH, Kelly E, Mathie A, et al. THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: transporters. Br J Pharmacol. 2019;176:S397‐S493.
    1. Gibson CM, Kastelein JJ, Phillips A, Aylward PE, Yee MK, Tendera M, Nicholls SJ, Pocock S, Goodman SG, Alexander JH, Lincoff AM, Bode C, Duffy D, Heise M, Berman G, Mears SJ, Tricoci P, Deckelbaum LI, Steg PG, Ridker P, Mehran R. Rationale and design of ApoA‐I Event Reducing in Ischemic Syndromes II (AEGIS‐II): A phase 3, multicenter, double‐blind, randomized, placebo‐controlled, parallel‐group study to investigate the efficacy and safety of CSL112 in subjects after acute myocardial infarction In preparation 2020.
    1. Yokoyama S. Unique features of high‐density lipoproteins in the Japanese: in population and in genetic factors. Nutrients. 2015;7:2359‐2381.
    1. Zitnanova I, Siarnik P, Fullop M, et al. Gender differences in LDL‐ and HDL‐cholesterol subfractions in patients after the acute ischemic stroke and their association with oxidative stress markers. J Clin Biochem Nutr. 2018;63(2):144‐148.
    1. Nagano M, Yamashita S, Hirano K, et al. Molecular mechanisms of cholesteryl ester transfer protein deficiency in Japanese. J Atheroscler Thromb. 2004;11(3):110‐121.
    1. Yancey PG, Bortnick AE, Kellner‐Weibel G, de la Llera‐Moya M, Phillips MC, Rothblat GH. Importance of different pathways of cellular cholesterol efflux. Arterioscler Thromb Vasc Biol. 2003;23:712‐719.
    1. Oram JF, Lawn RM, Garvin MR, Wade DP. ABCA1 is the cAMP‐inducible apolipoprotein receptor that mediates cholesterol secretion from macrophages. J Biol Chem. 2000;275(44):34508‐34511.
    1. Phillips MC. Molecular mechanisms of cellular cholesterol efflux. J Biol Chem. 2014;289(35):24020‐24029.
    1. de la Llera‐Moya M, Drazul‐Schrader D, Asztalos BF, Cuchel M, Rader DJ, Rothblat GH. The ability to promote efflux via ABCA1 determines the capacity of serum specimens with similar high‐density lipoprotein cholesterol to remove cholesterol from macrophages. Arterioscler Thromb Vasc Biol. 2010;30:796‐801.
    1. Soares AAS, Tavoni TM, de Faria EC, et al. HDL acceptor capacities for cholesterol efflux from macrophages and lipid transfer are both acutely reduced after myocardial infarction. Clin Chim Acta. 2018;478:51‐56.
    1. Potteaux S, Gautier EL, Hutchison SB, et al. Suppressed monocyte recruitment drives macrophage removal from atherosclerotic plaques of Apoe−/− mice during disease regression. J Clin Invest. 2011;121(5):2025‐2036.
    1. Feig JE, Rong JX, Shamir R, et al. HDL promotes rapid atherosclerosis regression in mice and alters inflammatory properties of plaque monocyte‐derived cells. Proc Natl Acad Sci USA. 2011;108(17):7166‐7171.
    1. Cimmino G, Ibanez B, Vilahur G, et al. Up‐regulation of reverse cholesterol transport key players and rescue from global inflammation by ApoA‐I (Milano). J Cell Mol Med. 2009;13(9B):3226‐3235.
    1. Murphy AJ, Funt S, Gorman D, Tall AR, Wang N. Pegylation of high‐density lipoprotein decreases plasma clearance and enhances antiatherogenic activity. Circ Res. 2013;113(1):e1‐e9.

Source: PubMed

3
Se inscrever