Clinical Pharmacokinetics and Pharmacodynamics of Evolocumab, a PCSK9 Inhibitor

Sreeneeranj Kasichayanula, Anita Grover, Maurice G Emery, Megan A Gibbs, Ransi Somaratne, Scott M Wasserman, John P Gibbs, Sreeneeranj Kasichayanula, Anita Grover, Maurice G Emery, Megan A Gibbs, Ransi Somaratne, Scott M Wasserman, John P Gibbs

Abstract

Proprotein convertase subtilisin/kexin type 9 (PCSK9) increases plasma low-density lipoprotein cholesterol (LDL-C) by decreasing expression of the LDL receptor on hepatic cells. Evolocumab is a human monoclonal immunoglobulin G2 that binds specifically to human PCSK9 to reduce LDL-C. Evolocumab exhibits nonlinear kinetics as a result of binding to PCSK9. Elimination is predominantly through saturable binding to PCSK9 at lower concentrations and a nonsaturable proteolytic pathway at higher concentrations. The effective half-life of evolocumab is 11-17 days. The pharmacodynamic effects of evolocumab on PCSK9 are rapid, with maximum suppression within 4 h. At steady state, peak reduction of LDL-C occurs approximately 1 week after a subcutaneous dose of 140 mg every 2 weeks (Q2W) and 2 weeks after a subcutaneous dose 420 mg once monthly (QM), and returns towards baseline over the dosing interval. In several clinical studies, these doses of evolocumab reduced LDL-C by approximately 55-75% compared with placebo. Evolocumab also reduced lipoprotein(a) [Lp(a)] levels and improved those of other lipids in clinical studies. No clinically meaningful differences in pharmacodynamic effects on LDL-C were observed in adult subjects regardless of mild/moderate hepatic impairment, renal impairment or renal failure, body weight, race, sex, or age. No clinically meaningful differences were observed for the pharmacodynamic effects of evolocumab on LDL-C between patients who received evolocumab alone or in combination with a statin, resulting in additional lowering of LDL-C when evolocumab was combined with a statin. No dose adjustment is necessary based on patient-specific factors or concomitant medication use.

Conflict of interest statement

Funding

This work was supported by Amgen Inc.

Conflict of interest

Drs. Somaratne and Wasserman are employees of and stockholders in Amgen, Inc. Drs. Somaratne and J.P. Gibbs are identified as inventors on at least one pending patent application owned by Amgen, Inc. relating to evolocumab. Dr. Wasserman appears on a number of pending patents owned by Amgen Inc. relating to evolocumab and PCSK9 inhibition. Drs. Kasichayanula, Grover, Emery, M. Gibbs, and J.P. Gibbs were employees of and stockholders in Amgen at the time this work was completed. Drs. Kasichayanula, M. Gibbs, and J.P. Gibbs are currently employed by AbbVie, Inc. Dr. Grover is currently employed by BioMarin Pharmaceutical Inc. Dr. Emery is currently employed by Seattle Genetics.

Figures

Fig. 1
Fig. 1
Mechanism of action: PCSK9 inhibition with evolocumab increases LDL-R and decreases serum concentrations of LDL-C [, , , , –55]. a The liver is responsible for catabolism of plasma LDL. Hepatocytes express LDL-R that bind LDL and remove it from the plasma. Upon internalization, vesicles containing the LDL–LDL-R complex fuse with endosomes. LDL-R cycles back to the hepatocyte surface to bind additional LDL. Free LDL in endosomes is degraded into lipids, free fatty acids, and amino acids. b PCSK9 is a protein that regulates the expression of LDL-R in the liver. Hepatocytes produce a precursor of PCSK9 that undergoes self-cleavage in the endoplasmic reticulum and ultimately is secreted into plasma as functional PCSK9. Extracellular PCSK9 binds to LDL-Rs on the surface of the hepatocyte and is internalized with the LDL–LDL-R complex. The LDL-R–PCSK9 complex is routed to the lysosome for degradation, thereby preventing the cycling of LDL-R back to the hepatocyte surface. The reduced concentration of LDL-R on the surface of hepatocytes results in a lower rate of plasma LDL elimination. c A monoclonal antibody directed against PCSK9 could lower LDL if binding to circulating PCSK9 blocks the interaction of PCSK9 with cell surface LDL-R. Internalized LDL-R could cycle back to the cell surface instead of being degraded in lysosomes, leading to increased concentrations of LDL-R on the cell surface. This could result in a higher LDL elimination rate by hepatocytes and an overall reduction in plasma LDL. LDL-C low-density lipoprotein cholesterol, LDL-R low-density lipoprotein receptor, PCSK9 proprotein convertase subtilisin/kexin type 9
Fig. 2
Fig. 2
Mean unbound evolocumab serum concentrations and geometric mean percent change from baseline in ultracentrifugation LDL-C and unbound PCSK9 in healthy subjects. a Single-dose SC evolocumab 140 mg. b Single-dose SC evolocumab 420 mg. LDL-C low-density lipoprotein cholesterol, PCSK9 proprotein convertase subtilisin/kexin type 9, SC subcutaneous. Amgen data on file
Fig. 3
Fig. 3
Percent change (mean and 95% confidence interval) from baseline for lipid parameters: time-averaged effect, mean of weeks 10 and 12, and week 12. ApoA1 apolipoprotein A1, ApoB apolipoprotein B, HDL-C high-density lipoprotein cholesterol, LDL-C low-density lipoprotein cholesterol, Q2W once every 2 weeks

References

    1. World Health Organization . World health statistics 2012. Geneva: WHO Press; 2012.
    1. Mozaffarian D, Benjamin EJ, Go AS, et al. Heart disease and stroke statistics–2015 update: a report from the American Heart Association. Circulation. 2015;131:e29–e322. doi: 10.1161/CIR.0000000000000152.
    1. Collaboration Cholesterol Treatment Trialists, Baigent C, Blackwell L, et al. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet. 2010;376:1670–1681. doi: 10.1016/S0140-6736(10)61350-5.
    1. Silverman MG, Ference BA, Im K, et al. Association between lowering LDL-C and cardiovascular risk reduction among different therapeutic interventions: a systematic review and meta-analysis. JAMA. 2016;316:1289–1297. doi: 10.1001/jama.2016.13985.
    1. Grundy SM, Cleeman JI, Merz CN, et al. Implications of recent clinical trials for the National Cholesterol Education Program Adult Treatment Panel III guidelines. Circulation. 2004;110:227–239. doi: 10.1161/01.CIR.0000133317.49796.0E.
    1. Waters DD, Brotons C, Chiang CW, et al. Lipid treatment assessment project 2: a multinational survey to evaluate the proportion of patients achieving low-density lipoprotein cholesterol goals. Circulation. 2009;120:28–34. doi: 10.1161/CIRCULATIONAHA.108.838466.
    1. Lemstra M, Blackburn D, Crawley A, et al. Proportion and risk indicators of nonadherence to statin therapy: a meta-analysis. Can J Cardiol. 2012;28:574–580. doi: 10.1016/j.cjca.2012.05.007.
    1. Mancini GB, Baker S, Bergeron J, et al. Diagnosis, prevention, and management of statin adverse effects and intolerance: Canadian Consensus Working Group Update (2016) Can J Cardiol. 2016;32:S35–S65. doi: 10.1016/j.cjca.2016.01.003.
    1. Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015;372:2387–2397. doi: 10.1056/NEJMoa1410489.
    1. Horton JD, Cohen JC, Hobbs HH. Molecular biology of PCSK9: its role in LDL metabolism. Trends Biochem Sci. 2007;32:71–77. doi: 10.1016/j.tibs.2006.12.008.
    1. Brown MS, Goldstein JL. Biomedicine. Lowering LDL–not only how low, but how long? Science. 2006;311:1721–1723. doi: 10.1126/science.1125884.
    1. Qian YW, Schmidt RJ, Zhang Y, et al. Secreted PCSK9 downregulates low density lipoprotein receptor through receptor-mediated endocytosis. J Lipid Res. 2007;48:1488–1498. doi: 10.1194/jlr.M700071-JLR200.
    1. Horton JD, Cohen JC, Hobbs HH. PCSK9: a convertase that coordinates LDL catabolism. J Lipid Res. 2009;50(Suppl):S172–S177. doi: 10.1194/jlr.R800091-JLR200.
    1. Abifadel M, Varret M, Rabes JP, et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet. 2003;34:154–156. doi: 10.1038/ng1161.
    1. Cohen J, Pertsemlidis A, Kotowski IK, et al. Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9. Nat Genet. 2005;37:161–165. doi: 10.1038/ng1509.
    1. Lopez D. PCSK9: an enigmatic protease. Biochim Biophys Acta. 2008;1781:184–191. doi: 10.1016/j.bbalip.2008.01.003.
    1. Abifadel M, Rabes JP, Devillers M, et al. Mutations and polymorphisms in the proprotein convertase subtilisin kexin 9 (PCSK9) gene in cholesterol metabolism and disease. Hum Mutat. 2009;30:520–529. doi: 10.1002/humu.20882.
    1. Koren MJ, Scott R, Kim JB, et al. Efficacy, safety, and tolerability of a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 as monotherapy in patients with hypercholesterolaemia (MENDEL): a randomised, double-blind, placebo-controlled, phase 2 study. Lancet. 2012;380:1995–2006. doi: 10.1016/S0140-6736(12)61771-1.
    1. Koren MJ, Lundqvist P, Bolognese M, et al. Anti-PCSK9 monotherapy for hypercholesterolemia: the MENDEL-2 randomized, controlled phase III clinical trial of evolocumab. J Am Coll Cardiol. 2014;63:2531–2540. doi: 10.1016/j.jacc.2014.03.018.
    1. Giugliano RP, Desai NR, Kohli P, et al. Efficacy, safety, and tolerability of a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 in combination with a statin in patients with hypercholesterolaemia (LAPLACE-TIMI 57): a randomised, placebo-controlled, dose-ranging, phase 2 study. Lancet. 2012;380:2007–2017. doi: 10.1016/S0140-6736(12)61770-X.
    1. Robinson JG, Nedergaard BS, Rogers WJ, et al. Effect of evolocumab or ezetimibe added to moderate- or high-intensity statin therapy on LDL-C lowering in patients with hypercholesterolemia: the LAPLACE-2 randomized clinical trial. JAMA. 2014;311:1870–1882. doi: 10.1001/jama.2014.4030.
    1. Raal F, Scott R, Somaratne R, et al. Low-density lipoprotein cholesterol-lowering effects of AMG 145, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 serine protease in patients with heterozygous familial hypercholesterolemia: the Reduction of LDL-C with PCSK9 Inhibition in Heterozygous Familial Hypercholesterolemia Disorder (RUTHERFORD) randomized trial. Circulation. 2012;126:2408–2417. doi: 10.1161/CIRCULATIONAHA.112.144055.
    1. Raal FJ, Stein EA, Dufour R, et al. PCSK9 inhibition with evolocumab (AMG 145) in heterozygous familial hypercholesterolaemia (RUTHERFORD-2): a randomised, double-blind, placebo-controlled trial. Lancet. 2015;385:331–340. doi: 10.1016/S0140-6736(14)61399-4.
    1. Sullivan D, Olsson AG, Scott R, et al. Effect of a monoclonal antibody to PCSK9 on low-density lipoprotein cholesterol levels in statin-intolerant patients: the GAUSS randomized trial. JAMA. 2012;308:2497–2506. doi: 10.1001/jama.2012.25790.
    1. Stroes E, Colquhoun D, Sullivan D, et al. Anti-PCSK9 antibody effectively lowers cholesterol in patients with statin intolerance: the GAUSS-2 randomized, placebo-controlled phase 3 clinical trial of evolocumab. J Am Coll Cardiol. 2014;63:2541–2548. doi: 10.1016/j.jacc.2014.03.019.
    1. Nissen SE, Stroes E, Dent-Acosta RE, et al. Efficacy and tolerability of evolocumab vs ezetimibe in patients with muscle-related statin intolerance: the GAUSS-3 randomized clinical trial. JAMA. 2016;315:1580–1590. doi: 10.1001/jama.2016.3608.
    1. Blom DJ, Hala T, Bolognese M, et al. A 52-week placebo-controlled trial of evolocumab in hyperlipidemia. N Engl J Med. 2014;370:1809–1819. doi: 10.1056/NEJMoa1316222.
    1. Sabatine MS, Giugliano RP, Wiviott SD, et al. Efficacy and safety of evolocumab in reducing lipids and cardiovascular events. N Engl J Med. 2015;372:1500–1509. doi: 10.1056/NEJMoa1500858.
    1. Stein EA, Honarpour N, Wasserman SM, et al. Effect of the proprotein convertase subtilisin/kexin 9 monoclonal antibody, AMG 145, in homozygous familial hypercholesterolemia. Circulation. 2013;128:2113–2120. doi: 10.1161/CIRCULATIONAHA.113.004678.
    1. Raal FJ, Honarpour N, Blom DJ, et al. Inhibition of PCSK9 with evolocumab in homozygous familial hypercholesterolaemia (TESLA Part B): a randomised, double-blind, placebo-controlled trial. Lancet. 2015;385:341–350. doi: 10.1016/S0140-6736(14)61374-X.
    1. Stein EA, Giugliano RP, Koren MJ, et al. Efficacy and safety of evolocumab (AMG 145), a fully human monoclonal antibody to PCSK9, in hyperlipidaemic patients on various background lipid therapies: pooled analysis of 1359 patients in four phase 2 trials. Eur Heart J. 2014;35:2249–2259. doi: 10.1093/eurheartj/ehu085.
    1. Nicholls SJ, Puri R, Anderson T, et al. Effect of evolocumab on progression of coronary disease in statin-treated patients: the GLAGOV randomized clinical trial. JAMA. 2016;316:2373–2384. doi: 10.1001/jama.2016.16951.
    1. Chan JC, Piper DE, Cao Q, et al. A proprotein convertase subtilisin/kexin type 9 neutralizing antibody reduces serum cholesterol in mice and nonhuman primates. Proc Natl Acad Sci USA. 2009;106:9820–9825. doi: 10.1073/pnas.0903849106.
    1. Dias CS, Shaywitz AJ, Wasserman SM, et al. Effects of AMG 145 on low-density lipoprotein cholesterol levels: results from 2 randomized, double-blind, placebo-controlled, ascending-dose phase 1 studies in healthy volunteers and hypercholesterolemic subjects on statins. J Am Coll Cardiol. 2012;60:1888–1898. doi: 10.1016/j.jacc.2012.08.986.
    1. Teramoto T, Shaywitz A, Hamilton L, et al. A phase 1, randomized, double-blind, placebo-controlled study of AMG 145 comparing the pharmacokinetics, pharmacodynamics, and safety in healthy Japanese and Caucasian subjects. In: Proceedings of 45th annual scientific meeting of the Japan Atherosclerosis Society, Tokyo; 2013.
    1. Gibbs JP, Slatter JG, Egbuna O, et al. Evaluation of evolocumab (AMG 145), a fully human anti-PCSK9 IgG2 monoclonal antibody, in subjects with hepatic impairment. J Clin Pharmacol. 2017;57:513–523. doi: 10.1002/jcph.832.
    1. Lee WP, Datta BN, Ong BB, et al. Defining the role of lipoprotein apheresis in the management of familial hypercholesterolemia. Am J Cardiovasc Drugs. 2011;11:363–370. doi: 10.2165/11594970-000000000-00000.
    1. Hirayama A, Honarpour N, Yoshida M, et al. Effects of evolocumab (AMG 145), a monoclonal antibody to PCSK9, in hypercholesterolemic, statin-treated Japanese patients at high cardiovascular risk–primary results from the phase 2 YUKAWA study. Circ J. 2014;78:1073–1082. doi: 10.1253/circj.CJ-14-0130.
    1. REPATHA® (evolocumab) Prescribing Information. Thousand Oaks: Amgen, Inc.; 2016.
    1. Gibbs JP, Doshi S, Kuchimanchi M, et al. Impact of target-mediated elimination on the dose and regimen of evolocumab, a human monoclonal antibody against proprotein convertase subtilisin/kexin type 9 (PCSK9) J Clin Pharmacol. 2017;57:616–626. doi: 10.1002/jcph.840.
    1. Colbert A, Umble-Romero A, Prokop S, et al. Characterization of a quantitative method to measure free proprotein convertase subtilisin/kexin type 9 in human serum. MAbs. 2014;6:1103–1113. doi: 10.4161/mabs.28719.
    1. Koren MJ, Doshi S, Castro R, et al. Comparisons of peak LDL-C reduction and duration of effect with lower or higher dosing regimens of the PCSK9 inhibitor evolocumab [abstract] Circulation. 2015;132:A12729.
    1. Rosenson RS, Jacobson TA, Preiss D, et al. Efficacy and safety of the PCSK9 inhibitor evolocumab in patients with mixed hyperlipidemia. Cardiovasc Drugs Ther. 2016;30:305–313. doi: 10.1007/s10557-016-6666-1.
    1. Raal FJ, Hovingh GK, Blom D, et al. Long-term treatment with evolocumab added to conventional drug therapy, with or without apheresis, in patients with homozygous familial hypercholesterolaemia: an interim subset analysis of the open-label TAUSSIG study. Lancet Diabetes Endocrinol. 2017;5:280–290. doi: 10.1016/S2213-8587(17)30044-X.
    1. Raal FJ, Giugliano RP, Sabatine MS, et al. PCSK9 inhibition-mediated reduction in Lp(a) with evolocumab: an analysis of 10 clinical trials and the LDL receptor’s role. J Lipid Res. 2016;57:1086–1096. doi: 10.1194/jlr.P065334.
    1. Watts GF, Chan DC, Dent R, et al. Factorial effects of evolocumab and atorvastatin on lipoprotein metabolism. Circulation. 2017;135:338–351. doi: 10.1161/CIRCULATIONAHA.116.025080.
    1. Desai NR, Giugliano RP, Wasserman SM, et al. Association between circulating baseline proprotein convertase subtilisin kexin type 9 levels and efficacy of evolocumab. JAMA Cardiol. 2017;2:556–560. doi: 10.1001/jamacardio.2016.5395.
    1. Sabatine MS, Giugliano RP, Keech AC, et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017;376:1713–1722. doi: 10.1056/NEJMoa1615664.
    1. Lee E, Gibbs J, Wasserman SM, et al. Pharmacokinetics and pharmacodynamics of evolocumab in patients with renal impairment [abstract] Eur Heart J. 2016;37(Suppl):343.
    1. Dubuc G, Chamberland A, Wassef H, et al. Statins upregulate PCSK9, the gene encoding the proprotein convertase neural apoptosis-regulated convertase-1 implicated in familial hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2004;24:1454–1459. doi: 10.1161/01.ATV.0000134621.14315.43.
    1. Mayne J, Dewpura T, Raymond A, et al. Plasma PCSK9 levels are significantly modified by statins and fibrates in humans. Lipids Health Dis. 2008;7:22. doi: 10.1186/1476-511X-7-22.
    1. Brown MS, Goldstein JL. Receptor-mediated endocytosis: insights from the lipoprotein receptor system. Proc Natl Acad Sci USA. 1979;76:3330–3337. doi: 10.1073/pnas.76.7.3330.
    1. Steinberg D, Witztum JL. Inhibition of PCSK9: a powerful weapon for achieving ideal LDL cholesterol levels. Proc Natl Acad Sci USA. 2009;106:9546–9547. doi: 10.1073/pnas.0904560106.
    1. Goldstein JL, Brown MS. The LDL receptor. Arterioscler Thromb Vasc Biol. 2009;29:431–438. doi: 10.1161/ATVBAHA.108.179564.
    1. Zhang DW, Lagace TA, Garuti R, et al. Binding of proprotein convertase subtilisin/kexin type 9 to epidermal growth factor-like repeat A of low density lipoprotein receptor decreases receptor recycling and increases degradation. J Biol Chem. 2007;282:18602–18612. doi: 10.1074/jbc.M702027200.

Source: PubMed

3
Subscribe