Changes in mipomersen dosing regimen provide similar exposure with improved tolerability in randomized placebo-controlled study of healthy volunteers

Joann D Flaim, John S Grundy, Brenda F Baker, Mary P McGowan, John J P Kastelein, Joann D Flaim, John S Grundy, Brenda F Baker, Mary P McGowan, John J P Kastelein

Abstract

Background: Mipomersen, an apolipoprotein B synthesis inhibitor, demonstrated significant reductions in low-density lipoprotein (LDL) cholesterol, non-high density lipoprotein cholesterol, and apolipoprotein B in 4 phase 3 studies at the FDA-approved subcutaneous dose of 200 mg once weekly.

Methods and results: A short-term phase 1 study in healthy volunteers was conducted to evaluate the relative bioavailability, safety, and tolerability of mipomersen in 2 test dose regimens in reference to the 200 mg weekly dose regimen. Eighty-four adults were randomized to 1 of 3 cohorts (30 mg once daily, 70 mg 3 times weekly, or 200 mg once weekly) and then mipomersen or placebo (3:1 ratio) for 3 weeks of treatment. Comparable mipomersen post-distribution phase plasma concentrations were observed across the 3 dose regimens suggesting similar tissue exposure. Injection site reactions were reported, but did not lead to treatment discontinuation. The median incidence of these responses per injection was decreased by lowering the dose. Signals from a diverse panel of systemic inflammation markers were essentially indistinguishable between dose regimens and placebo treatment. The one exception was a modest transient post-dose elevation of C-reactive protein (CRP) in the mipomersen 200 mg weekly group. This elevation was not associated with an increase in other proinflammatory markers.

Conclusions: This study demonstrated a similar drug exposure and overall safety profile between the 3 dosing regimens. Exploratory assessment of a diverse panel of biomarkers found no indication of a systemic inflammatory response to mipomersen treatment. These results support assessment of alternative dose regimens in longer-term studies.

Clinical trial registration url: http://www.clinicaltrials.gov. Unique identifier: NCT01061814.

Keywords: dosing; inhibitor; pharmacokinetics; randomized controlled trial; safety.

Figures

Figure 1.
Figure 1.
Evaluation of mipomersen in alternative dose regimens. Study design (A) and flow of participants (B) through the study. In the study design diagram, R denotes randomization, ratios are indicated above by cohort (R1) and active to placebo (R2), and the asterisk (*) indicates the time point for exploratory efficacy measurements, Day 28. †One subject discontinued dosing in the 30 mg group for asthenia and atrial flutter—both events were considered unlikely related to study drug. ‡Two subjects discontinued dosing in the 70 mg group: one subject for gastroesophageal reflux disease, this event was considered possibly related to study drug; and one subject for ALT and AST ≥3×ULN, muscle tightness, increase in lactate dehydrogenase and creatine kinase, these events were considered unrelated to study drug. ALT indicates alanine transaminase; AST, aspartate transaminase; QD, once daily; QW, once weekly; TIW, 3 times a week; ULN, upper limit of normal.
Figure 2.
Figure 2.
Mean mipomersen plasma concentrations over time by dose regimen. A, 0 to 24 hours after the first dose. B, 0 to 35 days after the last dose. QD indicates once daily; QW, once weekly; TIW, 3 times a week.
Figure 3.
Figure 3.
Effect of SC dosing regimen on serum IL‐6 and CRP levels. A, Relative median levels after first and last dose‐by‐dose regimen cohort. B, Individual maximum post‐dose changes after first and last dose by treatment groups. Median and interquartile range values are provided in Tables 5 and 6. *Dose day, samples collected pre‐dose. CRP indicates C‐reactive protein; IL, interleukin; MIPO, mipomersen; PBO, placebo; QD indicates once daily; QW, once weekly; SC, subcutaneous; TIW, 3 times a week.
Figure 4.
Figure 4.
Effect of SC dosing regimen on inflammation biomarkers. Type I interferons, IFN‐α and ‐β (A); chemokines, MIP‐1α, and MCP‐1 (B); immune cell signaling and activation, IL‐1β, and IL‐13 (C); complement split products, Bb and C5a (D); and IL‐6 and CRP (E). Data presented are the median absolute changes from baseline (BSLN), ± the interquartile range (IQR). Bb and C5a indicates complement split products; CRP, C‐reactive protein; IFN, interferon; IL, interleukin; MCP‐1, monocyte chemotactic protein 1; MIP‐1α, macrophage inflammatory protein 1 alpha; QD indicates once daily; QW, once weekly; SC, subcutaneous; TIW, 3 times a week.

References

    1. Toth PP. Emerging LDL therapies: mipomersen‐antisense oligonucleotide therapy in the management of hypercholesterolemia. J Clin Lipidol. 2013; 7:S6-S10
    1. Raal FJ, Santos RD, Blom DJ, Marais AD, Charng MJ, Cromwell WC, Lachmann RH, Gaudet D, Tan JL, Chasan‐Taber S, Tribble DL, Flaim JD, Crooke ST. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double‐blind, placebo‐controlled trial. Lancet. 2010; 375:998-1006
    1. McGowan M, Tardif JC, Ceska R, Burgess LJ, Soran H, Gouni‐Berthold I, Wagener G, Chasan‐Taber S. Randomized, placebo‐controlled trial of mipomersen in patients with severe hypercholesterolemia receiving maximally tolerated lipid‐lowering therapy. PLoS ONE. 2012; 7:e4900610.1371/journalpone.0049006
    1. Stein EA, Dufour R, Gagne C, Gaudet D, East C, Donovan JM, Chin W, Tribble DL, McGowan M. Apolipoprotein B synthesis inhibition with mipomersen in heterozygous familial hypercholesterolemia: results of a randomized, double‐blind, placebo controlled trial to assess efficacy and safety as add‐on therapy in patients with coronary artery disease. Circulation. 2012; 126:2283-2292
    1. Thomas GS, Cromwell W, Ali S, Flaim JD, Chin W, Davidson M. Mipomersen, an apolipoprotein B synthesis inhibitor, reduces atherogenic lipoproteins in patients with severe hypercholesterolemia at high cardiovascular risk: a randomized, double‐blind, placebo‐controlled trial. J Am Coll Cardiol. 2013; 62:2178-2184
    1. Kastelein JJ, Wedel MK, Baker BF, Su J, Bradley JD, Yu RZ, Chuang E, Graham MJ, Crooke RM. Potent reduction of apolipoprotein B and low‐density lipoprotein cholesterol by short‐term administration of an antisense inhibitor of apolipoprotein B. Circulation. 2006; 114:1729-1735
    1. Akdim F, Stroes ES, Sijbrands EJ, Tribble DL, Trip MD, Jukema JW, Flaim JD, Su J, Yu R, Baker BF, Wedel MK, Kastelein JJ. Efficacy and safety of mipomersen, an antisense inhibitor of apolipoprotein B, in hypercholesterolemic subjects receiving stable statin therapy. J Am Coll Cardiol. 2010; 55:1611-1618
    1. Akdim F, Tribble DL, Flaim JD, Yu R, Su J, Geary RS, Baker BF, Fuhr R, Wedel MK, Kastelein JJ. Efficacy of apolipoprotein B synthesis inhibition in subjects with mild‐to‐moderate hyperlipidaemia. Eur Heart J. 2011; 32:2650-2659
    1. Yu RZ, Lemonidis KM, Graham MJ, Matson JE, Crooke RM, Tribble DL, Wedel MK, Levin AA, Geary RS. Cross‐species comparison of in vivo PK/PD relationships for second‐generation antisense oligonucleotides targeting apolipoprotein B‐100. Biochem Pharmacol. 2009; 77:910-919
    1. Yu RZ, Kim TW, Hong A, Watanabe TA, Gaus HJ, Geary RS. Cross‐species pharmacokinetic comparison from mouse to man of a second‐generation antisense oligonucleotide, ISIS 301012, targeting human apolipoprotein B‐100. Drug Metab Dispos. 2007; 35:460-468
    1. Yu RZ, Grundy JS, Geary RS. Clinical pharmacokinetics of second generation antisense oligonucleotides. Expert Opin Drug Metab Toxicol. 2013; 9:169-182
    1. Libby P, Ridker PM, Hansson GKLeducq Transatlantic Network on Atherothrombosis. Inflammation in atherosclerosis: from pathophysiology to practice. J Am Coll Cardiol. 2009; 54:2129-2138
    1. Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Jr, Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, Macfadyen JG, Nordestgaard BG, Shepherd J, Willerson JT, Glynn RJJUPITER Trial Study Group. Reduction in C‐reactive protein and LDL cholesterol and cardiovascular event rates after initiation of rosuvastatin: a prospective study of the JUPITER trial. Lancet. 2009; 373:1175-1182
    1. IL6R Genetics Consortium Emerging Risk Factors Collaboration. Sarwar N, Butterworth AS, Freitag DF, Gregson J, Willeit P, Gorman DN, Gao P, Saleheen D, Rendon A, Nelson CP, Braund PS, Hall AS, Chasman DI, Tybjærg‐Hansen A, Chambers JC, Benjamin EJ, Franks PW, Clarke R, Wilde AA, Trip MD, Steri M, Witteman JC, Qi L, van der Schoot CE, de Faire U, Erdmann J, Stringham HM, Koenig W, Rader DJ, Melzer D, Reich D, Psaty BM, Kleber ME, Panagiotakos DB, Willeit J, Wennberg P, Woodward M, Adamovic S, Rimm EB, Meade TW, Gillum RF, Shaffer JA, Hofman A, Onat A, Sundström J, Wassertheil‐Smoller S, Mellström D, Gallacher J, Cushman M, Tracy RP, Kauhanen J, Karlsson M, Salonen JT, Wilhelmsen L, Amouyel P, Cantin B, Best LG, Ben‐Shlomo Y, Manson JE, Davey‐Smith G, de Bakker PI, O'Donnell CJ, Wilson JF, Wilson AG, Assimes TL, Jansson JO, Ohlsson C, Tivesten Å, Ljunggren Ö, Reilly MP, Hamsten A, Ingelsson E, Cambien F, Hung J, Thomas GN, Boehnke M, Schunkert H, Asselbergs FW, Kastelein JJ, Gudnason V, Salomaa V, Harris TB, Kooner JS, Allin KH, Nordestgaard BG, Hopewell JC, Goodall AH, Ridker PM, Hólm H, Watkins H, Ouwehand WH, Samani NJ, Kaptoge S, Di Angelantonio E, Harari O, Danesh J. Interleukin‐6 receptor pathways in coronary heart disease: a collaborative meta‐analysis of 82 studies. Lancet. 2012; 379:1205-1213
    1. Henry SP, Kim TW, Kramer‐Stickland K, Zanardi TA, Fey RA, Levin AA. In: Crooke ST. (ed.). Toxicologic properties of 2′‐O‐methoxyethyl chimeric antisense inhibitors in animals and man. Antisense Drug Technology: Principles, Strategies, and Applications. 20072nd edBoca Raton, FL: CRC Press/Taylor & Francis Group; 327-364
    1. Henry SP, Geary RS, Yu R, Levin AA. Drug properties of second‐generation antisense oligonucleotides: how do they measure up to their predecessors? Curr Opin Investig Drugs. 2001; 2:1444-1449
    1. Schroder K, Tschopp J. The inflammasomes. Cell. 2010; 140:821-832
    1. Kawai T, Akira S. Toll‐like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity. 2011; 34:637-650
    1. Guo L, Junttila IS, Paul WE. Cytokine‐induced cytokine production by conventional and innate lymphoid cells. Trends Immunol. 2012; 33:598-606
    1. Gabay C, Kushner I. Acute‐phase proteins and other systemic responses to inflammation. N Engl J Med. 1999; 340:448-454
    1. Krieg AM, Efler SM, Wittpoth M, Al Adhami MJ, Davis HL. Induction of systemic Th1‐like innate immunity in normal volunteers following subcutaneous but not intravenous administration of CPG 7909, a synthetic B‐class CpG oligodeoxynucleotide TLR9 agonist. J Immunother. 2004; 27:460-471
    1. Yu RZ, Geary RS, Flaim JD, Riley GC, Tribble DL, van Vliet AA, Wedel MK. Lack of pharmacokinetic interaction of mipomersen sodium (ISIS 301012), a 2′‐O‐methoxyethyl modified antisense oligonucleotide targeting apolipoprotein B‐100 messenger RNA, with simvastatin and ezetimibe. Clin Pharmacokinet. 2009; 48:39-50
    1. Borden EC, Sen GC, Uze G, Silverman RH, Ransohoff RM, Foster GR, Stark GR. Interferons at age 50: past, current and future impact on biomedicine. Nat Rev Drug Discov. 2007; 6:975-990
    1. Descotes J, Vial T. In: House RV, Descotes J. (eds.). Flu‐like syndrome and cytokines. Methods in Pharmacology and Toxicology: Cytokines in Human Health: Immunotoxicology, Pathology, and Therapeutic Applications. 2007Totowa, NJ: Humana Press Inc; 193-204
    1. Moser B, Wolf M, Walz A, Loetscher P. Chemokines: multiple levels of leukocyte migration control. Trends Immunol. 2004; 25:75-84
    1. Esche C, Stellato C, Beck LA. Chemokines: key players in innate and adaptive immunity. J Invest Dermatol. 2005; 125:615-628
    1. Bode JG, Albrecht U, Häussinger D, Heinrich PC, Schaper F. Hepatic acute phase proteins: regulation by IL‐6‐ and IL‐1‐type cytokines involving STAT3 and its crosstalk with NF‐κB‐dependent signaling. Eur J Cell Biol. 2012; 91:496-505
    1. Walport MJ. Complement. First of two parts. N Engl J Med. 2001; 344:1058-1066
    1. Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: a key system for immune surveillance and homeostasis. Nat Immunol. 2010; 11:785-797
    1. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER). Guidance for industry: statistical approaches to establishing bioequivalence. January 2001. Available at: Accessed November 1, 2013.
    1. Moreland LW, Schiff MH, Baumgartner SW, Tindall EA, Fleischmann RM, Bulpitt KJ, Weaver AL, Keystone EC, Furst DE, Mease PJ, Ruderman EM, Horwitz DA, Arkfeld DG, Garrison L, Burge DJ, Blosch CM, Lange ML, McDonnell ND, Weinblatt ME. Etanercept therapy in rheumatoid arthritis. A randomized, controlled trial. Ann Intern Med. 1999; 130:478-486
    1. Mikol DD, Barkhof F, Chang P, Coyle PK, Jeffery DR, Schwid SR, Stubinski B, Uitdehaag BREGARD study group. Comparison of subcutaneous interferon beta‐1a with glatiramer acetate in patients with relapsing multiple sclerosis (the REbif vs Glatiramer Acetate in Relapsing MS Disease [REGARD] study): a multicentre, randomised, parallel, open‐label trial. Lancet Neurol. 2008; 7:903-914
    1. Diamant M, Van Gaal L, Stranks S, Northrup J, Cao D, Taylor K, Trautmann M. Once weekly exenatide compared with insulin glargine titrated to target in patients with type 2 diabetes (DURATION‐3): an open‐label randomised trial. Lancet. 2010; 375:2234-2243
    1. Kerkmann M, Costa LT, Richter C, Rothenfusser S, Battiany J, Hornung V, Johnson J, Englert S, Ketterer T, Heckl W, Thalhammer S, Endres S, Hartmann G. Spontaneous formation of nucleic acid‐based nanoparticles is responsible for high interferon‐alpha induction by CpG‐A in plasmacytoid dendritic cells. J Biol Chem. 2005; 280:8086-8093
    1. Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, Matsumoto M, Hoshino K, Wagner H, Takeda K, Akira S. A Toll‐like receptor recognizes bacterial DNA. Nature. 2000; 408:740-745
    1. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006; 124:783-801
    1. Volanakis JE. Human C‐reactive protein: expression, structure, and function. Mol Immunol. 2001; 38:189-197
    1. Kofoed K, Andersen O, Kronborg G, Tvede M, Petersen J, Eugen‐Olsen J, Larsen K. Use of plasma C‐reactive protein, procalcitonin, neutrophils, macrophage migration inhibitory factor, soluble urokinase‐type plasminogen activator receptor, and soluble triggering receptor expressed on myeloid cells‐1 in combination to diagnose infections: a prospective study. Crit Care. 2007; 11:R38.
    1. El‐Sharrawy EA, El‐Hakim IE, Sameeh E. Attenuation of C‐reactive protein increases after exodontia by tramadol and ibuprofen. Anesth Prog. 2006; 53:78-82
    1. Neubauer O, König D, Wagner KH. Recovery after an Ironman triathlon: sustained inflammatory responses and muscular stress. Eur J Appl Physiol. 2008; 104:417-426
    1. Urbanski A, Schwarz T, Neuner P, Krutmann J, Kirnbauer R, Köck A, Luger TA. Ultraviolet light induces increased circulating interleukin‐6 in humans. J Invest Dermatol. 1990; 94:808-811
    1. C Reactive Protein Coronary Heart Disease Genetics Collaboration (CCGC). Wensley F, Gao P, Burgess S, Kaptoge S, Di Angelantonio E, Shah T, Engert JC, Clarke R, Davey‐Smith G, Nordestgaard BG, Saleheen D, Samani NJ, Sandhu M, Anand S, Pepys MB, Smeeth L, Whittaker J, Casas JP, Thompson SG, Hingorani AD, Danesh J. Association between C reactive protein and coronary heart disease: Mendelian randomisation analysis based on individual participant data. BMJ. 2011; 342:d548.
    1. Pearson TA, Mensah GA, Alexander RW, Anderson JL, Cannon RO, III, Criqui M, Fadl YY, Fortmann SP, Hong Y, Myers GL, Rifai N, Smith SC, Jr, Taubert K, Tracy RP, Vinicor F. Markers of inflammation and cardiovascular disease: application to clinical and public health practice: a statement for healthcare professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation. 2003; 107:499-511

Source: PubMed

3
Abonnere