First administration of the Fc-attenuated anti-β amyloid antibody GSK933776 to patients with mild Alzheimer's disease: a randomized, placebo-controlled study

Niels Andreasen, Monica Simeoni, Henrik Ostlund, Pia I Lisjo, Tormod Fladby, Amy E Loercher, Gerard J Byrne, Frances Murray, Paul T Scott-Stevens, Anders Wallin, Yinghua Y Zhang, Lena H Bronge, Henrik Zetterberg, Agneta K Nordberg, Astrid J Yeo, Shahid A Khan, Jan Hilpert, Prafull C Mistry, Niels Andreasen, Monica Simeoni, Henrik Ostlund, Pia I Lisjo, Tormod Fladby, Amy E Loercher, Gerard J Byrne, Frances Murray, Paul T Scott-Stevens, Anders Wallin, Yinghua Y Zhang, Lena H Bronge, Henrik Zetterberg, Agneta K Nordberg, Astrid J Yeo, Shahid A Khan, Jan Hilpert, Prafull C Mistry

Abstract

Objective: To assess the safety, tolerability, pharmacokinetics, and pharmacodynamics of the Fc-inactivated anti-β amyloid (Aβ) monoclonal antibody (mAb) GSK933776 in patients with mild Alzheimer's disease (AD) or mild cognitive impairment (MCI).

Methods: This was a two-part, single blind, placebo-controlled, first-time-in-human (FTIH) study of single (n = 18) and repeat dose (n = 32) intravenous GSK933776 0.001-6 mg/kg (ClinicalTrials.gov: NCT00459550). Additional safety data from an open-label, uncontrolled, single dose study of intravenous GSK933776 1-6 mg/kg (n = 18) are included (ClinicalTrials.gov: NCT01424436).

Results: There were no cases of amyloid-related imaging abnormalities-edema (ARIA-E) or -hemorrhage (ARIA-H) after GSK933776 administration in both studies. Three patients across the two studies developed anti-GSK933776 antibodies. Plasma GSK933776 half-life (t1/2) was 10-15 days after repeat dosing. After each of three administrations of GSK933776, plasma levels of total Aβ42 and Aβ increased whereas plasma levels of free Aβ decreased dose dependently; no changes were observed for placebo. For total Aβ42 the peak:trough ratio was ≤2 at doses ≥3 mg/kg; for total Aβ the ratio was ≤2 at 6 mg/kg. CSF concentrations of Aβ showed increases from baseline to week 12 for Aβ X-38 (week 12:baseline ratio: 1.65; 95%CI: 1.38, 1.93) and Aβ X-42 (week 12:baseline ratio: 1.18; 95%CI: 1.06, 1.30) for values pooled across doses.

Conclusion: In this FTIH study the Fc-inactivated anti-Aβ mAb GSK933776 engaged its target in plasma and CSF without causing brain ARIA-E/H in patients with mild AD or MCI.

Trial registration: ClinicalTrials.gov NCT00459550.

Conflict of interest statement

Competing Interests: MS, AL, FM, PSS, YZ, PM, AY, SK & JH are employees of GSK, whose company funded this study. PIL is an employee of TrialCo AB. LH is an employee of Aleris Diagnostic AB Sabbatsberg. The Independent Data Monitoring Committee included Trident Clinical Research Pty Ltd who provided study conduct and monitoring. There are no patents, products in development or marketed products to declare. This does not alter the authors’ adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Disposition of patients.
Fig 1. Disposition of patients.
A = active; P = placebo; AP = active followed by placebo; PA = placebo followed by active; AA = received active in both parts.
Fig 2. GSK933776 plasma pharmacokinetics.
Fig 2. GSK933776 plasma pharmacokinetics.
Time-course of plasma concentrations of GSK933776 by dose: medians (lines) and individual data (dots). LLQ is 100 ng/mL for the 0.1 mg/kg dose and 5 μg/mL for the 1, 3, and 6 mg/kg doses. SD = single dose; RD = repeat dose. Maximum plasma concentrations increased with dose.
Fig 3. GSK933776 plasma pharmacodynamics.
Fig 3. GSK933776 plasma pharmacodynamics.
A) Geometric mean plasma Aβ concentration–time plots over the three dosing intervals (semi-log plot). Plasma levels of total Aβ42 and Aβ increased whereas plasma levels of free Aβ decreased in dose-dependent manner. Peak:trough ratios for Aβ decreased with increasing dose of GSK933776. B) Week 12 ratio to baseline for CSF Aβ (Aβ1–42 and AβX–42) concentrations. Presented as individual values and mean (95%CI). There were no significant changes from baseline for Aβ1–42 or AβX–42.

References

    1. Karran E, Mercken M, De Strooper B (2011) The amyloid cascade hypothesis for Alzheimer’s disease: an appraisal for the development of therapeutics. Nat Rev Drug Discov 10: 698–712. 10.1038/nrd3505
    1. Salloway S, Sperling R, Gilman S, Fox NC, Blennow K, et al. (2009) A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology 73: 2061–2070. 10.1212/WNL.0b013e3181c67808
    1. Black RS, Sperling RA, Safirstein B, Motter RN, Pallay A, et al. (2010) A single ascending dose study of bapineuzumab in patients with Alzheimer disease. Alzheimer Dis Assoc Disord 24: 198–203. 10.1097/WAD.0b013e3181c53b00
    1. Rinne JO, Brooks DJ, Rossor MN, Fox NC, Bullock R, et al. (2010) 11C-PiB PET assessment of change in fibrillar amyloid-beta load in patients with Alzheimer’s disease treated with bapineuzumab: a phase 2, double-blind, placebo-controlled, ascending-dose study. Lancet Neurol 9: 363–372. 10.1016/S1474-4422(10)70043-0
    1. Blennow K, Zetterberg H, Rinne JO, Salloway S, Wei J, et al. (2012) Effect of immunotherapy with bapineuzumab on cerebrospinal fluid biomarker levels in patients with mild to moderate Alzheimer disease. Arch Neurol 69: 1002–1010.
    1. Ostrowitzki S, Deptula D, Thurfjell L, Barkhof F, Bohrmann B, et al. (2012) Mechanism of amyloid removal in patients with Alzheimer disease treated with gantenerumab. Arch Neurol 69: 198–207. 10.1001/archneurol.2011.1538
    1. Sperling R, Salloway S, Raskind M, Ferris S, Liu E, et al. (2012) A randomized, double-blind, placebo-controlled clinical trial of intravenous bapineuzumab in patients with Alzheimer’s disease who are apolipoprotein E ε4 carriers. Eur J Neurol 19 (Suppl. 1): 70.
    1. Salloway S, Sperling R, Honig L, Porsteinsson A, Sabbagh M, et al. (2012) A randomized, double-blind, placebo-controlled clinical trial of intravenous bapineuzumab in patients with Alzheimer’s disease who are apolipoprotein E ε4 non-carriers. Eur J Neurol 19 (Suppl. 1): 70.
    1. Salloway S, Sperling R, Fox NC, Blennow K, Klunk W, et al. (2014) Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N Engl J Med 370: 322–333. 10.1056/NEJMoa1304839
    1. Kerchner GA, Boxer AL (2010) Bapineuzumab. Expert Opin Biol Ther 10: 1121–1130. 10.1517/14712598.2010.493872
    1. Karran E, Hardy J (2014) Antiamyloid therapy for Alzheimer’s disease—are we on the right road? N Engl J Med 370: 377–378. 10.1056/NEJMe1313943
    1. Sperling RA, Jack CR, Black SE, Frosch MP, Greenberg SM, et al. (2011) Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: recommendations from the Alzheimer’s Association Research Roundtable Workgroup. Alzheimers Dement 7: 367–385. 10.1016/j.jalz.2011.05.2351
    1. Kabat EA, Wu TT, Perry HM, Gottesman KS, Foeller C (1991) Sequences of proteins of immunological interest. 5th ed Cambridge, MA: US Department of Health and Human Services.
    1. Leyhe T, Andreasen N, Simeoni M, Reich A, von Arnim CAF, et al. (2014). Modulation of beta amyloid by a single dose of GSK933776 in patients with mild Alzheimer’s disease: a Phase I study. Alzheimers Res Ther (in press).
    1. Folstein MF, Folstein SE, McHugh PR (1975) "Mini-Mental State". A practical method for grading the cognitive state of patients for the clinician. J Psych Res 12: 189–198.
    1. Sjögren M, Vanderstichele H, Agren H, Zachrisson O, Edsbagge M, et al. (2001) Tau and Abeta42 in cerebrospinal fluid from healthy adults 21–93 years of age: establishment of reference values. Clin Chem 10: 1776–1781.
    1. Zetterberg H, Wahlund LO, Blennow K (2003) Cerebrospinal fluid markers for prediction of Alzheimer’s disease. Neurosci Lett 352: 67–69.
    1. Doody RS, Thomas RG, Farlow M, Iwatsubo T, Vellas B, et al. (2014) Phase III trials of solanezumab for mild-to-moderate Alzheimer’s disease. N Engl J Med 370: 311–321 10.1056/NEJMoa1312889
    1. Carlson C, Estergard W, Oh J, Suhy J, Jack CR Jr, et al. (2011) Prevalence of asymptomatic vasogenic edema in pretreatment of Alzheimer’s disease study cohorts from phase 3 trials of semagacestat and solanezumab. Alzheimers Dement 7: 396–401. 10.1016/j.jalz.2011.05.2353
    1. Davda JP, Hansen RJ (2010) Properties of a general PK/PD model of antibody–ligand interactions for therapeutic antibodies that bind to soluble endogenous targets. MAbs 2: 576–588. 10.4161/mabs.2.5.12833
    1. Farlow M, Arnold SE, van Dyck CH, Aisen PS, Snider BJ, et al. (2012) Safety and biomarker effects of solanezumab in patients with Alzheimer’s disease. Alzheimers Dement 8: 261–271. 10.1016/j.jalz.2011.09.224

Source: PubMed

3
S'abonner