Pharmacodynamics of atabecestat (JNJ-54861911), an oral BACE1 inhibitor in patients with early Alzheimer's disease: randomized, double-blind, placebo-controlled study

Maarten Timmers, Johannes Rolf Streffer, Alberto Russu, Yushin Tominaga, Hiroko Shimizu, Ayako Shiraishi, Kanaka Tatikola, Pascale Smekens, Anne Börjesson-Hanson, Niels Andreasen, Jorge Matias-Guiu, Miquel Baquero, Mercè Boada, Ina Tesseur, Luc Tritsmans, Luc Van Nueten, Sebastiaan Engelborghs, Maarten Timmers, Johannes Rolf Streffer, Alberto Russu, Yushin Tominaga, Hiroko Shimizu, Ayako Shiraishi, Kanaka Tatikola, Pascale Smekens, Anne Börjesson-Hanson, Niels Andreasen, Jorge Matias-Guiu, Miquel Baquero, Mercè Boada, Ina Tesseur, Luc Tritsmans, Luc Van Nueten, Sebastiaan Engelborghs

Abstract

Background: β-Secretase enzyme (BACE) inhibition has been proposed as a priority treatment mechanism for Alzheimer's disease (AD), but treatment initiation may need to be very early. We present proof of mechanism of atabecestat (also known as JNJ-54861911), an oral BACE inhibitor for the treatment of AD, in Caucasian and Japanese populations with early AD who do not show signs of dementia.

Methods: In two similarly designed phase I studies, a sample of amyloid-positive elderly patients comprising 45 Caucasian patients with early AD diagnosed as preclinical AD (n = 15, Clinical Dementia Rating [CDR] = 0) or with mild cognitive impairment due to AD (n = 30, CDR = 0.5) and 18 Japanese patients diagnosed as preclinical AD (CDR-J = 0) were randomized 1:1:1 to atabecestat 10 or 50 mg or placebo (n = 6-8/treatment) daily for 4 weeks. Safety, pharmacokinetics (PK), and pharmacodynamics (PD) (i.e., reduction of cerebrospinal fluid [CSF] amyloid beta 1-40 [Aβ1-40] levels [primary endpoint] and effect on other AD biomarkers) of atabecestat were evaluated.

Results: In both populations, atabecestat was well tolerated and characterized by linear PK and high central nervous system penetrance of unbound drug. Atabecestat significantly reduced CSF Aβ1-40 levels from baseline at day 28 in both the 10-mg (67-68%) and 50-mg (87-90%) dose groups compared with placebo. For Caucasians with early AD, the least squares mean differences (95% CI) were - 69.37 (- 72.25; - 61.50) and - 92.74 (- 100.08; - 85.39), and for Japanese with preclinical AD, they were - 62.48 (- 78.32; - 46.64) and - 80.81 (- 96.13; - 65.49), respectively. PK/PD model simulations confirmed that once-daily 10 mg and 50 mg atabecestat can attain 60-70% and 90% Aβ1-40 reductions, respectively. The trend of the reduction was similar across the Aβ1-37, Aβ1-38, and Aβ1-42 fragments in both atabecestat dose groups, consistent with Aβ1-40. CSF amyloid precursor protein fragment (sAPPβ) levels declined from baseline, regardless of patient population, whereas CSF sAPPα levels increased compared with placebo. There were no relevant changes in either CSF total tau or phosphorylated tau 181P over a 4-week treatment period.

Conclusions: JNJ-54861911 at 10 and 50 mg daily doses after 4 weeks resulted in mean CSF Aβ1-40 reductions of 67% and up to 90% in both Caucasian and Japanese patients with early stage AD, confirming results in healthy elderly adults.

Trial registration: ALZ1005: ClinicalTrials.gov, NCT01978548. Registered on 7 November 2013. ALZ1008: ClinicalTrials.gov, NCT02360657. Registered on 10 February 2015.

Keywords: Alzheimer’s disease; Amyloid; Atabecestat; Aβ processing; BACE1 inhibitor; JNJ-54861911; PK/PD relationship.

Conflict of interest statement

Ethics approval and consent to participate

The study protocol and amendments were approved by the appropriate independent ethics committees/institutional review boards. Written informed consent was obtained from all participants before their participation. All study procedures followed were in accordance with current International Conference on Harmonization guidelines on good clinical practice, applicable regulatory and country-specific requirements, and the principles of the Declaration of Helsinki.

Consent for publication

Not applicable.

Competing interests

All authors, except ABH, NA, SE, JMG, MBa, and MBo, report receiving personal fees (employment) from Janssen Research & Development, a Division of Janssen Pharmaceutica N.V., Beerse, Belgium, or Janssen Research & Development, LLC, Raritan, NJ, USA, or Janssen Research and Development LLC, Titusville, NJ, USA, or Janssen Pharmaceutical K.K., Tokyo, Japan, and all own stock/stock options in the company. JRS is a former employee of Janssen Pharmaceutica NV and is currently affiliated with the University of Antwerp with a research advisory role at the Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium, and collects no consulting fees and receives no research funding. ABH is employed at Sahlgrenska University Hospital, Mölndal, Sweden, and has no disclosures to declare. NA is employed at Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Stockholm, Sweden, and has no disclosures to declare. SE is employed at the Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium, and at the Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium, and reports receiving research funding from Janssen Pharmaceutica N.V. and ADx Neurosciences (paid to institution). JMG is an employee of San Carlos Hospital Clinic, Universidad Complutense, Madrid, Spain, and receives salary as chief editor of a scientific journal indexed in Medline and Journal Citation Reports. MBa is an employee of Hospital Universitari i Politecnic La Fe, Valencia, Spain, and has no disclosures to declare. MBo is an employee of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain, and consults for Araclon, AstraZeneca, Grifols, Janssen, Kyowa Kirin, Lilly, MSD, Nutricia, Roche, Schwabe, and Servier. MBo has received fees for lectures from Araclon Biotech, Biogen, Grifols, Krka, Nutricia, Roche, and Schwabe, and reports receiving research funding from Araclon Biotech, Bioiberica, Grifols, Krka, Lilly, Merck, Piramal, Nutricia, Roche, Fundació Bancaria la Caixa, Carlos III Health Institute, the European Commission Horizon 2020 program, European Federation of Pharmaceutical Industries and Associations Innovative Medicines Initiative, and the European Research Area ERA-NET program.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Individual atabecestat (JNJ-54861911) pharmacokinetic (PK) parameters at steady state for AUC0–24 h (a) and maximum plasma concentration (Cmax) (b) vs. dose groups based on population PK model in Caucasian patients with early Alzheimer’s disease (ALZ1005) and in healthy elderly in the multiple ascending dose study (ALZ1002). Pharmacokinetic parameters were dose-normalized to 5 mg for all treatment and participant groups
Fig. 2
Fig. 2
Ratio of cerebrospinal fluid (CSF) and unbound plasma atabecestat (JNJ-54861911) concentration in Caucasian patients with preclinical Alzheimer’s disease (AD) and patients with mild cognitive impairment (MCI) due to AD (ALZ1005), by dose group
Fig. 3
Fig. 3
Percent change from baseline in cerebrospinal fluid (CSF) and plasma amyloid-β 1–40 (Aβ1–40) at day 28 (4 h postdose) across treatment groups for Caucasian patients with early Alzheimer’s disease (AD) and Japanese patients with preclinical AD. a Caucasian early AD and Japanese preclinical AD percent change from baseline in day 28 CSF Aβ1–40. b Caucasian early AD and Japanese preclinical AD percent change from baseline in day 28 plasma Aβ1–40
Fig. 4
Fig. 4
Observed day 28 cerebrospinal fluid (CSF) amyloid-β 1–40 (Aβ1–40) percentage reduction from baseline in Caucasian patients with preclinical Alzheimer’s disease (AD) and mild cognitive impairment (MCI) due to AD (ALZ1005) vs. model-based simulations from pharmacokinetic/pharmacodynamic modeling. The observed data of CSF Aβ1–40 percentage vs. baseline, stratified by patient population (preclinical AD vs. MCI due to AD) from Study ALZ1005, are overlaid on the model-predicted median and 90% prediction interval (5th and 95th percentiles, gray-shaded area) from 500 simulations per dose level
Fig. 5
Fig. 5
Day 28 mean change in computerized cognitive test battery elect from baseline across treatment groups in Caucasian patients with early Alzheimer’s disease (AD) (ALZ1005 safety population). a Paired associated learning (PAL) 6 pattern errors adjusted. b Reaction time (RTI) median five-choice reaction time. c Spatial working memory (SWM) between errors 4–8 boxes

References

    1. Citron M. Alzheimer’s disease: strategies for disease modification. Nat Rev Drug Discov. 2010;9:387–398. doi: 10.1038/nrd2896.
    1. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, et al. Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:280–292. doi: 10.1016/j.jalz.2011.03.003.
    1. Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, et al. Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science. 1999;286:735–741. doi: 10.1126/science.286.5440.735.
    1. Sinha S, Anderson JP, Barbour R, Basi GS, Caccavello R, Davis D, et al. Purification and cloning of amyloid precursor protein beta-secretase from human brain. Nature. 1999;402:537–540. doi: 10.1038/990114.
    1. Mulder SD, van der Flier WM, Verheijen JH, Mulder C, Scheltens P, Blankenstein MA, et al. BACE1 activity in cerebrospinal fluid and its relation to markers of AD pathology. J Alzheimers Dis. 2010;20:253–260. doi: 10.3233/JAD-2010-1367.
    1. Zetterberg H, Andreasson U, Hansson O, Wu G, Sankaranarayanan S, Andersson ME, et al. Elevated cerebrospinal fluid BACE1 activity in incipient Alzheimer disease. Arch Neurol. 2008;65:1102–1107. doi: 10.1001/archneur.65.8.1102.
    1. Zhong Z, Ewers M, Teipel S, Burger K, Wallin A, Blennow K, et al. Levels of beta-secretase (BACE1) in cerebrospinal fluid as a predictor of risk in mild cognitive impairment. Arch Gen Psychiatry. 2007;64:718–726. doi: 10.1001/archpsyc.64.6.718.
    1. Timmers M, Barao S, Van Broeck B, Tesseur I, Slemmon J, De Waepenaert K, et al. BACE1 dynamics upon inhibition with a BACE inhibitor and correlation to downstream Alzheimer’s disease markers in elderly healthy participants. J Alzheimers Dis. 2017;56:1437–1449. doi: 10.3233/JAD-160829.
    1. Timmers M, Van Broeck B, Ramael S, Slemmon J, De Waepenaert K, Russu A, Bogert JM, Stieltjes H, Shaw LM, Engelborghs S, Moechars D, Mercken M, Liu E, Sinha V, Kemp J, Van Nueten L, Tritsmans L, Streffer JR. Profiling the dynamics of CSF and plasma Aβ reduction after treatment with JNJ-54861911, a potent oral BACE inhibitor. Alzheimers Dement. 2016;2:202–212.
    1. Liu X, Wong H, Scearce-Levie K, Watts RJ, Coraggio M, Shin YG, et al. Mechanistic pharmacokinetic-pharmacodynamic modeling of BACE1 inhibition in monkeys: development of a predictive model for amyloid precursor protein processing. Drug Metab Dispos. 2013;41:1319–1328. doi: 10.1124/dmd.112.050864.
    1. Beal SL, Boeckmann AJ, Bauer RJ, editors. NONMEM 7.2.0 users guides. Ellicott City: Icon Development Solutions; 1989–2011.
    1. Timmerman P, White S, Dougall SM, Kall MA, Smeraglia J, Fjording MS, et al. Tiered approach into practice: scientific validation for chromatography-based assays in early development - a recommendation from the European bioanalysis forum. Bioanalysis 2015;7(18):2387–98.
    1. Struyfs H, Van Broeck B, Timmers M, Fransen E, Sleegers K, Van Broeckhoven C, et al. Diagnostic accuracy of cerebrospinal fluid amyloid-beta isoforms for early and differential dementia diagnosis. J Alzheimers Dis. 2015;45:813–822. doi: 10.3233/JAD-141986.
    1. Van Broeck B, Borgers M, Meulders G, Hermans B, Dhuyvetter D, Borghys H, Mercken M. Simultaneous evaluation of Aβ1-37/38/40/42 levels after treatment with secretase inhibitors and modulators using a novel immunoassay [abstract]. Neurodegener Dis. 2013;11(Suppl 1)
    1. Barao S, Zhou L, Adamczuk K, Vanhoutvin T, van Leuven F, Demedts D, et al. BACE1 levels correlate with phospho-tau levels in human cerebrospinal fluid. Curr Alzheimer Res. 2013;10:671–678. doi: 10.2174/15672050113109990138.
    1. Lewczuk P, Kamrowski-Kruck H, Peters O, Heuser I, Jessen F, Popp J, et al. Soluble amyloid precursor proteins in the cerebrospinal fluid as novel potential biomarkers of Alzheimer’s disease: a multicenter study. Mol Psychiatry. 2010;15:138–145. doi: 10.1038/mp.2008.84.
    1. Palmqvist S, Zetterberg H, Blennow K, Vestberg S, Andreasson U, Brooks DJ, et al. Accuracy of brain amyloid detection in clinical practice using cerebrospinal fluid beta-amyloid 42: a cross-validation study against amyloid positron emission tomography. JAMA Neurol. 2014;71:1282–1289. doi: 10.1001/jamaneurol.2014.1358.
    1. Donohue MC, Sperling RA, Petersen R, Sun CK, Weiner MW, Aisen PS, et al. Association between elevated brain amyloid and subsequent cognitive decline among cognitively normal persons. JAMA. 2017;317:2305–2316. doi: 10.1001/jama.2017.6669.
    1. Insel PS, Mattsson N, Mackin RS, Scholl M, Nosheny RL, Tosun D, et al. Accelerating rates of cognitive decline and imaging markers associated with beta-amyloid pathology. Neurology. 2016;86:1887–1896. doi: 10.1212/WNL.0000000000002683.
    1. Salloway S, Sperling R, Fox NC, Blennow K, Klunk W, Raskind M, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N Engl J Med. 2014;370:322–333. doi: 10.1056/NEJMoa1304839.
    1. Doody RS, Thomas RG, Farlow M, Iwatsubo T, Vellas B, Joffe S, et al. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N Engl J Med. 2014;370:311–321. doi: 10.1056/NEJMoa1312889.
    1. Yan R. Stepping closer to treating Alzheimer’s disease patients with BACE1 inhibitor drugs. Transl Neurodegener. 2016;5:13. doi: 10.1186/s40035-016-0061-5.
    1. Egan MF, Kost J, Tariot PN, Aisen PS, Cummings JL, Vellas B, et al. Randomized Trial of Verubecestat for Mild-to-Moderate Alzheimer's Disease. N Engl J Med. 2018;378:1691–703.

Source: PubMed

Подписаться