17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

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

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          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 [Aβ 1–40] 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 Aβ 1–40 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% Aβ 1–40 reductions, respectively. The trend of the reduction was similar across the Aβ 1–37, Aβ 1–38, and Aβ 1–42 fragments in both atabecestat dose groups, consistent with Aβ 1–40. 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 Aβ 1–40 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.

          Electronic supplementary material

          The online version of this article (10.1186/s13195-018-0415-6) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references3

          • Record: found
          • Abstract: found
          • Article: not found

          Alzheimer's disease: strategies for disease modification.

          Alzheimer's disease is the largest unmet medical need in neurology. Current drugs improve symptoms, but do not have profound disease-modifying effects. However, in recent years, several approaches aimed at inhibiting disease progression have advanced to clinical trials. Among these, strategies targeting the production and clearance of the amyloid-beta peptide - a cardinal feature of Alzheimer's disease that is thought to be important in disease pathogenesis - are the most advanced. Approaches aimed at modulating the abnormal aggregation of tau filaments (another key feature of the disease), and those targeting metabolic dysfunction, are also being evaluated in the clinic. This article discusses recent progress with each of these strategies, with a focus on anti-amyloid strategies, highlighting the lessons learned and the challenges that remain.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Accelerating rates of cognitive decline and imaging markers associated with β-amyloid pathology.

            To estimate points along the spectrum of β-amyloid pathology at which rates of change of several measures of neuronal injury and cognitive decline begin to accelerate.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mechanistic pharmacokinetic-pharmacodynamic modeling of BACE1 inhibition in monkeys: development of a predictive model for amyloid precursor protein processing.

              This study was conducted to determine the pharmacokinetics (PK) and pharmacodynamics (PD) of two novel inhibitors of β-site amyloid precursor protein (APP)-cleaving enzyme (BACE1), GNE-629 [(4S,4a'S,10a'S)-2-amino-8'-(2-fluoropyridin-3-yl)-1-methyl-3',4',4a',10a'-tetrahydro-1'H-spiro[imidazole-4,10'-pyrano[4,3-b]chromen]-5(1H)-one] and GNE-892 [(R)-2-amino-1,3',3'-trimethyl-7'-(pyrimidin-5-yl)-3',4'-dihydro-2'H-spiro[imidazole-4,1'-naphthalen]-5(1H)-one], and to develop a PK-PD model to predict in vivo effects based solely on in vitro activity and PK. GNE-629 and GNE-892 concentrations and PD biomarkers including amyloid β (Aβ) in the plasma and cerebrospinal fluid (CSF), and secreted APPβ (sAPPβ) and secreted APPα (sAPPα) in the CSF were measured after a single oral administration of GNE-629 (100 mg/kg) or GNE-892 (30 or 100 mg/kg) in cynomolgus monkeys. A mechanistic PK-PD model was developed to simultaneously characterize the plasma Aβ and CSF Aβ, sAPPα, and sAPPβ using GNE-629 in vivo data. This model was used to predict the in vivo effects of GNE-892 after adjustments based on differences in in vitro cellular activity and PK. The PK-PD model estimated GNE-629 CSF and free plasma IC₅₀ of 0.0033 μM and 0.065 μM, respectively. These differences in CSF and free plasma IC₅₀ suggest that different mechanisms are involved in Aβ formation in these two compartments. The predicted in vivo effects for GNE-892 using the PK-PD model were consistent with the observed data. In conclusion, a PK-PD model was developed to mechanistically describe the effects of BACE1 inhibition on Aβ, sAPPβ, and sAPPα in the CSF, and Aβ in the plasma. This model can be used to prospectively predict in vivo effects of new BACE1 inhibitors using just their in vitro activity and PK data.
                Bookmark

                Author and article information

                Contributors
                +32 14 60 8122 , mtimmer3@its.jnj.com
                johannes.streffer@uantwerpen.be , johannes.streffer@ucb.com
                arussu@its.jnj.com
                ytominag@its.jnj.com
                hshimizu@its.jnj.com
                ktatikol@its.jnj.com
                psmekens@its.jnj.com
                anne.borjesson@neuro.gu.se
                niels.andreasen@karolinska.se
                matiasguiu@gmail.com
                miquelbaquero@gmail.com
                mboada@fundacioace.com
                itesseur@its.jnj.com
                ltritsm1@its.jnj.com
                lvnueten@its.jnj.com
                sebastiaan.engelborghs@uantwerpen.be
                Journal
                Alzheimers Res Ther
                Alzheimers Res Ther
                Alzheimer's Research & Therapy
                BioMed Central (London )
                1758-9193
                23 August 2018
                23 August 2018
                2018
                : 10
                : 85
                Affiliations
                [1 ]ISNI 0000 0004 0623 0341, GRID grid.419619.2, Janssen Research and Development, , a Division of Janssen Pharmaceutica NV, ; Turnhoutseweg 30, 2340 Beerse, Belgium
                [2 ]ISNI 0000 0001 0790 3681, GRID grid.5284.b, Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, , University of Antwerp, ; Antwerp, Belgium
                [3 ]Janssen Pharmaceutical K.K, Tokyo, Japan
                [4 ]GRID grid.417429.d, Janssen Research and Development LLC, ; Raritan, NJ USA
                [5 ]ISNI 000000009445082X, GRID grid.1649.a, Sahlgrenska University Hospital, ; Mölndal, Sweden
                [6 ]ISNI 0000 0004 1937 0626, GRID grid.4714.6, Department Neurobiology, Care Sciences & Society, Center for Alzheimer Research, Division of Neurogeriatrics, , Karolinska Institutet, ; Stockholm, Sweden
                [7 ]ISNI 0000 0001 2157 7667, GRID grid.4795.f, Hospital Clinic San Carlos, Universidad Complutense, ; Madrid, Spain
                [8 ]ISNI 0000 0001 0360 9602, GRID grid.84393.35, Neurology Department, , Hospital Universitari I Politecnic La Fe, ; Valencia, Spain
                [9 ]ISNI 0000 0004 1765 5601, GRID grid.477255.6, Fundació ACE, Institut Català de Neurociències Aplicades, ; Barcelona, Spain
                [10 ]ISNI 0000 0004 0608 3935, GRID grid.416667.4, Department of Neurology and Memory Clinic, , Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, ; Antwerp, Belgium
                [11 ]GRID grid.421932.f, Present address: Head of Translational Medicine Neuroscience, UCB Biopharma SPRL, ; Chemin du Foriest, B-1420 Braine-l’Alleud, Belgium
                Article
                415
                10.1186/s13195-018-0415-6
                6106931
                30134967
                40d77551-3d01-4de0-a771-cd269bb16fe4
                © The Author(s). 2018

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 29 March 2018
                : 27 July 2018
                Funding
                Funded by: Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
                Funded by: Janssen Pharmaceutical K.K. Japan
                Funded by: Flanders Innovation & Entrepreneurship (VLAIO), Belgium
                Award ID: IWT 120834
                Award Recipient :
                Categories
                Research
                Custom metadata
                © The Author(s) 2018

                Neurology
                atabecestat,jnj-54861911,bace1 inhibitor,alzheimer’s disease,amyloid,aβ processing,pk/pd relationship

                Comments

                Comment on this article