20
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Safety, tolerability and efficacy of the glutaminyl cyclase inhibitor PQ912 in Alzheimer’s disease: results of a randomized, double-blind, placebo-controlled phase 2a 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

          PQ912 is an inhibitor of the glutaminyl cyclase enzyme that plays a central role in the formation of synaptotoxic pyroglutamate-A-beta oligomers. We report on the first clinical study with PQ912 in subjects with biomarker-proven Alzheimer’s disease (AD). The aim was to determine the maximal tolerated dose, target occupancy and treatment-related pharmacodynamic effects. The exploratory efficacy readouts selected were tailored to the patient population with early AD. The therapeutic approach focuses on synaptic dysfunction as captured by various measures such as electroencephalography (EEG), synaptic biomarkers and sensitive cognitive tests.

          Methods

          This was a randomized, double-blind, placebo-controlled trial evaluating the safety, tolerability and efficacy of PQ912 800 mg twice daily (bid) for 12 weeks in subjects with mild cognitive impairment or mild dementia due to AD. The 120 enrolled subjects were treatment-naïve at the start of the study, had confirmed AD biomarkers in their cerebrospinal fluid at screening and had a Mini Mental State Examination score between 21 and 30. After 1 week of treatment with 400 mg bid, patients were up-titrated to 800 mg bid for 11 weeks. Patients were randomized 1:1 to either PQ912 or placebo. The primary composite endpoints were to assess safety and tolerability based on the number of patients who discontinued due to (serious) adverse events (safety), and based on dose adjustment during the treatment period and/or nonadherence to randomized treatment (tolerability). All randomized subjects who took at least one dose of the study treatment or placebo were used for safety analyses.

          Results

          There was no significant difference between treatments in the number of subjects with (serious) adverse events, although there were slightly more patients with a serious adverse event in the PQ912 group compared to placebo. More subjects treated with PQ912 discontinued treatment due to adverse events, mostly related to gastrointestinal and skin/subcutaneous tissue disorders. PQ912 treatment resulted in a significant reduction in glutaminyl cyclase activity, which resulted in an average target occupancy of > 90%. A significant reduction of theta power in the EEG frequency analysis and a significant improvement in the One Back test of our Neuropsychological Test Battery was observed. The exploratory biomarker readouts, neurogranin for synaptic toxicity and YKL-40 as a marker of inflammation, appear to be sensitive enough to serve as efficacy markers in the next phase 2b study.

          Conclusions

          The maximal tolerated dose of PQ912 has been identified and the results support future studies at still lower doses reaching > 50% target occupancy, a longer up-titration phase to potentially induce adaptation and longer treatment periods to confirm the early signals of efficacy as seen in this study.

          Trial registration

          Clinicaltrials.gov, NCT 02389413. Registered on 17 March 2015.

          Electronic supplementary material

          The online version of this article (10.1186/s13195-018-0431-6) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references18

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

          Phase lag index: assessment of functional connectivity from multi channel EEG and MEG with diminished bias from common sources.

          To address the problem of volume conduction and active reference electrodes in the assessment of functional connectivity, we propose a novel measure to quantify phase synchronization, the phase lag index (PLI), and compare its performance to the well-known phase coherence (PC), and to the imaginary component of coherency (IC). The PLI is a measure of the asymmetry of the distribution of phase differences between two signals. The performance of PLI, PC, and IC was examined in (i) a model of 64 globally coupled oscillators, (ii) an EEG with an absence seizure, (iii) an EEG data set of 15 Alzheimer patients and 13 control subjects, and (iv) two MEG data sets. PLI and PC were more sensitive than IC to increasing levels of true synchronization in the model. PC and IC were influenced stronger than PLI by spurious correlations because of common sources. All measures detected changes in synchronization during the absence seizure. In contrast to PC, PLI and IC were barely changed by the choice of different montages. PLI and IC were superior to PC in detecting changes in beta band connectivity in AD patients. Finally, PLI and IC revealed a different spatial pattern of functional connectivity in MEG data than PC. The PLI performed at least as well as the PC in detecting true changes in synchronization in model and real data but, at the same token and like-wise the IC, it was much less affected by the influence of common sources and active reference electrodes. Copyright 2007 Wiley-Liss, Inc.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            EEG dynamics in patients with Alzheimer's disease.

            Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by cognitive and intellectual deficits and behavior disturbance. The electroencephalogram (EEG) has been used as a tool for diagnosing AD for several decades. The hallmark of EEG abnormalities in AD patients is a shift of the power spectrum to lower frequencies and a decrease in coherence of fast rhythms. These abnormalities are thought to be associated with functional disconnections among cortical areas resulting from death of cortical neurons, axonal pathology, cholinergic deficits, etc. This article reviews main findings of EEG abnormalities in AD patients obtained from conventional spectral analysis and nonlinear dynamical methods. In particular, nonlinear alterations in the EEG of AD patients, i.e. a decreased complexity of EEG patterns and reduced information transmission among cortical areas, and their clinical implications are discussed. For future studies, improvement of the accuracy of differential diagnosis and early detection of AD based on multimodal approaches, longitudinal studies on nonlinear dynamics of the EEG, drug effects on the EEG dynamics, and linear and nonlinear functional connectivity among cortical regions in AD are proposed to be investigated. EEG abnormalities of AD patients are characterized by slowed mean frequency, less complex activity, and reduced coherences among cortical regions. These abnormalities suggest that the EEG has utility as a valuable tool for differential and early diagnosis of AD.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              2015 Alzheimer's disease facts and figures.

              (2015)
              This report discusses the public health impact of Alzheimer’s disease (AD), including incidence and prevalence, mortality rates, costs of care and the overall effect on caregivers and society. It also examines the challenges encountered by health care providers when disclosing an AD diagnosis to patients and caregivers. An estimated 5.3 million Americans have AD; 5.1 million are age 65 years, and approximately 200,000 are age <65 years and have younger onset AD. By mid-century, the number of people living with AD in the United States is projected to grow by nearly 10 million, fueled in large part by the aging baby boom generation. Today, someone in the country develops AD every 67 seconds. By 2050, one new case of AD is expected to develop every 33 seconds, resulting in nearly 1 million new cases per year, and the estimated prevalence is expected to range from 11 million to 16 million. In 2013, official death certificates recorded 84,767 deaths from AD, making AD the sixth leading cause of death in the United States and the fifth leading cause of death in Americans age 65 years. Between 2000 and 2013, deaths resulting from heart disease, stroke and prostate cancer decreased 14%, 23% and 11%, respectively, whereas deaths from AD increased 71%. The actual number of deaths to which AD contributes (or deaths with AD) is likely much larger than the number of deaths from AD recorded on death certificates. In 2015, an estimated 700,000 Americans age 65 years will die with AD, and many of them will die from complications caused by AD. In 2014, more than 15 million family members and other unpaid caregivers provided an estimated 17.9 billion hours of care to people with AD and other dementias, a contribution valued at more than $217 billion. Average per-person Medicare payments for services to beneficiaries age 65 years with AD and other dementias are more than two and a half times as great as payments for all beneficiaries without these conditions, and Medicaid payments are 19 times as great. Total payments in 2015 for health care, long-term care and hospice services for people age 65 years with dementia are expected to be $226 billion. Among people with a diagnosis of AD or another dementia, fewer than half report having been told of the diagnosis by their health care provider. Though the benefits of a prompt, clear and accurate disclosure of an AD diagnosis are recognized by the medical profession, improvements to the disclosure process are needed. These improvements may require stronger support systems for health care providers and their patients.
                Bookmark

                Author and article information

                Contributors
                p.scheltens@vumc.nl
                Merja.Hallikainen@kuh.fi
                t.grimmer@tum.de
                Thomas.Duning@ukmuenster.de
                AA.Gouw@vumc.nl
                c.teunissen@vumc.nl
                a.wink@vumc.nl
                pmaruff@cogstate.com
                johncpc@btinternet.com
                G.C.M.vanBaal@umcutrecht.nl
                suzanne.bruins@probiodrug.de
                inge.lues@probiodrug.de
                n.Prins@brainresearchcenter.nl
                Journal
                Alzheimers Res Ther
                Alzheimers Res Ther
                Alzheimer's Research & Therapy
                BioMed Central (London )
                1758-9193
                12 October 2018
                12 October 2018
                2018
                : 10
                : 107
                Affiliations
                [1 ]ISNI 0000 0004 0435 165X, GRID grid.16872.3a, Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, , VU University Medical Center, ; Amsterdam, The Netherlands
                [2 ]ISNI 0000 0001 0726 2490, GRID grid.9668.1, University of Eastern Finland, Institute of Clinical Medicine, ; Kuopio, Finland
                [3 ]ISNI 0000000123222966, GRID grid.6936.a, Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, , Technische Universität München, ; Munich, Germany
                [4 ]ISNI 0000 0001 2172 9288, GRID grid.5949.1, Department of Neurology, , University of Münster, ; Münster, Germany
                [5 ]ISNI 0000 0004 0435 165X, GRID grid.16872.3a, Department of Clinical Neurophysiology and MEG Center, Amsterdam Neuroscience, , VU University Medical Center, ; Amsterdam, The Netherlands
                [6 ]ISNI 0000 0004 0435 165X, GRID grid.16872.3a, Department of Clinical Chemistry, Neurochemistry Laboratory and Biobank, Amsterdam Neuroscience, , VU University Medical Center, ; Amsterdam, The Netherlands
                [7 ]ISNI 0000 0004 0435 165X, GRID grid.16872.3a, Department of Radiology, Nuclear Medicine and PET Research, Amsterdam Neuroscience, , VU University Medical Center, ; Amsterdam, The Netherlands
                [8 ]Cogstate Ltd, Melbourne, Australia
                [9 ]ISNI 0000 0001 2322 6764, GRID grid.13097.3c, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, ; London, UK
                [10 ]ISNI 0000000090126352, GRID grid.7692.a, Julius Center for Health Sciences and Primary Care, UMC Utrecht, ; Utrecht, The Netherlands
                [11 ]GRID grid.435222.0, Probiodrug AG, ; Halle, Germany
                [12 ]Brain Research Center, Amsterdam, The Netherlands
                Author information
                http://orcid.org/0000-0002-1046-6408
                Article
                431
                10.1186/s13195-018-0431-6
                6182869
                30309389
                25615717-98ec-4948-8d9c-a4ef4a9b7144
                © 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
                : 15 May 2018
                : 10 September 2018
                Funding
                Funded by: Probiodrug AG
                Categories
                Research
                Custom metadata
                © The Author(s) 2018

                Neurology
                alzheimer’s disease,glutaminyl cyclase inhibitor,pq912,phase 2a
                Neurology
                alzheimer’s disease, glutaminyl cyclase inhibitor, pq912, phase 2a

                Comments

                Comment on this article