Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Inge Lues is active.

Publication


Featured researches published by Inge Lues.


Neurobiology of Aging | 2015

An anti-pyroglutamate-3 Aβ vaccine reduces plaques and improves cognition in APPswe/PS1ΔE9 mice

Jeffrey L. Frost; Bin Liu; Jens-Ulrich Rahfeld; Martin Kleinschmidt; Brian O'Nuallain; Kevin X. Le; Inge Lues; Stephan Schilling; Hans-Ulrich Demuth; Cynthia A. Lemere

Pyroglutamate-3 amyloid-beta (pGlu-3 Aβ) is an N-terminally truncated Aβ isoform likely playing a decisive role in Alzheimers disease pathogenesis. Here, we describe a prophylactic passive immunization study in APPswe/PS1ΔE9 mice using a novel pGlu-3 Aβ immunoglobulin G1 (IgG1) monoclonal antibody, 07/1 (150 and 500 μg, intraperitoneal, weekly) and compare its efficacy with a general Aβ IgG1 monoclonal antibody, 3A1 (200 μg, intraperitoneal, weekly) as a positive control. After 28 weeks of treatment, plaque burden was reduced and cognitive performance of 07/1-immunized Tg mice, especially at the higher dose, was normalized to wild-type levels in 2 hippocampal-dependent tests and partially spared compared with phosphate-buffered saline-treated Tg mice. Mice that received 3A1 had reduced plaque burden but showed no cognitive benefit. In contrast with 3A1, treatment with 07/1 did not increase the concentration of Aβ in plasma, suggesting different modes of Aβ plaque clearance. In conclusion, early selective targeting of pGlu-3 Aβ by immunotherapy may be effective in lowering cerebral Aβ plaque burden and preventing cognitive decline in the clinical setting. Targeting this pathologically modified form of Aβ thereby is unlikely to interfere with potential physiologic function(s) of Aβ that have been proposed.


Journal of Pharmacology and Experimental Therapeutics | 2017

Glutaminyl Cyclase Inhibitor PQ912 improves cognition in mouse models of Alzheimer's disease - studies on relation to effective target occupancy

Torsten Hoffmann; Antje Meyer; Ulrich Heiser; Stephan Kurat; Livia Böhme; Martin Kleinschmidt; Karl-Ulrich Bühring; Birgit Hutter-Paier; Martina Farcher; Hans-Ulrich Demuth; Inge Lues; Stephan Schilling

Numerous studies suggest that the majority of amyloid-β (Aβ) peptides deposited in Alzheimer’s disease (AD) are truncated and post-translationally modified at the N terminus. Among these modified species, pyroglutamyl-Aβ (pE-Aβ, including N3pE-Aβ40/42 and N11pE-Aβ40/42) has been identified as particularly neurotoxic. The N-terminal modification renders the peptide hydrophobic, accelerates formation of oligomers, and reduces degradation by peptidases, leading ultimately to the accumulation of the peptide and progression of AD. It has been shown that the formation of pyroglutamyl residues is catalyzed by glutaminyl cyclase (QC). Here, we present data about the pharmacological in vitro and in vivo efficacy of the QC inhibitor (S)-1-(1H-benzo[d]imidazol-5-yl)-5-(4-propoxyphenyl)imidazolidin-2-one (PQ912), the first-in-class compound that is in clinical development. PQ912 inhibits human, rat, and mouse QC activity, with Ki values ranging between 20 and 65 nM. Chronic oral treatment of hAPPSLxhQC double-transgenic mice with approximately 200 mg/kg/day via chow shows a significant reduction of pE-Aβ levels and concomitant improvement of spatial learning in a Morris water maze test paradigm. This dose results in a brain and cerebrospinal fluid concentration of PQ912 which relates to a QC target occupancy of about 60%. Thus, we conclude that >50% inhibition of QC activity in the brain leads to robust treatment effects. Secondary pharmacology experiments in mice indicate a fairly large potency difference for Aβ cyclization compared with cyclization of physiologic substrates, suggesting a robust therapeutic window in humans. This information constitutes an important translational guidance for predicting the therapeutic dose range in clinical studies with PQ912.


Alzheimer's & Dementia: Translational Research & Clinical Interventions | 2015

A phase 1 study to evaluate the safety and pharmacokinetics of PQ912, a glutaminyl cyclase inhibitor, in healthy subjects

Inge Lues; Frank Weber; Antje Meyer; Uli Bühring; Torsten Hoffmann; Kerstin Kühn-Wache; Susanne Manhart; Ulrich Heiser; Rolf Pokorny; Joseph Chiesa; Konrad Glund

Pyroglutamate‐amyloid‐β (pE‐Aβ) peptides are major components of Aβ‐oligomers and Aβ‐plaques, which are regarded as key culprits of Alzheimers disease (AD) pathology. PQ912 is a competitive inhibitor of the enzyme glutaminyl cyclase (QC), essential for the formation of pE‐Aβ peptides.


Journal of Biological Chemistry | 2017

Structural and functional analyses of pyroglutamate-amyloid-β-specific antibodies as a basis for Alzheimer immunotherapy

Anke Piechotta; Christoph Parthier; Martin Kleinschmidt; Kathrin Gnoth; Thierry Pillot; Inge Lues; Hans-Ulrich Demuth; Stephan Schilling; Jens-Ulrich Rahfeld; Milton T. Stubbs

Alzheimer disease is associated with deposition of the amyloidogenic peptide Aβ in the brain. Passive immunization using Aβ-specific antibodies has been demonstrated to reduce amyloid deposition both in vitro and in vivo. Because N-terminally truncated pyroglutamate (pE)-modified Aβ species (AβpE3) exhibit enhanced aggregation potential and propensity to form toxic oligomers, they represent particularly attractive targets for antibody therapy. Here we present three separate monoclonal antibodies that specifically recognize AβpE3 with affinities of 1–10 nm and inhibit AβpE3 fibril formation in vitro. In vivo application of one of these resulted in improved memory in AβpE3 oligomer-treated mice. Crystal structures of Fab-AβpE3 complexes revealed two distinct binding modes for the peptide. Juxtaposition of pyroglutamate pE3 and the F4 side chain (the “pEF head”) confers a pronounced bulky hydrophobic nature to the AβpE3 N terminus that might explain the enhanced aggregation properties of the modified peptide. The deep burial of the pEF head by two of the antibodies explains their high target specificity and low cross-reactivity, making them promising candidates for the development of clinical antibodies.


Alzheimer's Research & Therapy | 2017

Glutaminyl cyclase activity correlates with levels of Aβ peptides and mediators of angiogenesis in cerebrospinal fluid of Alzheimer’s disease patients

Claire Bridel; Torsten Hoffmann; Antje Meyer; Sisi Durieux; Marleen Koel-Simmelink; Matthias Orth; Philip Scheltens; Inge Lues; Charlotte E. Teunissen

BackgroundPyroglutamylation of truncated Aβ peptides, which is catalysed by enzyme glutaminyl cyclase (QC), generates pE-Aβ species with enhanced aggregation propensities and resistance to most amino-peptidases and endo-peptidases. pE-Aβ species have been identified as major constituents of Aβ plaques and reduction of pE-Aβ species is associated with improvement of cognitive tasks in animal models of Alzheimer’s disease (AD). Pharmacological inhibition of QC has thus emerged as a promising therapeutic approach for AD. Here, we question whether cerebrospinal fluid (CSF) QC enzymatic activity differs between AD patients and controls and whether inflammatory or angiogenesis mediators, some of which are potential QC substrates, and/or Aβ peptides may serve as pharmacodynamic read-outs for QC inhibition.MethodsQC activity, Aβ peptides and inflammatory or angiogenesis mediators were measured in CSF of a clinically well-characterized cohort of 20 mild AD patients, 20 moderate AD patients and 20 subjective memory complaints (SMC) controls. Correlation of these parameters with core diagnostic CSF AD biomarkers (Aβ42, tau and p-tau) and clinical features was evaluated.ResultsQC activity shows a tendency to decrease with AD progression (p = 0.129). The addition of QC activity to biomarkers tau and p-tau significantly increases diagnostic power (ROC-AUCTAU = 0.878, ROC-AUCTAU&QC = 0.939 and ROC-AUCpTAU = 0.820, ROC-AUCpTAU&QC = 0.948). In AD and controls, QC activity correlates with Aβ38 (r = 0.83, p < 0.0001) and Aβ40 (r = 0.84, p < 0.0001), angiogenesis mediators (Flt1, Tie2, VEGFD, VCAM-1 and ICAM-1, r > 0.5, p < 0.0001) and core diagnostic biomarkers (r > 0.35, p = <0.0057). QC activity does not correlate with MMSE or ApoE genotype.ConclusionsAβ38, Aβ40 and angiogenesis mediators (Flt1, Tie2, VEGFD, VCAM-1 and ICAM-1) are potential pharmacodynamic markers of QC inhibition, because their levels closely correlate with QC activity in AD patients. The addition of QC activity to core diagnostic CSF biomarkers may be of specific interest in clinical cases with discordant imaging and biochemical biomarker results.


Alzheimers & Dementia | 2014

TOXIC PGLU-ABETA IS ENHANCED AND GLUTAMINYL CYCLASE (QC) UP-REGULATED EARLY IN ALZHEIMER'S DISEASE (AD): INHIBITORS OF QC BLOCKING PGLU-ABETA FORMATION ARE IN CLINICAL DEVELOPMENT

Hans-Ulrich Demuth; Stephan Schilling; Steffen Roßner; Markus Morawski; Maike Hartlage-Rübsamen; Inge Lues; Konrad Glund

O1-10-02 TOXIC PGLU-ABETA IS ENHANCED AND GLUTAMINYL CYCLASE (QC) UP-REGULATED EARLY IN ALZHEIMER’S DISEASE (AD): INHIBITORS OF QC BLOCKING PGLU-ABETA FORMATION ARE IN CLINICAL DEVELOPMENT Hans-Ulrich Demuth, Stephan Schilling, Steffen Roßner, Markus Morawski, Maike Hartlage-R€ubsamen, Inge Lues, Konrad Glund, Probiodrug AG, Halle (Saale), Germany; University of Leipzig, Leipzig, Germany. Contact e-mail: hans-ulrich.demuth@ probiodrug.de


Molecules | 2018

Passive Aβ Immunotherapy: Current Achievements and Future Perspectives

Stephan Schilling; Jens-Ulrich Rahfeld; Inge Lues; Cynthia A. Lemere

Passive immunotherapy has emerged as a very promising approach for the treatment of Alzheimer’s disease and other neurodegenerative disorders, which are characterized by the misfolding and deposition of amyloid peptides. On the basis of the amyloid hypothesis, the majority of antibodies in clinical development are directed against amyloid β (Aβ), the primary amyloid component in extracellular plaques. This review focuses on the current status of Aβ antibodies in clinical development, including their characteristics and challenges that came up in clinical trials with these new biological entities (NBEs). Emphasis is placed on the current view of common side effects observed with passive immunotherapy, so-called amyloid-related imaging abnormalities (ARIAs), and potential ways to overcome this issue. Among these new ideas, a special focus is placed on molecules that are directed against post-translationally modified variants of the Aβ peptide, an emerging approach for development of new antibody molecules.


Molecules | 2018

Immunohistochemical Evidence from APP-Transgenic Mice for Glutaminyl Cyclase as Drug Target to Diminish pE-Abeta Formation

Maike Hartlage-Rübsamen; Alexandra Bluhm; Anke Piechotta; Miriam Linnert; Jens-Ulrich Rahfeld; Hans-Ulrich Demuth; Inge Lues; Peer-Hendrik Kuhn; Stefan F. Lichtenthaler; Steffen Roßner; Corinna Höfling

Oligomeric assemblies of neurotoxic amyloid beta (Abeta) peptides generated by proteolytical processing of the amyloid precursor protein (APP) play a key role in the pathogenesis of Alzheimer’s disease (AD). In recent years, a substantial heterogeneity of Abeta peptides with distinct biophysical and cell biological properties has been demonstrated. Among these, a particularly neurotoxic and disease-specific Abeta variant is N-terminally truncated and modified to pyroglutamate (pE-Abeta). Cell biological and animal experimental studies imply the catalysis of this modification by the enzyme glutaminyl cyclase (QC). However, direct histopathological evidence in transgenic animals from comparative brain region and cell type-specific expression of transgenic hAPP and QC, on the one hand, and on the formation of pE-Abeta aggregates, on the other, is lacking. Here, using single light microscopic, as well as triple immunofluorescent, labeling, we report the deposition of pE-Abeta only in the brain regions of APP-transgenic Tg2576 mice with detectable human APP and endogenous QC expression, such as the hippocampus, piriform cortex, and amygdala. Brain regions showing human APP expression without the concomitant presence of QC (the anterodorsal thalamic nucleus and perifornical nucleus) do not display pE-Abeta plaque formation. However, we also identified brain regions with substantial expression of human APP and QC in the absence of pE-Abeta deposition (the Edinger-Westphal nucleus and locus coeruleus). In these brain regions, the enzymes required to generate N-truncated Abeta peptides as substrates for QC might be lacking. Our observations provide additional evidence for an involvement of QC in AD pathogenesis via QC-catalyzed pE-Abeta formation.


Alzheimers & Dementia | 2018

COMBINATION OF A GLUTAMINYL CYCLASE INHIBITOR (PQ912) AND A PYROGLUTAMATE-Aβ SPECIFIC ANTIBODY (PBD-M06) SHOWS ADDITIVE EFFECTS IN A MOUSE MODEL WITH ALZHEIMER’S DISEASE-LIKE PATHOLOGY

Torsten Hoffmann; Martina Farcher; Birgit Hutter-Paier; Cynthia A. Lemere; Stephan Schilling; Inge Lues

P1-099 COMBINATION OFA GLUTAMINYL CYCLASE INHIBITOR (PQ912) AND A PYROGLUTAMATE-Ab SPECIFIC ANTIBODY (PBD-M06) SHOWS ADDITIVE EFFECTS IN A MOUSE MODELWITH ALZHEIMER’S DISEASE-LIKE PATHOLOGY Torsten Hoffmann, Martina Farcher, Birgit Hutter-Paier, Cynthia A. Lemere, Stephan Schilling, Inge Lues, Probiodrug, Halle, Germany; QPS Austria, Grambach, Austria; QPS Austria GmbH, Grambach, Austria; Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA; Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany. Contact e-mail: stephan.schilling@izi. fraunhofer.de


Alzheimers & Dementia | 2013

Safety, pharmacokinetics and pharmacodynamics of PQ912, the first glutaminyl cyclase (QC) inhibitor to treat Alzheimer's disease, in healthy elderly

Ronald S. Black; Inge Lues; Frank Weber; Antje Meyer; Torsten Hoffman; Rolf Pokorny; Hans-Ulrich Demuth; Konrad Glund

Collaboration


Dive into the Inge Lues's collaboration.

Top Co-Authors

Avatar

Torsten Hoffmann

Martin Luther University of Halle-Wittenberg

View shared research outputs
Top Co-Authors

Avatar

Cynthia A. Lemere

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bin Liu

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Brian O'Nuallain

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jeffrey L. Frost

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Kevin X. Le

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philip Scheltens

VU University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge