Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Eur J Pharm Sci ; 156: 105581, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33035662

ABSTRACT

Amyloid-ß (Aß) plays a central role in the development and progression of Alzheimer's disease (AD) with Aß oligomers representing the most toxic species. The all-d-enantiomeric peptide RD2, which recently successfully completed clinical phase I, specifically eliminates Aß oligomers in vitro as well as in vivo and improves cognitive deficits in various transgenic AD mouse models even after oral administration. To further enhance the oral absorption of RD2, folic acid has been conjugated to the d-peptide promoting an endocytosis-mediated uptake via a folate receptor located in the intestine. Two different conjugation strategies were selected to obtain prodrugs with folic acid being cleaved after intestinal absorption releasing unmodified RD2 in order to enable RD2's unaltered systemic efficacy. Both conjugates remained stable in simulated gastrointestinal fluids. But only one of them was suitable as prodrug as it was cleaved to RD2 in vitro in human blood plasma and liver microsomes and in vivo in mice after intravenous injection leading to a systemic release of RD2. Furthermore, the conjugate's permeability in vitro and after oral administration in mice was strongly enhanced compared to unconjugated RD2 demonstrating the prodrug's functionality. However, the conjugate seemed to have impaired the mice's wellbeing shortly after oral administration possibly resulting from strain-specific hypersensitivity to folic acid. Nevertheless, we assume that the prodrug is actually non-toxic, especially in lower concentrations as verified by a cell viability test. Furthermore, lower dosages can be applied with unaltered efficacy due to its enhanced oral absorption.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/drug therapy , Animals , Folic Acid , Mice , Mice, Transgenic , Stereoisomerism
2.
Mol Neurobiol ; 56(3): 2211-2223, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30003517

ABSTRACT

Oligomers of the amyloid-ß (Aß) protein are suspected to be responsible for the development and progression of Alzheimer's disease. Thus, the development of compounds that are able to eliminate already formed toxic Aß oligomers is very desirable. Here, we describe the in vivo efficacy of the compound RD2, which was developed to directly and specifically eliminate toxic Aß oligomers. In a truly therapeutic, rather than a preventive study, oral treatment with RD2 was able to reverse cognitive deficits and significantly reduce Aß pathology in old-aged transgenic Alzheimer's Disease mice with full-blown pathology and behavioral deficits. For the first time, we demonstrate the in vivo target engagement of RD2 by showing a significant reduction of Aß oligomers in the brains of RD2-treated mice compared to placebo-treated mice. The correlation of Aß elimination in vivo and the reversal of cognitive deficits in old-aged transgenic mice support the hypothesis that Aß oligomers are relevant not only for disease development and progression, but also offer a promising target for the causal treatment of Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Brain/drug effects , Cognition/drug effects , Peptides/therapeutic use , Spatial Learning/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/metabolism , Cognition/physiology , Male , Mice , Mice, Transgenic , Peptides/pharmacology , Spatial Learning/physiology
3.
Neurobiol Dis ; 124: 36-45, 2019 04.
Article in English | MEDLINE | ID: mdl-30391539

ABSTRACT

Alzheimer's disease, a multifactorial incurable disorder, is mainly characterised by progressive neurodegeneration, extracellular accumulation of amyloid-ß protein (Aß), and intracellular aggregation of hyperphosphorylated tau protein. During the last years, Aß oligomers have been claimed to be the disease causing agent. Consequently, development of compounds that are able to disrupt already existing Aß oligomers is highly desirable. We developed d-enantiomeric peptides, consisting solely of d-enantiomeric amino acid residues, for the direct and specific elimination of toxic Aß oligomers. The drug candidate RD2 did show high oligomer elimination efficacy in vitro and the in vivo efficacy of RD2 was demonstrated in treatment studies by enhanced cognition in transgenic mouse models of amyloidosis. Here, we report on the in vitro and in vivo efficacy of the compound towards pyroglutamate-Aß, a particular aggressive Aß species. Using the transgenic TBA2.1 mouse model, which develops pyroglutamate-Aß(3-42) induced neurodegeneration, we are able to show that oral RD2 treatment resulted in a significant deceleration of the progression of the phenotype. The in vivo efficacy against this highly toxic Aß species further validates RD2 as a drug candidate for the therapeutic use in humans.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Brain/metabolism , Oligopeptides/pharmacology , Peptide Fragments/metabolism , Administration, Oral , Alzheimer Disease/pathology , Animals , Brain/pathology , Disease Models, Animal , Mice, Transgenic , Motor Activity/drug effects , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Phenotype , Stereoisomerism
4.
J Alzheimers Dis ; 64(3): 859-873, 2018.
Article in English | MEDLINE | ID: mdl-29966196

ABSTRACT

Diffusible amyloid-ß (Aß) oligomers are currently presumed to be the most cytotoxic Aß assembly and held responsible to trigger the pathogenesis of Alzheimer's disease (AD). Thus, Aß oligomers are a prominent target in AD drug development. Previously, we reported on our solely D-enantiomeric peptide D3 and its derivatives as AD drug candidates. Here, we compare one of the most promising D3 derivatives, ANK6, with its tandem version (tANK6), and its head-to-tail cyclized isoform (cANK6r). In vitro tests investigating the D-peptides' potencies to inhibit Aß aggregation, eliminate Aß oligomers, and reduce Aß-induced cytotoxicity revealed that all three D-peptides efficiently target Aß. Subsequent preclinical pharmacokinetic studies of the three all-D-peptides in wildtype mice showed promising blood-brain barrier permeability with cANK6r yielding the highest levels in brain. The peptides' potencies to lower Aß toxicity and their remarkable brain/plasma ratios make them promising AD drug candidates.


Subject(s)
Amyloid beta-Peptides/metabolism , Oligopeptides/pharmacokinetics , Oligopeptides/therapeutic use , Peptide Fragments/metabolism , Protein Aggregates/drug effects , Amyloid beta-Peptides/drug effects , Animals , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Oligopeptides/cerebrospinal fluid , Oligopeptides/chemistry , PC12 Cells/drug effects , PC12 Cells/metabolism , Peptide Fragments/drug effects , Protein Binding/drug effects , Protein Isoforms/pharmacokinetics , Rats , Stereoisomerism , Tissue Distribution/drug effects , Tritium/cerebrospinal fluid , Tritium/pharmacokinetics
5.
Article in English | MEDLINE | ID: mdl-29248770

ABSTRACT

During preclinical drug development, a method for quantification of unlabeled compounds in blood plasma samples from treatment or pharmacokinetic studies in mice is required. In the current work, a rapid, specific, sensitive and validated liquid chromatography mass-spectrometric UHPLC-ESI-QTOF-MS method was developed for the quantification of the therapeutic compound RD2 in mouse plasma. RD2 is an all-D-enantiomeric peptide developed for the treatment of Alzheimer's disease, a progressive neurodegenerative disease finally leading to dementia. Due to RD2's highly hydrophilic properties, the sample preparation and the chromatographic separation and quantification were very challenging. The chromatographic separation of RD2 and its internal standard were accomplished on an Acquity UPLC BEH C18 column (2.1 × 100 mm, 1.7 µm particle size) within 6.5 min at 50 °C with a flow rate of 0.5 mL/min. Mobile phases consisted of water and acetonitrile with 1% formic acid and 0.025% heptafluorobutyric acid, respectively. Ions were generated by electrospray ionization (ESI) in the positive mode and the peptide was quantified by QTOF-MS. The developed extraction method for RD2 from mouse plasma revealed complete recovery. The linearity of the calibration curve was in the range of 5.3 ng/mL to 265 ng/mL (r2 > 0.999) with a lower limit of detection (LLOD) of 2.65 ng/mL and a lower limit of quantification (LLOQ) of 5.3 ng/mL. The intra-day and inter-day accuracy and precision of RD2 in plasma ranged from -0.54% to 2.21% and from 1.97% to 8.18%, respectively. Moreover, no matrix effects were observed and RD2 remained stable in extracted mouse plasma at different conditions. Using this validated bioanalytical method, plasma samples of unlabeled RD2 or placebo treated mice were analyzed. The herein developed UHPLC-ESI-QTOF-MS method is a suitable tool for the quantitative analysis of unlabeled RD2 in plasma samples of treated mice.


Subject(s)
Amyloid beta-Peptides/blood , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/isolation & purification , Animals , Hydrophobic and Hydrophilic Interactions , Limit of Detection , Linear Models , Male , Mice , Mice, Transgenic , Oligonucleotides/isolation & purification , Oligonucleotides/metabolism , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization/methods , Stereoisomerism
6.
Eur J Pharm Sci ; 114: 93-102, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29225107

ABSTRACT

Alzheimer's disease (AD), until now, is an incurable progressive neurodegenerative disease. To target toxic amyloid ß oligomers in AD patients' brains and to convert them into non-toxic aggregation-incompetent species, we designed peptides consisting solely of d-enantiomeric amino acid residues. The original lead compound was named D3 and several D3 derivatives were designed to enhance beneficial properties. Here, we compare four d-peptides concerning their efficiencies to pass the blood-brain barrier (BBB). We demonstrate that the d-peptides' concentrations in murine brain directly correlate with concentrations in cerebrospinal fluid. The cyclic d-enantiomeric peptide cRD2D3 is characterized by the highest efficiency to pass the BBB. For in total three cyclic peptides we show that administration of cyclic peptides resulted in up to tenfold higher peak concentrations in brain as compared to their linear equivalents which have partially been characterized before (Jiang et al., 2015; Leithold et al., 2016a). These results suggest that cyclic peptides pass the murine BBB more efficiently than their linear equivalents. cRD2D3's proteolytic stability, oral bioavailability, long duration of action and its favorable brain/plasma ratio reveal that it may become a suitable drug for long-term AD-treatment from a pharmacokinetic point of view.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Biological Transport/physiology , Blood-Brain Barrier/drug effects , Brain/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Peptides, Cyclic/administration & dosage , Stereoisomerism , Tissue Distribution/drug effects , Tissue Distribution/physiology
7.
Sci Rep ; 7(1): 16275, 2017 11 24.
Article in English | MEDLINE | ID: mdl-29176708

ABSTRACT

While amyloid-ß protein (Aß) aggregation into insoluble plaques is one of the pathological hallmarks of Alzheimer's disease (AD), soluble oligomeric Aß has been hypothesized to be responsible for synapse damage, neurodegeneration, learning, and memory deficits in AD. Here, we investigate the in vitro and in vivo efficacy of the D-enantiomeric peptide RD2, a rationally designed derivative of the previously described lead compound D3, which has been developed to efficiently eliminate toxic Aß42 oligomers as a promising treatment strategy for AD. Besides the detailed in vitro characterization of RD2, we also report the results of a treatment study of APP/PS1 mice with RD2. After 28 days of treatment we observed enhancement of cognition and learning behaviour. Analysis on brain plaque load did not reveal significant changes, but a significant reduction of insoluble Aß42. Our findings demonstrate that RD2 was significantly more efficient in Aß oligomer elimination in vitro compared to D3. Enhanced cognition without reduction of plaque pathology in parallel suggests that synaptic malfunction due to Aß oligomers rather than plaque pathology is decisive for disease development and progression. Thus, Aß oligomer elimination by RD2 treatment may be also beneficial for AD patients.


Subject(s)
Plaque, Amyloid/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cognition/drug effects , Disease Models, Animal , Female , Mice , Peptides/chemistry , Peptides/therapeutic use , Plaque, Amyloid/drug therapy
8.
Biochim Biophys Acta ; 1858(11): 2717-2724, 2016 11.
Article in English | MEDLINE | ID: mdl-27423267

ABSTRACT

The application of small peptides targeting amyloid beta (Aß) is one of many drug development strategies for the treatment of Alzheimer's disease (AD). We have previously identified several peptides consisting solely of D-enantiomeric amino acid residues obtained from mirror-image phage display selection, which bind to Aß in different assembly states and eliminate toxic Aß aggregates. Some of these D-peptides show both diagnostic and therapeutic potential in vitro and in vivo. Here we have analysed the similarity of the arginine-rich D-peptide D3 to the arginine-rich motif (ARM) of the human immunodeficiency virus type 1 transactivator of transcription (HIV-Tat) protein, and examined its in vivo blood-brain barrier (BBB) permeability using wild type mice and transgenic mouse models of Alzheimer's disease. We are able to demonstrate that D3 rapidly enters the brain where it can be found associated with amyloid plaques suggesting a direct penetration of BBB.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/chemistry , Blood-Brain Barrier/drug effects , Neuroprotective Agents/pharmacokinetics , Oligopeptides/metabolism , Administration, Oral , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Capillary Permeability , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Oligopeptides/chemical synthesis , Oligopeptides/pharmacokinetics , Sequence Alignment , tat Gene Products, Human Immunodeficiency Virus/chemistry
9.
Eur J Pharm Sci ; 89: 31-8, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27086111

ABSTRACT

Peptides are more and more considered for the development of drug candidates. However, they frequently exhibit severe disadvantages such as instability and unfavourable pharmacokinetic properties. Many peptides are rapidly cleared from the organism and oral bioavailabilities as well as in vivo half-lives often remain low. In contrast, some peptides consisting solely of d-enantiomeric amino acid residues were shown to combine promising therapeutic properties with high proteolytic stability and enhanced pharmacokinetic parameters. Recently, we have shown that D3 and RD2 have highly advantageous pharmacokinetic properties. Especially D3 has already proven promising properties suitable for treatment of Alzheimer's disease. Here, we analyse the pharmacokinetic profiles of D3D3 and RD2D3, which are head-to-tail tandem d-peptides built of D3 and its derivative RD2. Both D3D3 and RD2D3 show proteolytic stability in mouse plasma and organ homogenates for at least 24h and in murine and human liver microsomes for 4h. Notwithstanding their high affinity to plasma proteins, both peptides are taken up into the brain following i.v. as well as i.p. administration. Although both peptides contain identical d-amino acid residues, they are arranged in a different sequence order and the peptides show differences in pharmacokinetic properties. After i.p. administration RD2D3 exhibits lower plasma clearance and higher bioavailability than D3D3. We therefore concluded that the amino acid sequence of RD2 leads to more favourable pharmacokinetic properties within the tandem peptide, which underlines the importance of particular sequence motifs, even in short peptides, for the design of further therapeutic d-peptides.


Subject(s)
Alzheimer Disease/drug therapy , Peptides/pharmacology , Amino Acid Sequence , Amino Acids/metabolism , Animals , Biological Availability , Blood Proteins/metabolism , Brain/drug effects , Brain/metabolism , Half-Life , Humans , Kinetics , Mice , Mice, Inbred C57BL , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Peptides/pharmacokinetics , Stereoisomerism
10.
Pharm Res ; 33(2): 328-36, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26381279

ABSTRACT

PURPOSE: It has been shown that amyloid ß (Aß) oligomers play an important role in the pathology of Alzheimer's disease (AD). D3, a peptide consisting solely of D-enantiomeric amino acid residues, was developed to specifically eliminate Aß oligomers and is therapeutically active in transgenic AD mice. D-peptides have several advantages over L-peptides, but little is known about their pharmacokinetic potential in vivo. Here, we analysed the pharmacokinetic properties of RD2, a rationally designed and potent D3 derivative. METHODS: The pharmacokinetic analysis was performed using (3)H-RD2 after administration via several routes in mice. The time dependent amount of radiolabelled RD2 was measured in plasma and several organ homogenates by liquid scintillation counting. Furthermore, binding to plasma proteins was estimated. RESULTS: RD2 penetrates into the brain, where it is thought to implement its therapeutic function. All administration routes result in a maximal brain concentration per dose (Cmax/D) of 0.06 (µg/g)/(mg/kg) with brain/plasma ratios ranging between 0.7 and 1.0. RD2 shows a small elimination constant and a long terminal half-life in plasma of more than 2 days. It also exhibits high bioavailability after i.p., s.c. or p.o. administration. CONCLUSIONS: These excellent pharmacokinetic properties confirm that RD2 is a very promising drug candidate for AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Brain/metabolism , Peptides/pharmacokinetics , Alzheimer Disease/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Humans , Male , Mice, Inbred C57BL , Peptides/blood , Peptides/pharmacology
11.
PLoS One ; 10(6): e0128553, 2015.
Article in English | MEDLINE | ID: mdl-26046986

ABSTRACT

Targeting toxic amyloid beta (Aß) oligomers is currently a very attractive drug development strategy for treatment of Alzheimer´s disease. Using mirror-image phage display against Aß1-42, we have previously identified the fully D-enantiomeric peptide D3, which is able to eliminate Aß oligomers and has proven therapeutic potential in transgenic Alzheimer´s disease animal models. However, there is little information on the pharmacokinetic behaviour of D-enantiomeric peptides in general. Therefore, we conducted experiments with the tritium labelled D-peptide D3 (3H-D3) in mice with different administration routes to study its distribution in liver, kidney, brain, plasma and gastrointestinal tract, as well as its bioavailability by i.p. and p.o. administration. In addition, we investigated the metabolic stability in liver microsomes, mouse plasma, brain, liver and kidney homogenates, and estimated the plasma protein binding. Based on its high stability and long biological half-life, our pharmacokinetic results support the therapeutic potential of D-peptides in general, with D3 being a new promising drug candidate for Alzheimer´s disease treatment.


Subject(s)
Alzheimer Disease/drug therapy , Oligopeptides/therapeutic use , Animals , Area Under Curve , Blood Proteins/chemistry , Blood Proteins/metabolism , Chromatography, Thin Layer , Disease Models, Animal , Drug Evaluation, Preclinical , Half-Life , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microsomes, Liver/metabolism , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Protein Binding , ROC Curve , Stereoisomerism , Tritium/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...