Network


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

Hotspot


Dive into the research topics where Britt-Marie Swahn is active.

Publication


Featured researches published by Britt-Marie Swahn.


Journal of Neurochemistry | 2010

Characterization of AZD4694, a novel fluorinated Abeta plaque neuroimaging PET radioligand

Anders Juréus; Britt-Marie Swahn; Johan Sandell; Fredrik Jeppsson; Allan E. Johnson; Peter Johnström; Jan Neelissen; Dan Sunnemark; Lars Farde; Samuel P.S. Svensson

J. Neurochem. (2010) 114, 784–794.


Journal of Biological Chemistry | 2012

Discovery of AZD3839, a Potent and Selective BACE1 Inhibitor Clinical Candidate for the Treatment of Alzheimer Disease

Fredrik Jeppsson; Susanna Eketjäll; Juliette Janson; Sofia Karlström; Susanne Gustavsson; Lise-Lotte Olsson; Bart Ploeger; Gvido Cebers; Karin Kolmodin; Britt-Marie Swahn; Stefan Berg; Tjerk Bueters; Johanna Fälting

Background: BACE1 inhibitors target the first step in Aβ formation and are tractable drugs for halting disease progression in Alzheimer disease. Results: AZD3839 is a novel BACE1 inhibitor that effectively reduces brain and CSF Aβ levels in several preclinical species. Conclusion: Based on the preclinical profile, AZD3839 was progressed into Phase I. Significance: AZD3839 may have disease-modifying potential in the treatment of Alzheimer disease. β-Site amyloid precursor protein cleaving enzyme1 (BACE1) is one of the key enzymes involved in the processing of the amyloid precursor protein (APP) and formation of amyloid β peptide (Aβ) species. Because cerebral deposition of Aβ species might be critical for the pathogenesis of Alzheimer disease, BACE1 has emerged as a key target for the treatment of this disease. Here, we report the discovery and comprehensive preclinical characterization of AZD3839, a potent and selective inhibitor of human BACE1. AZD3839 was identified using fragment-based screening and structure-based design. In a concentration-dependent manner, AZD3839 inhibited BACE1 activity in a biochemical fluorescence resonance energy transfer (FRET) assay, Aβ and sAPPβ release from modified and wild-type human SH-SY5Y cells and mouse N2A cells as well as from mouse and guinea pig primary cortical neurons. Selectivity against BACE2 and cathepsin D was 14 and >1000-fold, respectively. AZD3839 exhibited dose- and time-dependent lowering of plasma, brain, and cerebrospinal fluid Aβ levels in mouse, guinea pig, and non-human primate. Pharmacokinetic/pharmacodynamic analyses of mouse and guinea pig data showed a good correlation between the potency of AZD3839 in primary cortical neurons and in vivo brain effects. These results suggest that AZD3839 effectively reduces the levels of Aβ in brain, CSF, and plasma in several preclinical species. It might, therefore, have disease-modifying potential in the treatment of Alzheimer disease and related dementias. Based on the overall pharmacological profile and its drug like properties, AZD3839 has been progressed into Phase 1 clinical trials in man.


Journal of Medicinal Chemistry | 2012

Design and synthesis of β-site amyloid precursor protein cleaving enzyme (BACE1) inhibitors with in vivo brain reduction of β-amyloid peptides.

Britt-Marie Swahn; Karin Kolmodin; Sofia Karlström; Stefan Berg; Peter Söderman; Jörg Holenz; Johan Lindström; M. Sundstrom; Jacob Kihlström; Can Slivo; Lars I. Andersson; David Pyring; Didier Rotticci; Liselotte Öhberg; Annika Kers; Krisztián Bogár; Fredrik von Kieseritzky; Margareta Bergh; Lise-Lotte Olsson; Juliette Janson; Susanna Eketjäll; Biljana Georgievska; Fredrik Jeppsson; Johanna Fälting

The evaluation of a series of aminoisoindoles as β-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors and the discovery of a clinical candidate drug for Alzheimers disease, (S)-32 (AZD3839), are described. The improvement in permeability properties by the introduction of fluorine adjacent to the amidine moiety, resulting in in vivo brain reduction of Aβ40, is discussed. Due to the basic nature of these compounds, they displayed affinity for the human ether-a-go-go related gene (hERG) ion channel. Different ways to reduce hERG inhibition and increase hERG margins for this series are described, culminating in (S)-16 and (R)-41 showing large in vitro margins with BACE1 cell IC(50) values of 8.6 and 0.16 nM, respectively, and hERG IC(50) values of 16 and 2.8 μM, respectively. Several compounds were advanced into pharmacodynamic studies and demonstrated significant reduction of β-amyloid peptides in mouse brain following oral dosing.


Journal of Medicinal Chemistry | 2012

New Aminoimidazoles as β-Secretase (BACE-1) Inhibitors Showing Amyloid-β (Aβ) Lowering in Brain

Ylva Gravenfors; Jenny Viklund; Jan Blid; Tobias Ginman; Sofia Karlström; Jacob Kihlström; Karin Kolmodin; Johan Lindström; Stefan Berg; Fredrik von Kieseritzky; Can Slivo; Britt-Marie Swahn; Lise-Lotte Olsson; Patrik Johansson; Susanna Eketjäll; Johanna Fälting; Fredrik Jeppsson; Kia Strömberg; Juliette Janson; Fredrik Rahm

Amino-2H-imidazoles are described as a new class of BACE-1 inhibitors for the treatment of Alzheimers disease. Synthetic methods, crystal structures, and structure-activity relationships for target activity, permeability, and hERG activity are reported and discussed. Compound (S)-1m was one of the most promising compounds in this report, with high potency in the cellular assay and a good overall profile. When guinea pigs were treated with compound (S)-1m, a concentration and time dependent decrease in Aβ40 and Aβ42 levels in plasma, brain, and CSF was observed. The maximum reduction of brain Aβ was 40-50%, 1.5 h after oral dosing (100 μmol/kg). The results presented highlight the potential of this new class of BACE-1 inhibitors with good target potency and with low effect on hERG, in combination with a fair CNS exposure in vivo.


Bioorganic & Medicinal Chemistry Letters | 2012

Aminoimidazoles as Bace-1 Inhibitors: The Challenge to Achieve in Vivo Brain Efficacy

Britt-Marie Swahn; Jörg Holenz; Jacob Kihlström; Karin Kolmodin; Johan Lindström; Niklas Plobeck; Didier Rotticci; Fernando Sehgelmeble; M. Sundstrom; Stefan Berg; Johanna Fälting; Biljana Georgievska; Susanne Gustavsson; Jan Neelissen; Margareta Ek; Lise-Lotte Olsson

The evaluation of a series of bicyclic aminoimidazoles as potent BACE-1 inhibitors is described. The crystal structures of compounds 14 and 23 in complex with BACE-1 reveal hydrogen bond interactions with the protein important for achieving potent inhibition. The optimization of permeability and efflux properties of the compounds is discussed as well as the importance of these properties for attaining in vivo brain efficacy. Compound (R)-25 was selected for evaluation in vivo in wild type mice and 1.5h after oral co-administration of 300μmol/kg (R)-25 and efflux inhibitor GF120918 the brain Aβ40 level was reduced by 17% and the plasma Aβ40 level by 76%.


Bioorganic & Medicinal Chemistry Letters | 2011

Strategies to improve in vivo toxicology outcomes for basic candidate drug molecules

Tim Luker; Lilian Alcaraz; Kamaldeep K. Chohan; Niklas Blomberg; Dearg S. Brown; Roger John Butlin; Thomas Elebring; Andrew Griffin; Simon D. Guile; Stephen St-Gallay; Britt-Marie Swahn; Steve Swallow; Michael J. Waring; Mark C. Wenlock; Paul D. Leeson

A valid PLS-DA model to predict attrition in pre-clinical toxicology for basic oral candidate drugs was built. A combination of aromatic/aliphatic balance, flatness, charge distribution and size descriptors helped predict the successful progression of compounds through a wide range of toxicity testing. Eighty percent of an independent test set of marketed post-2000 basic drugs could be successfully classified using the model, indicating useful forward predictivity. The themes within this work provide additional guidance for medicinal design chemists and complement other literature property guidelines.


The Journal of Neuroscience | 2013

AZ-4217: a high potency BACE inhibitor displaying acute central efficacy in different in vivo models and reduced amyloid deposition in Tg2576 mice.

Susanna Eketjäll; Juliette Janson; Fredrik Jeppsson; Alexander Svanhagen; Karin Kolmodin; Susanne Gustavsson; Ann-Cathrin Radesäter; Kristina Eliason; Sveinn Briem; Paulina Appelkvist; Camilla Niva; Anna-Lena Berg; Sofia Karlström; Britt-Marie Swahn; Johanna Fälting

Aβ, the product of APP (amyloid precursor protein), has been implicated in the pathophysiology of Alzheimers disease (AD). β-Site APP cleaving enzyme1 (BACE1) is the enzyme initiating the processing of the APP to Aβ peptides. Small molecule BACE1 inhibitors are expected to decrease Aβ-peptide generation and thereby reduce amyloid plaque formation in the brain, a neuropathological hallmark of AD. BACE1 inhibition thus addresses a key mechanism in AD and its potential as a therapeutic target is currently being addressed in clinical studies. Here, we report the discovery and the pharmacokinetic and pharmacodynamic properties of BACE1 inhibitor AZ-4217, a high potency compound (IC50 160 pm in human SH-SY5Y cells) with an excellent in vivo efficacy. Central efficacy of BACE1 inhibition was observed after a single dose in C57BL/6 mice, guinea pigs, and in an APP transgenic mouse model of cerebral amyloidosis (Tg2576). Furthermore, we demonstrate that in a 1 month treatment paradigm BACE1 inhibition of Aβ production does lower amyloid deposition in 12-month-old Tg2576 mice. These results strongly support BACE1 inhibition as concretely impacting amyloid deposition and therefore potentially an important approach for therapeutic intervention in AD.


Synapse | 2010

Radiosynthesis of the candidate β‐amyloid radioligand [11C]AZD2184: Positron emission tomography examination and metabolite analysis in cynomolgus monkeys

Jan D. Andersson; Katarina Varnäs; Zsolt Cselényi; Balázs Gulyás; David Wensbo; Sjoerd J. Finnema; Britt-Marie Swahn; Samuel P.S. Svensson; Svante Nyberg; Lars Farde; Christer Halldin

β‐Amyloid accumulation is associated with the pathogenesis of Alzheimers disease (AD). AZD2184, a new radioligand for high‐contrast positron emission tomography (PET) imaging of Aβ‐deposits, has recently been developed and characterized in vitro and in rodents ex vivo. The objective of this study was to label AZD2184 with carbon‐11, to perform in vivo characterization of [11C]AZD2184 ([11C]5) in the cynomolgus monkey brain as well as whole‐body dosimetry, and to examine the metabolism of the labeled radioligand. [11C]5 was prepared by a two‐step radiosynthesis starting with the reaction of 5‐(6‐(tert‐butyldimethylsilyloxy)benzo[d]thiazol‐2‐yl)pyridin‐2‐amine with [11C]methyl iodide followed by deprotection using water. Four brain PET measurements in two cynomolgus monkeys and one whole‐body PET measurement were performed with [11C]5. There was a high and rapid brain uptake (2.2–3.4% of injected dose at 2 min). The distribution of brain radioactivity was fairly uniform, with early to late‐brain concentration ratios (peak vs. 60 min) higher for [11C]5 than ratios previously reported for [11C]PIB (8.2 and 4.6, respectively). Based on the whole‐body data, it was estimated that an effective dose in an adult male would be 6.2 μSv/MBq and thus would be safe from a radiation point of view for multiple scans within the same year. [11C]5 shows binding characteristics, suggesting low levels of white‐matter retention, and may thus provide improved contrast when compared with currently used PET radioligands for visualization of Aβ‐deposits. On the basis of the labeling chemistry and the results of the biological evaluation, we conclude that [11C]5 should be useful for routine clinical studies. Synapse 64:733–741, 2010.


Bioorganic & Medicinal Chemistry Letters | 2010

Synthesis and evaluation of 2-pyridylbenzothiazole, 2-pyridylbenzoxazole and 2-pyridylbenzofuran derivatives as 11C-PET imaging agents for β-amyloid plaques

Britt-Marie Swahn; David Wensbo; Johan Sandell; Daniel Sohn; Can Slivo; David Pyring; Jonas Malmström; Erwan Arzel; Michaela Vallin; Margareta Bergh; Fredrik Jeppsson; Allan E. Johnson; Anders Juréus; Jan Neelissen; Samuel P.S. Svensson

The syntheses and SAR of new series of beta-amyloid binding agents are reported. The effort to optimize signal-to-background ratios for these ligands are described. Compounds 8, 21 and 30 displayed desirable lipophilicity and pharmacokinetic properties. Compounds 8 and 21 were evaluated with in vitro autoradiographic studies and in vivo in APP/PS1 transgenic mice. It is shown that it was possible to increase the signal-to-background ratios compared to PIB 1, as demonstrated by compounds 8 and 21.


Bioorganic & Medicinal Chemistry Letters | 2012

Synthesis and evaluation of pyridylbenzofuran, pyridylbenzothiazole and pyridylbenzoxazole derivatives as 18F-PET imaging agents for β-amyloid plaques

Britt-Marie Swahn; Johan Sandell; David Pyring; Margareta Bergh; Fredrik Jeppsson; Anders Juréus; Jan Neelissen; Peter Johnström; Magnus Schou; Samuel P.S. Svensson

The synthesis and SAR of new β-amyloid binding agents are reported. Evaluation of important properties for achieving good signal-to-background ratio is described. Compounds 27, 33, and 36 displayed desirable lipophilic and pharmacokinetic properties. Compound 27 was further evaluated with autoradiographic studies in vitro on human brain tissue and in vivo in Tg2576 mice. Compound 27 showed an increased signal-to-background ratio compared to flutemetamol 4, indicating its suitability as PET ligand for β-amyloid deposits in AD patients. The preparation of the corresponding (18)F-labeled PET radioligand of compound 27 is presented.

Collaboration


Dive into the Britt-Marie Swahn's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge