Network


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

Hotspot


Dive into the research topics where Lise-Lotte Olsson is active.

Publication


Featured researches published by Lise-Lotte Olsson.


Current Topics in Medicinal Chemistry | 2007

An Integrated Approach to Fragment-Based Lead Generation:Philosophy, Strategy and Case Studies from AstraZenecas Drug Discovery Programmes

Jeffrey S. Albert; Niklas Blomberg; Alexander L. Breeze; Alastair J. H. Brown; Jeremy N. Burrows; Philip Duke Edwards; Rutger H. A. Folmer; Stefan Geschwindner; Ed J. Griffen; Peter W. Kenny; Thorsten Nowak; Lise-Lotte Olsson; Hitesh Sanganee; Adam B. Shapiro

Fragment-based lead generation (FBLG) has recently emerged as an alternative to traditional high throughput screening (HTS) to identify initial chemistry starting points for drug discovery programs. In comparison to HTS screening libraries, the screening sets for FBLG tend to contain orders of magnitude fewer compounds, and the compounds themselves are less structurally complex and have lower molecular weight. This report summarises the advent of FBLG within the industry and then describes the FBLG experience at AstraZeneca. We discuss (1) optimising the design of screening libraries, (2) hit detection methodologies, (3) evaluation of hit quality and use of ligand efficiency calculations, and (4) approaches to evolve fragment-based, low complexity hits towards drug-like leads. Furthermore, we exemplify our use of FBLG with case studies in the following drug discovery areas: antibacterial enzyme targets, GPCRs (melanocortin 4 receptor modulators), prostaglandin D2 synthase inhibitors, phosphatase inhibitors (protein tyrosine phosphotase 1B), and protease inhibitors (b-secretase).


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%.


PLOS ONE | 2014

Engineering Neprilysin Activity and Specificity to Create a Novel Therapeutic for Alzheimer's Disease

Carl Webster; Matthew Burrell; Lise-Lotte Olsson; Susan B. Fowler; Sarah Digby; Alan Sandercock; Arjan Snijder; Jan Tebbe; Ulrich Haupts; Joanna Grudzinska; Lutz Jermutus; Christin Andersson

Neprilysin is a transmembrane zinc metallopeptidase that degrades a wide range of peptide substrates. It has received attention as a potential therapy for Alzheimer’s disease due to its ability to degrade the peptide amyloid beta. However, its broad range of peptide substrates has the potential to limit its therapeutic use due to degradation of additional peptides substrates that tightly regulate many physiological processes. We sought to generate a soluble version of the ectodomain of neprilysin with improved activity and specificity towards amyloid beta as a potential therapeutic for Alzheimer’s disease. Extensive amino acid substitutions were performed at positions surrounding the active site and inner surface of the enzyme and variants screened for activity on amyloid beta 1–40, 1–42 and a variety of other physiologically relevant peptides. We identified several mutations that modulated and improved both enzyme selectivity and intrinsic activity. Neprilysin variant G399V/G714K displayed an approximately 20-fold improved activity on amyloid beta 1–40 and up to a 3,200-fold reduction in activity on other peptides. Along with the altered peptide substrate specificity, the mutant enzyme produced a markedly altered series of amyloid beta cleavage products compared to the wild-type enzyme. Crystallisation of the mutant enzyme revealed that the amino acid substitutions result in alteration of the shape and size of the pocket containing the active site compared to the wild-type enzyme. The mutant enzyme offers the potential for the more efficient degradation of amyloid beta in vivo as a therapeutic for the treatment of Alzheimer’s disease.


Bioorganic & Medicinal Chemistry Letters | 2012

Synthesis and Structure-Activity Relationship of 4-(1,3-Benzothiazol-2-Yl)-Thiophene-2-Sulfonamides as Cyclin-Dependent Kinase 5 (Cdk5)/P25 Inhibitors.

Jonas Malmström; Jenny Viklund; Can Slivo; Ana R. Costa; Mickaël Maudet; Catrin Sandelin; Gösta Hiller; Lise-Lotte Olsson; Anna Aagaard; Stefan Geschwindner; Yafeng Xue; Mervi Vasänge

4-(1,3-Benzothiazol-2-yl)thiophene-2-sulfonamide (4a) was found to be a moderately potent inhibitor of cyclin-dependent kinase 5 (cdk5) from a HTS screen. The synthesis and SAR around this hit is described. The X-ray coordinates of ligand 4a with cdk5 are also reported, showing an unusual binding mode to the hinge region via a water molecule.


Bioorganic & Medicinal Chemistry Letters | 2016

Design and Structure-Activity Relationships of Novel Inhibitors of Human Rhinovirus 3C Protease.

Sameer Kawatkar; M. Gagnon; Valerie Hoesch; Choi-Lai Tiong-Yip; Kenneth D. Johnson; Margareta Ek; E. Nilsson; T. Lister; Lise-Lotte Olsson; J. Patel; Qin Yu

Human rhinovirus (HRV) is a primary cause of common cold and is linked to exacerbation of underlying respiratory diseases such as asthma and COPD. HRV 3C protease, which is responsible for cleavage of viral polyprotein in to proteins essential for viral life-cycle, represents an important target. We have designed proline- and azetidine-based analogues of Rupintrivir that target the P2 pocket of the binding site. Potency optimization, aided with X-ray crystallography and quantum mechanical calculations, led to compounds with activity against a broad spectrum of HRV serotypes. Altogether, these compounds represent alternative starting points to identify promising leads in our continual efforts to treat HRV infections.


MedChemComm | 2013

A monomeric form of iNOS can rationalise observed SAR for inhibitors of dimerisation: quantum mechanics and docking compared

Andrew G. Leach; Lise-Lotte Olsson; Daniel Jon Warner

Two series of compounds that inhibit dimerisation of iNOS have been reported in the literature and studied crystallographically. We have applied a range of docking techniques to these compounds and shown that the geometry of the complexes can be reproduced well but that even within a series of related compounds the docking scores do not rank compounds well. Quantum mechanical studies using a model system for the protein and a range of density functionals are able to reproduce the geometry of the complexes and the corresponding affinities. The combination of docking to generate geometries and quantum mechanical calculations to predict complexation energies is a powerful one for rationalising observed changes and designing improved compounds.


Journal of Medicinal Chemistry | 2007

Application of Fragment-Based Lead Generation to the Discovery of Novel, Cyclic Amidine β-Secretase Inhibitors with Nanomolar Potency, Cellular Activity, and High Ligand Efficiency

Philip Duke Edwards; Jeffrey S. Albert; Mark Sylvester; David Aharony; Donald W. Andisik; Owen Callaghan; James B. Campbell; Robin A. E. Carr; Gianni Chessari; Miles Congreve; Martyn Frederickson; Rutger H. A. Folmer; Stefan Geschwindner; Gerard M. Koether; Karin Kolmodin; Jennifer R. Krumrine; Russell C. Mauger; Christopher W. Murray; Lise-Lotte Olsson; Sahil Patel; Nate Spear; Gaochao Tian

Collaboration


Dive into the Lise-Lotte Olsson'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
Researchain Logo
Decentralizing Knowledge