Network


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

Hotspot


Dive into the research topics where Andrea Olland is active.

Publication


Featured researches published by Andrea Olland.


Journal of Medicinal Chemistry | 2010

Design and Synthesis of 5,5′-Disubstituted Aminohydantoins as Potent and Selective Human β-Secretase (BACE1) Inhibitors

Michael S. Malamas; Jim Erdei; Iwan Gunawan; Jim Turner; Yun Hu; Erik Wagner; Kristi Fan; Rajiv Chopra; Andrea Olland; Steve Jacobsen; Ronald L. Magolda; Menelas N. Pangalos; Albert Jean Robichaud

The identification of small molecule aminohydantoins as potent and selective human beta-secretase inhibitors is reported. These analogues exhibit low nannomolar potency for BACE1, show comparable activity in a cell-based (ELISA) assay, and demonstrate >100x selectivity for the other structurally related aspartyl proteases BACE2, cathepsinD, renin, and pepsin. On the basis of the cocrystal structure of the HTS-hit 2 in the BACE1 active site and by use of a structure-based drug design approach, we methodically explored the comparatively large binding pocket of the BACE1 enzyme and identified key interactions between the ligand and the protein that contributed to the affinity. One of the more potent compounds, (S)-55, displayed an IC(50) value for BACE1 of 10 nM and exhibited comparable cellular activity (EC(50) = 20 nM) in the ELISA assay. Acute oral administration of (S)-55 at 100 mg/kg resulted in a 69% reduction of plasma A beta(40) at 8 h in a Tg2576 mouse (p < 0.001).


Journal of Medicinal Chemistry | 2009

Aminoimidazoles as potent and selective human beta-secretase (BACE1) inhibitors.

Michael S. Malamas; Jim Erdei; Iwan Gunawan; Keith Douglas Barnes; Matthew S. Johnson; Yu Hui; Jim Turner; Yun Hu; Erik Wagner; Kristi Fan; Andrea Olland; Albert Jean Robichaud

The identification of small molecule aminoimidazoles as potent and selective human beta-secretase inhibitors is reported. These analogues demonstrate low nannomolar potency for BACE1 in a FRET assay, exhibit comparable activity in a cell-based (ELISA) assay, and show >100x selectivity for the other structurally related aspartyl proteases BACE2, cathepsin D, renin, and pepsin. Our design strategy was supported by molecular modeling studies based on the cocrystal structure of the HTS-hit 3 in the BACE1 active site. These strategies enabled us to integrate pyridine and pyrimidine groups on 3 extending deep into the S3 region of the BACE1 binding pocket and enhancing the ligands potency. Compound (R)-37 displayed an IC50 value for BACE1 of 20 nM, cellular activity of 90 nM, and >100-fold selectivity over related aspartyl proteases. Acute oral administration of (R)-37 at 30 mg/kg resulted in a significant 71% reduction of plasma Abeta40 measured at the 6 h time point in a Tg2576 mouse model (p < 0.001).


Bioorganic & Medicinal Chemistry Letters | 2010

Design and synthesis of aminohydantoins as potent and selective human β-secretase (BACE1) inhibitors with enhanced brain permeability.

Michael S. Malamas; Albert J. Robichaud; Jim Erdei; Dominick Anthony Quagliato; William Ronald Solvibile; Ping Zhou; Koi Michele Morris; Jim Turner; Erik Wagner; Kristi Fan; Andrea Olland; Steve Jacobsen; Peter Reinhart; David Riddell; Menelas N. Pangalos

The identification of small molecule aminohydantoins as potent and selective human β-secretase inhibitors is reported. These analogs exhibit good brain permeability (40-70%), low nanomolar potency for BACE1, and demonstrate >100-fold selectivity for the structurally related aspartyl proteases cathepsin D, renin and pepsin. Alkyl and alkoxy groups at the meta-position of the P1 phenyl, which extend toward the S3 region of the enzyme, have contributed to the ligands reduced affinity for the efflux transporter protein P-gp, and decreased topological polar surface area, thus resulting in enhanced brain permeability. A fluorine substitution at the para-position of the P1 phenyl has contributed to 100-fold decrease of CYP3A4 inhibition and enhancement of compound metabolic stability. The plasma and brain protein binding properties of these new analogs are affected by substitutions at the P1 phenyl moiety. Higher compound protein binding was observed in the brain than in the plasma. Two structurally diverse potent BACE1 inhibitors (84 and 89) reduced 30% plasma Aβ40 in the Tg2576 mice in vivo model at 30 mg/kg p.o..


Protein Science | 2009

Triad of polar residues implicated in pH specificity of acidic mammalian chitinase

Andrea Olland; James Strand; Eleonora Presman; Robert M. Czerwinski; Diane Joseph-McCarthy; Rustem Krykbaev; Gerhard Schlingmann; Rajiv Chopra; Laura Lin; Margaret Fleming; Ron Kriz; Mark Stahl; William Stuart Somers; Lori Fitz; Lidia Mosyak

Acidic mammalian chitinase (AMCase) is a mammalian chitinase that has been implicated in allergic asthma. One of only two active mammalian chinases, AMCase, is distinguished from other chitinases by several unique features. Here, we present the novel structure of the AMCase catalytic domain, both in the apo form and in complex with the inhibitor methylallosamidin, determined to high resolution by X‐ray crystallography. These results provide a structural basis for understanding some of the unique characteristics of this enzyme, including the low pH optimum and the preference for the β‐anomer of the substrate. A triad of polar residues in the second‐shell is found to modulate the highly conserved chitinase active site. As a novel target for asthma therapy, structural details of AMCase activity will help guide the future design of specific and potent AMCase inhibitors.


Journal of Biological Chemistry | 2013

Crystal Structure of A Human IκB Kinase β Asymmetric Dimer

Shenping Liu; Yohann R. Misquitta; Andrea Olland; Mark Johnson; Kerry S. Kelleher; Ron Kriz; Laura Lin; Mark Stahl; Lidia Mosyak

Background: IκB kinase β is a key regulator in the NκB signaling pathway. Results: Crystal structure of a human IKKβ asymmetric dimer shows one kinase active site phosphorylated and in the active conformation and the other unphosphorylated and inactive. Conclusion: Depending on the phosphorylation state, IKKβ can adopt distinct dimeric geometry. Significance: High resolution structure of hIKKβ provides structural basis for its activation and potential use of inhibitor design. Phosphorylation of inhibitor of nuclear transcription factor κB (IκB) by IκB kinase (IKK) triggers the degradation of IκB and migration of cytoplasmic κB to the nucleus where it promotes the transcription of its target genes. Activation of IKK is achieved by phosphorylation of its main subunit, IKKβ, at the activation loop sites. Here, we report the 2.8 Å resolution crystal structure of human IKKβ (hIKKβ), which is partially phosphorylated and bound to the staurosporine analog K252a. The hIKKβ protomer adopts a trimodular structure that closely resembles that from Xenopus laevis (xIKKβ): an N-terminal kinase domain (KD), a central ubiquitin-like domain (ULD), and a C-terminal scaffold/dimerization domain (SDD). Although hIKKβ and xIKKβ utilize a similar dimerization mode, their overall geometries are distinct. In contrast to the structure resembling closed shears reported previously for xIKKβ, hIKKβ exists as an open asymmetric dimer in which the two KDs are further apart, with one in an active and the other in an inactive conformation. Dimer interactions are limited to the C-terminal six-helix bundle that acts as a hinge between the two subunits. The observed domain movements in the structures of IKKβ may represent trans-phosphorylation steps that accompany IKKβ activation.


Journal of Biological Chemistry | 2013

Atypical Antigen Recognition Mode of a Shark Immunoglobulin New Antigen Receptor (IgNAR) Variable Domain Characterized by Humanization and Structural Analysis

Oleg V. Kovalenko; Andrea Olland; Nicole Piche-Nicholas; Adarsh Godbole; Daniel King; Kristine Svenson; Valerie Calabro; Mischa R. Müller; Caroline Barelle; William S. Somers; Davinder Gill; Lidia Mosyak; Lioudmila Tchistiakova

Background: Single domain variable regions of shark antibodies (V-NARs) are promising biotherapeutic candidates. Results: A V-NAR specific for human serum albumin was humanized, and its crystal structure in complex with the antigen was solved, revealing an unusual recognition mode. Conclusion: Humanization preserved antigen binding properties and activity of the parental shark antibody. Significance: A structural framework for humanization of shark antibodies was established. The immunoglobulin new antigen receptors (IgNARs) are a class of Ig-like molecules of the shark immune system that exist as heavy chain-only homodimers and bind antigens by their single domain variable regions (V-NARs). Following shark immunization and/or in vitro selection, V-NARs can be generated as soluble, stable, and specific high affinity monomeric binding proteins of ∼12 kDa. We have previously isolated a V-NAR from an immunized spiny dogfish shark, named E06, that binds specifically and with high affinity to human, mouse, and rat serum albumins. Humanization of E06 was carried out by converting over 60% of non-complementarity-determining region residues to those of a human germ line Vκ1 sequence, DPK9. The resulting huE06 molecules have largely retained the specificity and affinity of antigen binding of the parental V-NAR. Crystal structures of the shark E06 and its humanized variant (huE06 v1.1) in complex with human serum albumin (HSA) were determined at 3- and 2.3-Å resolution, respectively. The huE06 v1.1 molecule retained all but one amino acid residues involved in the binding site for HSA. Structural analysis of these V-NARs has revealed an unusual variable domain-antigen interaction. E06 interacts with HSA in an atypical mode that utilizes extensive framework contacts in addition to complementarity-determining regions that has not been seen previously in V-NARs. On the basis of the structure, the roles of various elements of the molecule are described with respect to antigen binding and V-NAR stability. This information broadens the general understanding of antigen recognition and provides a framework for further design and humanization of shark IgNARs.


Bioorganic & Medicinal Chemistry Letters | 2010

Novel pyrrolyl 2-aminopyridines as potent and selective human beta-secretase (BACE1) inhibitors.

Michael S. Malamas; Keith Douglas Barnes; Yu Hui; Matthew S. Johnson; Frank Lovering; Jeff Condon; William Floyd Fobare; William Ronald Solvibile; Jim Turner; Yun Hu; Eric S. Manas; Kristi Fan; Andrea Olland; Rajiv Chopra; Menelas N. Pangalos; Peter Reinhart; Albert J. Robichaud

The proteolytic enzyme beta-secretase (BACE1) plays a central role in the synthesis of the pathogenic beta-amyloid in Alzheimers disease. Recently, we reported small molecule acylguanidines as potent BACE1 inhibitors. However, many of these acylguanidines have a high polar surface area (e.g. as measured by the topological polar surface area or TPSA), which is unfavorable for crossing the blood-brain barrier. Herein, we describe the identification of the 2-aminopyridine moiety as a bioisosteric replacement of the acylguanidine moiety, which resulted in inhibitors with lower TPSA values and superior brain penetration. X-ray crystallographic studies indicated that the 2-aminopyridine moiety interacts directly with the catalytic aspartic acids Asp32 and Asp228 via a hydrogen-bonding network.


Journal of Medicinal Chemistry | 2010

Identification and Characterization of Acidic Mammalian Chitinase Inhibitors

Derek Cecil Cole; Andrea Olland; Jaison Jacob; Jon Brooks; Matthew G. Bursavich; Robert M. Czerwinski; Charlene DeClercq; Mark R. Johnson; Diane Joseph-McCarthy; John W. Ellingboe; Laura Lin; Pawel Wojciech Nowak; Ella Presman; James Strand; Amy Tam; Cara Williams; Shihua Yao; Désirée H. H. Tsao; Lori Fitz

Acidic mammalian chitinase (AMCase) is a member of the glycosyl hydrolase 18 family (EC 3.2.1.14) that has been implicated in the pathophysiology of allergic airway disease such as asthma. Small molecule inhibitors of AMCase were identified using a combination of high-throughput screening, fragment screening, and virtual screening techniques and characterized by enzyme inhibition and NMR and Biacore binding experiments. X-ray structures of the inhibitors in complex with AMCase revealed that the larger more potent HTS hits, e.g. 5-(4-(2-(4-bromophenoxy)ethyl)piperazine-1-yl)-1H-1,2,4-triazol-3-amine 1, spanned from the active site pocket to a hydrophobic pocket. Smaller fragments identified by FBS occupy both these pockets independently and suggest potential strategies for linking fragments. Compound 1 is a 200 nM AMCase inhibitor which reduced AMCase enzymatic activity in the bronchoalveolar lavage fluid in allergen-challenged mice after oral dosing.


Bioorganic & Medicinal Chemistry Letters | 2010

4-(3-Aryloxyaryl)quinoline sulfones are potent liver X receptor agonists.

Ronald C. Bernotas; Robert R. Singhaus; David H. Kaufman; Jeremy M. Travins; John W. Ullrich; Rayomand J. Unwalla; Elaine Quinet; Mark J. Evans; Ponnal Nambi; Andrea Olland; Björn Kauppi; Anna Wilhelmsson; Annika Goos-Nilsson; Jay E. Wrobel

A series of 4-(3-aryloxyaryl)quinolines with sulfone substituents on the terminal aryl ring (7) was prepared as LXR agonists. High affinity LXR ligands with excellent agonist potency and efficacy in functional assays of LXR activity were identified. In general, these sulfone agonists were equal to or superior to previously described alcohol and amide analogs in terms of affinity, functional potency, and microsomal stability. Many of the sulfones had LXRbeta binding IC(50) values <10nM while the most potent compounds in an ABCA1 mRNA induction assay in J774 mouse cells had EC(50) values <10nM and were as efficacious as T0901317.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of a stable macrocyclic o-aminobenzamide Hsp90 inhibitor which significantly decreases tumor volume in a mouse xenograft model.

Christoph Wolfgang Zapf; Jonathan David Bloom; Zhong Li; Russell Dushin; Thomas Nittoli; Mercy Otteng; Antonia Nikitenko; Jennifer M. Golas; Hao Liu; Judy Lucas; Frank Boschelli; Erik Vogan; Andrea Olland; Mark Johnson; Jeremy I. Levin

An extension of our previously reported series of macrocyclic ortho-aminobenzamide Hsp90 inhibitors is reported. Addition of a second methyl group to the tether provided analogs that show increased potency in binding as well as cell-proliferation assays and, more importantly, are stable toward microsomes. We wish to disclose the discovery of a macrocycle which showed impressive biomarker activity 24-h post dosing and which demonstrated prolonged exposure in tumors. When studied in a lung cancer xenograft model, the compound demonstrated significant tumor size reduction.

Collaboration


Dive into the Andrea Olland's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yun Hu

Princeton University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Keith Douglas Barnes

Rensselaer Polytechnic Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge