I03-Macromolecular Crystallography
|
Frederick W.
Goldberg
,
Attilla K. T.
Ting
,
David
Beattie
,
Gillian M.
Lamont
,
Charlene
Fallan
,
M. Raymond V.
Finlay
,
Beth
Williamson
,
Marianne
Schimpl
,
Alexander R.
Harmer
,
Oladipupo B.
Adeyemi
,
Pär
Nordell
,
Anna S.
Cronin
,
Mercedes
Vazquez-Chantada
,
Derek
Barratt
,
Antonio
Ramos-Montoya
,
Elaine B.
Cadogan
,
Barry R.
Davies
Diamond Proposal Number(s):
[20015]
Abstract: The DNA-PK complex is activated by double-strand DNA breaks and regulates the non-homologous end-joining repair pathway; thus, targeting DNA-PK by inhibiting the DNA-PK catalytic subunit (DNA-PKcs) is potentially a useful therapeutic approach for oncology. A previously reported series of neutral DNA-PKcs inhibitors were modified to incorporate a basic group, with the rationale that increasing the volume of distribution while maintaining good metabolic stability should increase the half-life. However, adding a basic group introduced hERG activity, and basic compounds with modest hERG activity (IC50 = 10–15 μM) prolonged QTc (time from the start of the Q wave to the end of the T wave, corrected by heart rate) in an anaesthetized guinea pig cardiovascular model. Further optimization was necessary, including modulation of pKa, to identify compound 18, which combines low hERG activity (IC50 = 75 μM) with excellent kinome selectivity and favorable pharmacokinetic properties.
|
Jul 2022
|
|
I03-Macromolecular Crystallography
I04-1-Macromolecular Crystallography (fixed wavelength)
|
Jeffrey W.
Johannes
,
Amber
Balazs
,
Derek
Barratt
,
Michal
Bista
,
Matthew D.
Chuba
,
Sabina
Cosulich
,
Susan E.
Critchlow
,
Sébastien L.
Degorce
,
Paolo
Di Fruscia
,
Scott D.
Edmondson
,
Kevin
Embrey
,
Stephen
Fawell
,
Avipsa
Ghosh
,
Sonja J.
Gill
,
Anders
Gunnarsson
,
Sudhir M.
Hande
,
Tom D.
Heightman
,
Paul
Hemsley
,
Giuditta
Illuzzi
,
Jordan
Lane
,
Carrie
Larner
,
Elisabetta
Leo
,
Lina
Liu
,
Andrew
Madin
,
Scott
Martin
,
Lisa
Mcwilliams
,
Mark J.
O'Connor
,
Jonathan P.
Orme
,
Fiona
Pachl
,
Martin J.
Packer
,
Xiaohui
Pei
,
Andrew
Pike
,
Marianne
Schimpl
,
Hongyao
She
,
Anna D.
Staniszewska
,
Verity
Talbot
,
Elizabeth
Underwood
,
Jeffrey G.
Varnes
,
Lin
Xue
,
Tieguang
Yao
,
Ke
Zhang
,
Andrew X.
Zhang
,
Xiaolan
Zheng
Diamond Proposal Number(s):
[14631, 17180, 20015]
Abstract: Poly-ADP-ribose-polymerase (PARP) inhibitors have achieved regulatory approval in oncology for homologous recombination repair deficient tumors including BRCA mutation. However, some have failed in combination with first-line chemotherapies, usually due to overlapping hematological toxicities. Currently approved PARP inhibitors lack selectivity for PARP1 over PARP2 and some other 16 PARP family members, and we hypothesized that this could contribute to toxicity. Recent literature has demonstrated that PARP1 inhibition and PARP1–DNA trapping are key for driving efficacy in a BRCA mutant background. Herein, we describe the structure- and property-based design of 25 (AZD5305), a potent and selective PARP1 inhibitor and PARP1–DNA trapper with excellent in vivo efficacy in a BRCA mutant HBCx-17 PDX model. Compound 25 is highly selective for PARP1 over other PARP family members, with good secondary pharmacology and physicochemical properties and excellent pharmacokinetics in preclinical species, with reduced effects on human bone marrow progenitor cells in vitro.
|
Sep 2021
|
|
I03-Macromolecular Crystallography
|
William
Mccoull
,
Scott
Boyd
,
Martin R.
Brown
,
Muireann
Coen
,
Olga
Collingwood
,
Nichola L.
Davies
,
Ann
Doherty
,
Gary
Fairley
,
Kristin
Goldberg
,
Elizabeth
Hardaker
,
Guang
He
,
Edward J.
Hennessy
,
Philip
Hopcroft
,
George
Hodgson
,
Anne
Jackson
,
Xiefeng
Jiang
,
Ankur
Karmokar
,
Anne-Laure
Lainé
,
Nicola
Lindsay
,
Yumeng
Mao
,
Roshini
Markandu
,
Lindsay
Mcmurray
,
Neville
Mclean
,
Lorraine
Mooney
,
Helen
Musgrove
,
J. Willem M.
Nissink
,
Alexander
Pflug
,
Venkatesh Pilla
Reddy
,
Philip B.
Rawlins
,
Emma
Rivers
,
Marianne
Schimpl
,
Graham F.
Smith
,
Sharon
Tentarelli
,
Jon
Travers
,
Robert I.
Troup
,
Josephine
Walton
,
Cheng
Wang
,
Stephen
Wilkinson
,
Beth
Williamson
,
Jon
Winter-Holt
,
Dejian
Yang
,
Yuting
Zheng
,
Qianxiu
Zhu
,
Paul D.
Smith
Diamond Proposal Number(s):
[17180]
Abstract: Inhibition of Mer and Axl kinases has been implicated as a potential way to improve the efficacy of current immuno-oncology therapeutics by restoring the innate immune response in the tumor microenvironment. Highly selective dual Mer/Axl kinase inhibitors are required to validate this hypothesis. Starting from hits from a DNA-encoded library screen, we optimized an imidazo[1,2-a]pyridine series using structure-based compound design to improve potency and reduce lipophilicity, resulting in a highly selective in vivo probe compound 32. We demonstrated dose-dependent in vivo efficacy and target engagement in Mer- and Axl-dependent efficacy models using two structurally differentiated and selective dual Mer/Axl inhibitors. Additionally, in vivo efficacy was observed in a preclinical MC38 immuno-oncology model in combination with anti-PD1 antibodies and ionizing radiation.
|
Sep 2021
|
|
I04-1-Macromolecular Crystallography (fixed wavelength)
I04-Macromolecular Crystallography
|
J. Willem M.
Nissink
,
Sana
Bazzaz
,
Carolyn
Blackett
,
Matthew A.
Clark
,
Olga
Collingwood
,
Jeremy S.
Disch
,
Diana
Gikunju
,
Kristin
Goldberg
,
John P.
Guilinger
,
Elizabeth
Hardaker
,
Edward J.
Hennessy
,
Rachael
Jetson
,
Anthony D.
Keefe
,
William
Mccoull
,
Lindsay
Mcmurray
,
Allison
Olszewski
,
Ross
Overman
,
Alexander
Pflug
,
Marian
Preston
,
Philip B.
Rawlins
,
Emma
Rivers
,
Marianne
Schimpl
,
Paul
Smith
,
Caroline
Truman
,
Elizabeth
Underwood
,
Juli
Warwicker
,
Jon
Winter-Holt
,
Simon
Woodcock
,
Ying
Zhang
Abstract: Mer is a member of the TAM (Tyro3, Axl, Mer) kinase family that has been associated with cancer progression, metastasis, and drug resistance. Their essential function in immune homeostasis has prompted an interest in their role as modulators of antitumor immune response in the tumor microenvironment. Here we illustrate the outcomes of an extensive lead-generation campaign for identification of Mer inhibitors, focusing on the results from concurrent, orthogonal high-throughput screening approaches. Data mining, HT (high-throughput), and DECL (DNA-encoded chemical library) screens offered means to evaluate large numbers of compounds. We discuss campaign strategy and screening outcomes, and exemplify series resulting from prioritization of hits that were identified. Concurrent execution of HT and DECL screening successfully yielded a large number of potent, selective, and novel starting points, covering a range of selectivity profiles across the TAM family members and modes of kinase binding, and offered excellent start points for lead development.
|
Mar 2021
|
|
I03-Macromolecular Crystallography
I04-Macromolecular Crystallography
|
Tom E. H.
Ogden
,
Ji-Chun
Yang
,
Marianne
Schimpl
,
Laura E.
Easton
,
Elizabeth
Underwood
,
Philip b.
Rawlins
,
Michael m
Mccauley
,
Marie-France
Langelier
,
John m.
Pascal
,
Kevin j.
Embrey
,
David
Neuhaus
Diamond Proposal Number(s):
[17180, 20015]
Open Access
Abstract: PARP-1 is a key early responder to DNA damage in eukaryotic cells. An allosteric mechanism links initial sensing of DNA single-strand breaks by PARP-1’s F1 and F2 domains via a process of further domain assembly to activation of the catalytic domain (CAT); synthesis and attachment of poly(ADP-ribose) (PAR) chains to protein sidechains then signals for assembly of DNA repair components. A key component in transmission of the allosteric signal is the HD subdomain of CAT, which alone bridges between the assembled DNA-binding domains and the active site in the ART subdomain of CAT. Here we present a study of isolated CAT domain from human PARP-1, using NMR-based dynamics experiments to analyse WT apo-protein as well as a set of inhibitor complexes (with veliparib, olaparib, talazoparib and EB-47) and point mutants (L713F, L765A and L765F), together with new crystal structures of the free CAT domain and inhibitor complexes. Variations in both dynamics and structures amongst these species point to a model for full-length PARP-1 activation where first DNA binding and then substrate interaction successively destabilise the folded structure of the HD subdomain to the point where its steric blockade of the active site is released and PAR synthesis can proceed.
|
Jan 2021
|
|
I04-Macromolecular Crystallography
|
Diamond Proposal Number(s):
[17180]
Open Access
Abstract: Helicobacter pylori (H. pylori) uses several outer membrane proteins for adhering to its host's gastric mucosa, an important step in establishing and preserving colonization. Several adhesins (SabA, BabA, HopQ) have been characterized in terms of their three-dimensional structure. A recent addition to the growing list of outer membrane porins is LabA (LacdiNAc-binding adhesin), which is thought to bind specifically to GalNAcβ1-4GlcNAc, occurring in the gastric mucosa. LabA47-496 protein expressed as His-tagged protein in the periplasm of E. coli and purified via subtractive IMAC after TEV cleavage and subsequent size exclusion chromatography, resulted in bipyramidal crystals with good diffraction properties. Here, we describe the 2.06 Å resolution structure of the exodomain of LabA from H. pylori strain J99 (PDB ID: 6GMM). Strikingly, despite the relatively low levels of sequence identity with the other three structurally characterized adhesins (20–49%), LabA shares an L-shaped fold with SabA and BabA. The ‘head’ region contains a 4 + 3 α-helix bundle, with a small insertion domain consisting of a short antiparallel beta sheet and an unstructured region, not resolved in the crystal structure. Sequence alignment of LabA from different strains shows a high level of conservation in the N- and C-termini, and identifies two main types based on the length of the insertion domain (‘crown’ region), the ‘J99-type’ (insertion ~31 amino acids), and the H. pylori ‘26695 type’ (insertion ~46 amino acids). Analysis of ligand binding using Native Electrospray Ionization Mass Spectrometry (ESI-MS) together with solid phase-bound, ELISA-type assays could not confirm the originally described binding of GalNAcβ1-4GlcNAc-containing oligosaccharides, in line with other recent reports, which also failed to confirm LacdiNAc binding.
|
Dec 2020
|
|
I02-Macromolecular Crystallography
I03-Macromolecular Crystallography
I04-Macromolecular Crystallography
|
Diamond Proposal Number(s):
[14631, 17180, 20015]
Open Access
Abstract: The activation loop (A-loop) plays a key role in regulating the catalytic activity of protein kinases. Phosphorylation in this region enhances the phosphoryl transfer rate of the kinase domain and increases its affinity for ATP. Furthermore, the A-loop possesses autoinhibitory functions in some kinases, where it collapses onto the protein surface and blocks substrate binding when unphosphorylated. Due to its flexible nature, the A-loop is usually disordered and untraceable in kinase domain crystal structures. The resulting lack of structural information is regrettable as it impedes the design of drug A-loop contacts, which have proven favourable in multiple cases. Here we characterize the binding with A-loop engagement between type 1.5 kinase inhibitor ‘example 172’ (EX172) and Mer tyrosine kinase (MerTK). With the help of crystal structures and binding kinetics we portray how the recruitment of the A-loop elicits a two-step binding mechanism which results in a drug-target complex characterized by high affinity and long residence time. In addition, the type 1.5 compound possesses excellent kinome selectivity and a remarkable preference for the phosphorylated over the dephosphorylated form of MerTK. We discuss these unique characteristics in the context of known type 1 and type 2 inhibitors and highlight opportunities for future kinase inhibitor design.
|
Oct 2020
|
|
I03-Macromolecular Crystallography
I04-Macromolecular Crystallography
|
Kirsten
Mcaulay
,
Emily A.
Hoyt
,
Morgan
Thomas
,
Marianne
Schimpl
,
Michael S.
Bodnarchuk
,
Hilary J.
Lewis
,
Derek
Barratt
,
Deepa
Bhavsar
,
David M.
Robinson
,
Michael J.
Deery
,
Derek
Ogg
,
Gonçalo J. I.
Bernardes
,
Richard A.
Ward
,
Michael J.
Waring
,
Jason G.
Kettle
Diamond Proposal Number(s):
[12419, 20015]
Abstract: With a resurgence in interest in covalent drugs, there is need to identify new moieties capable of cysteine bond formation that are differentiated from commonly employed systems such as acrylamide. Herein, we report on the discovery of new alkynyl benzoxazine and dihydroquinazoline moieties capable of covalent reaction with cysteine. Their utility as alternative electrophilic warheads for chemical biological probes and drug molecules is demonstrated through site-selective protein modification and incorporation into kinase drug scaffolds. A potent covalent inhibitor of JAK3 kinase was identified with superior selectivity across the kinome and improvements in in vitro pharmacokinetic profile relative to the related acrylamide-based inhibitor. In addition, the use of a novel heterocycle as cysteine reactive warhead is employed to target Cys788 in c-KIT where acrylamide has previously failed to form covalent interactions. These new reactive and selective heterocyclic warheads supplement the current repertoire for cysteine covalent modification whilst avoiding some of the limitations generally associated with established moieties.
|
May 2020
|
|
I03-Macromolecular Crystallography
|
Frederick Woolf
Goldberg
,
M. Raymond V.
Finlay
,
Attilla
Ting
,
David
Beattie
,
Gillian
Lamont
,
Charlene
Fallan
,
Gail
Wrigley
,
Marianne
Schimpl
,
Martin R.
Howard
,
Beth
Williamson
,
Mercedes
Vazquez-Chantada
,
Derek
Barratt
,
Barry
Davies
,
Elaine
Cadogan
,
Antonio
Ramos Montoya
,
Emma
Dean
Abstract: DNA-PK is a key component within the DNA damage response, as it is responsible for recognizing and repairing double-strand DNA breaks (DSBs) via non-homologous end joining. Historically it has been challenging to identify inhibitors of the DNA-PK catalytic subunit (DNA-PKcs) with good selectivity versus the structurally related PI3 (lipid) and PI3K-related protein kinases. We screened our corporate collection for DNA-PKcs inhibitors with good PI3 kinase selectivity, identifying compound 1. Optimization focused on further improving selectivity whilst improving physical and pharmacokinetic properties, notably co-optimization of permeability and metabolic stability, to identify compound 16 (AZD7648). Compound 16 had no significant off-targets in the protein kinome, and only weak activity versus PI3Kα/γ lipid kinases. Monotherapy activity in murine xenograft models was observed, and regressions were observed when combined with inducers of DSBs (doxorubicin or irradiation) or PARP inhibition (olaparib). These data support progression into clinical studies (NCT03907969).
|
Dec 2019
|
|
I02-Macromolecular Crystallography
I04-1-Macromolecular Crystallography (fixed wavelength)
|
Jason G.
Kettle
,
Rana
Anjum
,
Evan
Barry
,
Deepa
Bhavsar
,
Crystal
Brown
,
Scott
Boyd
,
Andrew
Campbell
,
Kristin
Goldberg
,
Michael
Grondine
,
Sylvie
Guichard
,
Christopher J.
Hardy
,
Tom
Hunt
,
Rhys D. O.
Jones
,
Xiuwei
Li
,
Olga
Moleva
,
Derek
Ogg
,
Ross C.
Overman
,
Martin J.
Packer
,
Stuart
Pearson
,
Marianne
Schimpl
,
Wenlin
Shao
,
Aaron
Smith
,
James M.
Smith
,
Darren
Stead
,
Steve
Stokes
,
Michael
Tucker
,
Yang
Ye
Diamond Proposal Number(s):
[12419, 14631]
Abstract: While the treatment of gastrointestinal stromal tumors (GISTs) has been revolutionized by the application of targeted tyrosine kinase inhibitors capable of inhibiting KIT-driven proliferation, diverse mutations to this kinase drive resistance to established therapies. Here we describe the identification of potent pan-KIT mutant kinase inhibitors that can be dosed without being limited by the tolerability issues seen with multitargeted agents. This effort focused on identification and optimization of an existing kinase scaffold through the use of structure-based design. Starting from a series of previously reported phenoxyquinazoline and quinoline based inhibitors of the tyrosine kinase PDGFRα, potency against a diverse panel of mutant KIT driven Ba/F3 cell lines was optimized, with a particular focus on reducing activity against a KDR driven cell model in order to limit the potential for hypertension commonly seen in second and third line GIST therapies. AZD3229 demonstrates potent single digit nM growth inhibition across a broad cell panel, with good margin to KDR-driven effects. Selectivity over KDR can be rationalized predominantly by the interaction of water molecules with the protein and ligand in the active site, and its kinome selectivity is similar to the best of the approved GIST agents. This compound demonstrates excellent cross-species pharmacokinetics, shows strong pharmacodynamic inhibition of target, and is active in several in vivo models of GIST.
|
Sep 2018
|
|