I04-1-Macromolecular Crystallography (fixed wavelength)
I04-Macromolecular Crystallography
|
Sabrina R.
Mackinnon
,
Tobias
Krojer
,
William R.
Foster
,
Laura
Diaz-Saez
,
Manshu
Tang
,
Kilian V. M.
Huber
,
Frank
Von Delft
,
Kent
Lai
,
Paul
Brennan
,
Gustavo
Arruda Bezerra
,
Wyatt W.
Yue
Diamond Proposal Number(s):
[18145]
Abstract: Classic galactosemia is caused by loss-of-function mutations in
galactose-1-phosphate uridylyltransferase (GALT) that lead to toxic
accumulation of its substrate, galactose-1-phosphate. One proposed therapy
is to inhibit the biosynthesis of galactose-1-phosphate, catalyzed by
galactokinase 1 (GALK1). Existing inhibitors of human GALK1 (hGALK1)
are primarily ATP-competitive with limited clinical utility to date. Here, we
determined crystal structures of hGALK1 bound with reported ATP-
competitive inhibitors of the spiro-benzoxazole series, to reveal their binding
mode in the active site. Spurred by the need for additional chemotypes of
hGALK1 inhibitors, desirably targeting a nonorthosteric site, we also
performed crystallography-based screening by soaking hundreds of hGALK1
crystals, already containing active site ligands, with fragments from a custom library. Two fragments were found to bind close to the ATP binding site, and a further eight were found in a hotspot distal from the active site, highlighting the strength of this method in identifying previously uncharacterized allosteric sites. To generate inhibitors of improved potency and selectivity targeting the newly identified binding hotspot, new compounds were designed by merging overlapping fragments. This yielded two micromolar inhibitors of hGALK1 that were not competitive with respect to either substrate (ATP or galactose) and demonstrated good selectivity over hGALK1 homologues, galactokinase 2 and mevalonate kinase. Our findings are therefore the first to demonstrate inhibition of hGALK1 from an allosteric site, with potential for further development of potent and selective inhibitors to provide novel therapeutics for classic galactosemia.
|
Mar 2021
|
|
I03-Macromolecular Crystallography
I24-Microfocus Macromolecular Crystallography
|
Sarah L.
Kidd
,
Elaine
Fowler
,
Till
Reinhardt
,
Thomas
Compton
,
Natalia
Mateu
,
Hector
Newman
,
Dom
Bellini
,
Romain
Talon
,
Joseph
Mcloughlin
,
Tobias
Krojer
,
Anthony
Aimon
,
Anthony
Bradley
,
Michael
Fairhead
,
Paul
Brear
,
Laura
Diaz-Saez
,
Katherine
Mcauley
,
Hannah F.
Sore
,
Andrew
Madin
,
Daniel H.
O'Donovan
,
Kilian
Huber
,
Marko
Hyvonen
,
Frank
Von Delft
,
Christopher G.
Dowson
,
David R.
Spring
Diamond Proposal Number(s):
[18145, 15649, 14303, 14493]
Open Access
Abstract: Organic synthesis underpins the evolution of weak fragment hits into potent lead compounds. Deficiencies within current screening collections often result in the requirement of significant synthetic investment to enable multidirectional fragment growth, limiting the efficiency of the hit evolution process. Diversity-oriented synthesis (DOS)-derived fragment libraries are constructed in an efficient and modular fashion and thus are well-suited to address this challenge. To demonstrate the effective nature of such libraries within fragment-based drug discovery, we herein describe the screening of a 40-member DOS library against three functionally distinct biological targets using X-Ray crystallography. Firstly, we demonstrate the importance for diversity in aiding hit identification with four fragment binders resulting from these efforts. Moreover, we also exemplify the ability to readily access a library of analogues from cheap commercially available materials, which ultimately enabled the exploration of a minimum of four synthetic vectors from each molecule. In total, 10–14 analogues of each hit were rapidly accessed in three to six synthetic steps. Thus, we showcase how DOS-derived fragment libraries enable efficient hit derivatisation and can be utilised to remove the synthetic limitations encountered in early stage fragment-based drug discovery.
|
May 2020
|
|
I02-Macromolecular Crystallography
I03-Macromolecular Crystallography
I24-Microfocus Macromolecular Crystallography
|
Vincent
Fagan
,
Catrine
Johansson
,
Carina
Gileadi
,
Octovia
Monteiro
,
James Edward
Dunford
,
Reshma
Nibhani
,
Martin
Philpott
,
Jessica
Malzahn
,
Graham
Wells
,
Ruth
Farham
,
Adam
Cribbs
,
Nadia
Halidi
,
Fengling
Li
,
Irene
Chau
,
Holger
Greschik
,
Srikannathasan
Velupillai
,
Abdellah
Allali-Hassani
,
James M.
Bennett
,
Thomas
Christott
,
Charline
Giroud
,
Andrew M.
Lewis
,
Kilian V. M.
Huber
,
Nick
Athanasou
,
Chas
Bountra
,
Manfred
Jung
,
Roland
Schüle
,
Masoud
Vedadi
,
Cheryl H.
Arrowsmith
,
Yan
Xiong
,
Jian
Jin
,
Oleg
Fedorov
,
Gillian
Farnie
,
Paul E.
Brennan
,
Udo C. T.
Oppermann
Diamond Proposal Number(s):
[10619, 15433]
Abstract: Modifications of histone tails, including lysine/arginine methylation, provide the basis of a 'chromatin or histone code'. Proteins that con-tain 'reader' domains can bind to these modifications and form specific effector complexes, which ultimately mediate chromatin function. The spindlin1 (SPIN1) protein contains three Tudor methyl-lysine/arginine reader domains and was identified as a putative onco-gene and transcriptional co-activator. Here we report a SPIN1 chemi-cal probe inhibitor with low nanomolar in vitro activity, exquisite selectivity on a panel of methyl reader and writer proteins, and with submicromolar cellular activity. X-ray crystallography showed that this Tudor domain chemical probe simultaneously engages Tudor domains 1 and 2 via a bidentate binding mode. Small molecule inhibition and siRNA knockdown of SPIN1, as well as chemoproteomic studies, iden-tified genes which are transcriptionally regulated by SPIN1 in squa-mous cell carcinoma and suggest that SPIN1 may have a roll in cancer related inflammation and/or cancer metastasis.
|
Sep 2019
|
|
I03-Macromolecular Crystallography
I04-1-Macromolecular Crystallography (fixed wavelength)
|
Efrat
Resnick
,
Anthony
Bradley
,
Jinrui
Gan
,
Alice
Douangamath
,
Tobias
Krojer
,
Ritika
Sethi
,
Paul P.
Geurink
,
Anthony
Aimon
,
Gabriel
Amitai
,
Dom
Bellini
,
James
Bennett
,
Michael
Fairhead
,
Oleg
Fedorov
,
Ronen
Gabizon
,
Jin
Gan
,
Jingxu
Guo
,
Alexander
Plotnikov
,
Nava
Reznik
,
Gian Filippo
Ruda
,
Laura
Diaz-Saez
,
Verena M.
Straub
,
Tamas
Szommer
,
Srikannathasan
Velupillai
,
Daniel
Zaidman
,
Yanling
Zhang
,
Alun R.
Coker
,
Christopher G.
Dowson
,
Haim
Barr
,
Chu
Wang
,
Kilian V. M.
Huber
,
Paul E.
Brennan
,
Huib
Ovaa
,
Frank
Von Delft
,
Nir
London
Abstract: Covalent probes can display unmatched potency, selectivity and duration of action; however, their discovery is challenging. In principle, fragments that can irreversibly bind their target can overcome the low affinity that limits reversible fragment screening, but such electrophilic fragments were considered non-selective and were rarely screened. We hypothesized that mild electrophiles might overcome the selectivity challenge and constructed a library of 993 mildly electrophilic fragments. We characterized this library by a new high-throughput thiol-reactivity assay and screened them against ten cysteine-containing proteins. Highly reactive and promiscuous fragments were rare and could be easily eliminated. By contrast, we found hits for most targets. Combining our approach with high-throughput crystallography allowed rapid progression to potent and selective probes for two enzymes, the deubiquitinase OTUB2 and the pyrophosphatase NUDT7. No inhibitors were previously known for either. This study highlights the potential of electrophile-fragment screening as a practical and efficient tool for covalent-ligand discovery.
|
May 2019
|
|
I04-Macromolecular Crystallography
|
Saleta
Vazquez-Rodriguez
,
Miranda
Wright
,
Catherine M.
Rogers
,
Adam P.
Cribbs
,
Srikannathasan
Velupillai
,
Martin
Philpott
,
Henry
Lee
,
James E.
Dunford
,
Kilian V. M.
Huber
,
Matthew B.
Robers
,
James D.
Vasta
,
Marie-Laetitia
Thezenas
,
Sarah
Bonham
,
Benedikt
Kessler
,
James
Bennett
,
Oleg
Fedorov
,
Florence
Raynaud
,
Adam
Donovan
,
Julian
Blagg
,
Vassilios
Bavetsias
,
Udo
Oppermann
,
Chas
Bountra
,
Akane
Kawamura
,
Paul E.
Brennan
Diamond Proposal Number(s):
[15433]
Open Access
Abstract: Histone lysine demethylases (KDMs) are involved in the dynamic regulation of gene expression and they play a critical role in several biological processes. Achieving selectivity over the different KDMs has been a major challenge for KDM inhibitor development. Here we report potent and selective KDM5 covalent inhibitors designed to target cysteine residues only present in the KDM5 sub‐family. The covalent binding to the targeted proteins was confirmed by MS and time‐dependent inhibition. Additional competition assays show that compounds were non 2‐OG competitive. Target engagement and ChIP‐seq analysis showed that the compounds inhibited the KDM5 members in cells at nano‐ to micromolar levels and induce a global increase of the H3K4me3 mark at transcriptional start sites.
|
Jan 2019
|
|
I04-Macromolecular Crystallography
|
Matous
Hrdinka
,
Lisa
Schlicher
,
Bing
Dai
,
Daniel M.
Pinkas
,
Joshua C.
Bufton
,
Sarah
Picaud
,
Jennifer A.
Ward
,
Catherine
Rogers
,
Chalada
Suebsuwong
,
Sameer
Nikhar
,
Gregory D.
Cuny
,
Kilian V. M.
Huber
,
Panagis
Filippakopoulos
,
Alex N.
Bullock
,
Alexei
Degterev
,
Mads
Gyrd‐hansen
Diamond Proposal Number(s):
[15433]
Open Access
Abstract: RIPK2 mediates inflammatory signaling by the bacteria‐sensing receptors NOD1 and NOD2. Kinase inhibitors targeting RIPK2 are a proposed strategy to ameliorate NOD‐mediated pathologies. Here, we reveal that RIPK2 kinase activity is dispensable for NOD2 inflammatory signaling and show that RIPK2 inhibitors function instead by antagonizing XIAP‐binding and XIAP‐mediated ubiquitination of RIPK2. We map the XIAP binding site on RIPK2 to the loop between β2 and β3 of the N‐lobe of the kinase, which is in close proximity to the ATP‐binding pocket. Through characterization of a new series of ATP pocket‐binding RIPK2 inhibitors, we identify the molecular features that determine their inhibition of both the RIPK2‐XIAP interaction, and of cellular and in vivoNOD2 signaling. Our study exemplifies how targeting of the ATP‐binding pocket in RIPK2 can be exploited to interfere with the RIPK2‐XIAP interaction for modulation of NOD signaling.
|
Jul 2018
|
|
I02-Macromolecular Crystallography
|
Léa
Bouché
,
Clara D.
Christ
,
Stephan
Siegel
,
Amaury E.
Fernández-Montalván
,
Simon J.
Holton
,
Oleg
Fedorov
,
Antonius
Ter Laak
,
Tatsuo
Sugawara
,
Detlef
Stöckigt
,
Cynthia
Tallant
,
James
Bennett
,
Octovia
Monteiro
,
Laura
Díaz-Sáez
,
Paulina
Siejka
,
Julia
Meier
,
Vera
Pütter
,
Jörg
Weiske
,
Susanne
Müller
,
Kilian V. M.
Huber
,
Ingo V.
Hartung
,
Bernard
Haendler
Diamond Proposal Number(s):
[15558, 10619]
Abstract: Bromodomains (BD) are readers of lysine acetylation marks present in numerous proteins associated with chromatin. Here we describe a dual inhibitor of the bromodomain and PHD finger (BRPF) family member BRPF2 and the TATA box binding protein-associated factors TAF1 and TAF1L. These proteins are found in large chromatin complexes and play important roles in transcription regulation. The substituted benzoisoquinolinedione series was identified by high-throughput screening, and subsequent structure–activity relationship optimization allowed generation of low nanomolar BRPF2 BD inhibitors with strong selectivity against BRPF1 and BRPF3 BDs. In addition, a strong inhibition of TAF1/TAF1L BD2 was measured for most derivatives. The best compound of the series was BAY-299, which is a very potent, dual inhibitor with an IC50 of 67 nM for BRPF2 BD, 8 nM for TAF1 BD2, and 106 nM for TAF1L BD2. Importantly, no activity was measured for BRD4 BDs. Furthermore, cellular activity was evidenced using a BRPF2– or TAF1–histone H3.3 or H4 interaction assay.
|
May 2017
|
|
I02-Macromolecular Crystallography
I04-1-Macromolecular Crystallography (fixed wavelength)
|
Kilian V. M.
Huber
,
Eidarus
Salah
,
Branka
Radic
,
Manuela
Gridling
,
Jonathan M.
Elkins
,
Alexey
Stukalov
,
Ann-Sofie
Jemth
,
Camilla
Göktürk
,
Kumar
Sanjiv
,
Kia
Strömberg
,
Therese
Pham
,
Ulrika Warpman
Berglund
,
Jacques
Colinge
,
Keiryn L.
Bennett
,
Joanna I.
Loizou
,
Thomas
Helleday
,
Stefan
Knapp
,
Giulio
Superti-Furga
Diamond Proposal Number(s):
[8421]
Abstract: Activated RAS GTPase signalling is a critical driver of oncogenic transformation and malignant disease. Cellular models of RAS-dependent cancers have been used to identify experimental small molecules, such as SCH51344, but their molecular mechanism of action remains generally unknown. Here, using a chemical proteomic approach, we identify the target of SCH51344 as the human mutT homologue MTH1 (also known as NUDT1), a nucleotide pool sanitizing enzyme. Loss-of-function of MTH1 impaired growth of KRAS tumour cells, whereas MTH1 overexpression mitigated sensitivity towards SCH51344. Searching for more drug-like inhibitors, we identified the kinase inhibitor crizotinib as a nanomolar suppressor of MTH1 activity. Surprisingly, the clinically used (R)-enantiomer of the drug was inactive, whereas the (S)-enantiomer selectively inhibited MTH1 catalytic activity. Enzymatic assays, chemical proteomic profiling, kinome-wide activity surveys and MTH1 co-crystal structures of both enantiomers provide a rationale for this remarkable stereospecificity. Disruption of nucleotide pool homeostasis via MTH1 inhibition by (S)-crizotinib induced an increase in DNA single-strand breaks, activated DNA repair in human colon carcinoma cells, and effectively suppressed tumour growth in animal models. Our results propose (S)-crizotinib as an attractive chemical entity for further pre-clinical evaluation, and small-molecule inhibitors of MTH1 in general as a promising novel class of anticancer agents.
|
Apr 2014
|
|
I02-Macromolecular Crystallography
|
Oleg
Fedorov
,
Kilian
Huber
,
Andreas
Eisenreich
,
Panagis
Filippakopoulos
,
Oliver
King
,
Alex N.
Bullock
,
Damian
Szklarczyk
,
Lars J.
Jensen
,
Doriano
Fabbro
,
Jörg
Trappe
,
Ursula
Rauch
,
Franz
Bracher
,
Stefan
Knapp
Open Access
Abstract: There is a growing recognition of the importance of protein kinases in the control of alternative splicing. To define the underlying regulatory mechanisms, highly selective inhibitors are needed. Here, we report the discovery and characterization of the dichloroindolyl enaminonitrile KH-CB19, a potent and highly specific inhibitor of the CDC2-like kinase isoforms 1 and 4 (CLK1/CLK4). Cocrystal structures of KH-CB19 with CLK1 and CLK3 revealed a non-ATP mimetic binding mode, conformational changes in helix αC and the phosphate binding loop and halogen bonding to the kinase hinge region. KH-CB19 effectively suppressed phosphorylation of SR (serine/arginine) proteins in cells, consistent with its expected mechanism of action. Chemical inhibition of CLK1/CLK4 generated a unique pattern of splicing factor dephosphorylation and had at low nM concentration a profound effect on splicing of the two tissue factor isoforms flTF (full-length TF) and asHTF (alternatively spliced human TF).
|
Jan 2011
|
|