|
William
Mccorkindale
,
Kadi L.
Saar
,
Daren
Fearon
,
Melissa
Boby
,
Haim
Barr
,
Amir
Ben-Shmuel
,
Nir
London
,
Frank
Von Delft
,
John D.
Chodera
,
Alpha. A.
Lee
,
The
Covid Moonshot Consortium
Open Access
Abstract: A common challenge in drug design pertains to finding chemical modifications to a ligand that increases its affinity to the target protein. An underutilized advance is the increase in structural biology throughput, which has progressed from an artisanal endeavor to a monthly throughput of hundreds of different ligands against a protein in modern synchrotrons. However, the missing piece is a framework that turns high-throughput crystallography data into predictive models for ligand design. Here, we designed a simple machine learning approach that predicts protein–ligand affinity from experimental structures of diverse ligands against a single protein paired with biochemical measurements. Our key insight is using physics-based energy descriptors to represent protein–ligand complexes and a learning-to-rank approach that infers the relevant differences between binding modes. We ran a high-throughput crystallography campaign against the SARS-CoV-2 main protease (MPro), obtaining parallel measurements of over 200 protein–ligand complexes and their binding activities. This allows us to design one-step library syntheses which improved the potency of two distinct micromolar hits by over 10-fold, arriving at a noncovalent and nonpeptidomimetic inhibitor with 120 nM antiviral efficacy. Crucially, our approach successfully extends ligands to unexplored regions of the binding pocket, executing large and fruitful moves in chemical space with simple chemistry.
|
Mar 2023
|
|
I03-Macromolecular Crystallography
|
Diamond Proposal Number(s):
[23459]
Open Access
Abstract: The worldwide public health and socioeconomic consequences caused by the COVID-19 pandemic highlight the importance of increasing preparedness for viral disease outbreaks by providing rapid disease prevention and treatment strategies. The NSP3 macrodomain of coronaviruses including SARS-CoV-2 is among the viral protein repertoire that was identified as a potential target for the development of antiviral agents, due to its critical role in viral replication and consequent pathogenicity in the host. By combining virtual and biophysical screening efforts, we discovered several experimental small molecules and FDA-approved drugs as inhibitors of the NSP3 macrodomain. Analogue characterisation of the hit matter and crystallographic studies confirming binding modes, including that of the antibiotic compound aztreonam, to the active site of the macrodomain provide valuable structure–activity relationship information that support current approaches and open up new avenues for NSP3 macrodomain inhibitor development.
|
Feb 2023
|
|
|
Stefan
Gahbauer
,
Galen J.
Correy
,
Marion
Schuller
,
Matteo P.
Ferla
,
Yagmur Umay
Doruk
,
Moira
Rachman
,
Taiasean
Wu
,
Morgan
Diolaiti
,
Siyi
Wang
,
R. Jeffrey
Neitz
,
Daren
Fearon
,
Dmytro S.
Radchenko
,
Yurii S.
Moroz
,
John J.
Irwin
,
Adam R.
Renslo
,
Jenny C.
Taylor
,
Jason E.
Gestwicki
,
Frank
Von Delft
,
Alan
Ashworth
,
Ivan
Ahel
,
Brian K.
Shoichet
,
James S.
Fraser
Open Access
Abstract: The nonstructural protein 3 (NSP3) of the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) contains a conserved macrodomain enzyme (Mac1) that is critical for pathogenesis and lethality. While small-molecule inhibitors of Mac1 have great therapeutic potential, at the outset of the COVID-19 pandemic, there were no well-validated inhibitors for this protein nor, indeed, the macrodomain enzyme family, making this target a pharmacological orphan. Here, we report the structure-based discovery and development of several different chemical scaffolds exhibiting low- to sub-micromolar affinity for Mac1 through iterations of computer-aided design, structural characterization by ultra-high-resolution protein crystallography, and binding evaluation. Potent scaffolds were designed with in silico fragment linkage and by ultra-large library docking of over 450 million molecules. Both techniques leverage the computational exploration of tangible chemical space and are applicable to other pharmacological orphans. Overall, 160 ligands in 119 different scaffolds were discovered, and 153 Mac1-ligand complex crystal structures were determined, typically to 1 Å resolution or better. Our analyses discovered selective and cell-permeable molecules, unexpected ligand-mediated conformational changes within the active site, and key inhibitor motifs that will template future drug development against Mac1.
|
Jan 2023
|
|
I04-1-Macromolecular Crystallography (fixed wavelength)
|
Open Access
Abstract: A library of thiazoles and selenothiazoles were synthesized via Ir-catalyzed ylide insertion chemistry. This process is a functional group, particularly heterocycle-substituent tolerant. This was applied to the synthesis of fanetizole, an anti-inflammatory drug, and a thiazole-containing drug fragment that binds to the peptidyl-tRNA hydrolase (Pth) in Neisseria gonorrheae bacteria.
|
Nov 2022
|
|
I04-1-Macromolecular Crystallography (fixed wavelength)
I04-Macromolecular Crystallography
|
Diamond Proposal Number(s):
[16816, 18582, 19190, 25586, 30588]
Open Access
Abstract: Polyomaviruses are a family of ubiquitous double-stranded DNA viruses many of which are human pathogens. These include BK polyomavirus which causes severe urinary tract infection in immunocompromised patients and Merkel cell polyomavirus associated with aggressive cancers. The small genome of polyomaviruses lacks conventional drug targets, and no specific drugs are available at present. Here we focus on the main structural protein VP1 of BK polyomavirus which is responsible for icosahedral capsid formation. To provide a foundation towards rational drug design, we crystallized truncated VP1 pentamers and subjected them to a high-throughput screening for binding drug-like fragments through a direct X-ray analysis. To enable a highly performant screening, rigorous optimization of the crystallographic pipeline and processing with the latest generation PanDDA2 software were necessary. As a result, a total of 144 binding hits were established. Importantly, the hits are well clustered in six surface pockets. Three pockets are located on the outside of the pentamer and map on the regions where the ‘invading’ C-terminal arm of another pentamer is attached upon capsid assembly. Another set of three pockets is situated within the wide pore along the five-fold axis of the VP1 pentamer. These pockets are situated at the interaction interface with the minor capsid protein VP2 which is indispensable for normal functioning of the virus. Here we systematically analyse the three outside pockets which are highly conserved across various polyomaviruses, while point mutations in these pockets are detrimental for viral replication. We show that one of the pockets can accommodate antipsychotic drug trifluoperazine. For each pocket, we derive pharmacophore features which enable the design of small molecules preventing the interaction between VP1 pentamers and therefore inhibiting capsid assembly. Our data lay a foundation towards a rational development of first-in-class drugs targeting polyomavirus capsid.
|
May 2022
|
|
I03-Macromolecular Crystallography
|
Arathy
Jose
,
Daniel
Guest
,
Remi
Legay
,
Graham J.
Tizzard
,
Simon
Coles
,
Mariliza
Derveni
,
Edward
Wright
,
Lester
Marrison
,
Alpha A.
Lee
,
Aaron
Morris
,
Matt
Robinson
,
Frank
Von Delft
,
Daren
Fearon
,
Lizbe
Koekemoer
,
Tetiana
Matviuk
,
Anthony
Aimon
,
Christopher J.
Schofield
,
Tika R.
Malla
,
Nir
London
,
Barnaby W.
Greenland
,
Mark C.
Bagley
,
John
Spencer
Diamond Proposal Number(s):
[19301]
Open Access
Abstract: The pentafluorosulfanyl (-SF5) functional group is of increasing interest as a bioisostere in medicinal chemistry. A library of SF5-containing compounds, including amide, isoxazole, and oxindole derivatives, was synthesised using a range of solution-based and solventless methods, including microwave and ball-mill techniques. The library was tested against targets including human dihydroorotate dehydrogenase (HDHODH). A subsequent focused approach led to synthesis of analogues of the clinically used disease modifying anti-rheumatic drugs (DMARDs), Teriflunomide and Leflunomide, considered for potential COVID-19 use, where SF5 bioisostere deployment led to improved inhibition of HDHODH compared with the parent drugs. The results demonstrate the utility of the SF5 group in medicinal chemistry.
|
Feb 2022
|
|
I04-1-Macromolecular Crystallography (fixed wavelength)
|
Joseph A.
Newman
,
Alice
Douangamath
,
Setayesh
Yadzani
,
Yuliana
Yosaatmadja
,
Anthony
Aimon
,
Jose
Brandao-Neto
,
Louise
Dunnett
,
Tyler
Gorrie-Stone
,
Rachael
Skyner
,
Daren
Fearon
,
Matthieu
Schapira
,
Frank
Von Delft
,
Opher
Gileadi
Diamond Proposal Number(s):
[26998]
Open Access
Abstract: There is currently a lack of effective drugs to treat people infected with SARS-CoV-2, the cause of the global COVID-19 pandemic. The SARS-CoV-2 Non-structural protein 13 (NSP13) has been identified as a target for anti-virals due to its high sequence conservation and essential role in viral replication. Structural analysis reveals two “druggable” pockets on NSP13 that are among the most conserved sites in the entire SARS-CoV-2 proteome. Here we present crystal structures of SARS-CoV-2 NSP13 solved in the APO form and in the presence of both phosphate and a non-hydrolysable ATP analog. Comparisons of these structures reveal details of conformational changes that provide insights into the helicase mechanism and possible modes of inhibition. To identify starting points for drug development we have performed a crystallographic fragment screen against NSP13. The screen reveals 65 fragment hits across 52 datasets opening the way to structure guided development of novel antiviral agents.
|
Aug 2021
|
|
I04-1-Macromolecular Crystallography (fixed wavelength)
|
G. D.
Noske
,
A. M.
Nakamura
,
V. O.
Gawriljuk
,
R. S.
Fernandes
,
G.
M. A. Lima
,
H. V. D.
Rosa
,
H. D.
Pereira
,
A. C. M.
Zeri
,
A. A. F. Z.
Nascimento
,
M. C. L. C.
Freire
,
D.
Fearon
,
A.
Douangamath
,
F.
Von Delft
,
G.
Oliva
,
A. S.
Godoy
Diamond Proposal Number(s):
[27963]
Abstract: SARS-CoV-2 is the causative agent of COVID-19. The dimeric form of the viral Mpro is responsible for the cleavage of the viral polyprotein in 11 sites, including its own N and C-terminus. The lack of structural information for intermediary forms of Mpro is a setback for the understanding its self-maturation process. Herein, we used X-ray crystallography combined with biochemical data to characterize multiple forms of SARS-CoV-2 Mpro. For the immature form, we show that extra N-terminal residues caused conformational changes in the positioning of domain-three over the active site, hampering the dimerization and diminishing its activity. We propose that this form preludes the cis and trans-cleavage of N-terminal residues. Using fragment screening, we probe new cavities in this form which can be used to guide therapeutic development. Furthermore, we characterized a serine site-directed mutant of the Mpro bound to its endogenous N and C-terminal residues during dimeric association stage of the maturation process. We suggest this form is a transitional state during the C-terminal trans-cleavage. This data sheds light in the structural modifications of the SARS-CoV-2 main protease during its self-maturation process.
|
Jun 2021
|
|
I04-1-Macromolecular Crystallography (fixed wavelength)
|
Daniel
Zaidman
,
Paul
Gehrtz
,
Mihajlo
Filep
,
Daren
Fearon
,
Ronen
Gabizon
,
Alice
Douangamath
,
Jaime
Prilusky
,
Shirly
Duberstein
,
Galit
Cohen
,
C. David
Owen
,
Efrat
Resnick
,
Claire
Strain-Damerell
,
Petra
Lukacik
,
Haim
Barr
,
Martin A.
Walsh
,
Frank
Von Delft
,
Nir
London
Diamond Proposal Number(s):
[18145, 27963]
Abstract: Designing covalent inhibitors is increasingly important, although it remains challenging. Here, we present covalentizer, a computational pipeline for identifying irreversible inhibitors based on structures of targets with non-covalent binders. Through covalent docking of tailored focused libraries, we identify candidates that can bind covalently to a nearby cysteine while preserving the interactions of the original molecule. We found ∼11,000 cysteines proximal to a ligand across 8,386 complexes in the PDB. Of these, the protocol identified 1,553 structures with covalent predictions. In a prospective evaluation, five out of nine predicted covalent kinase inhibitors showed half-maximal inhibitory concentration (IC50) values between 155 nM and 4.5 μM. Application against an existing SARS-CoV Mpro reversible inhibitor led to an acrylamide inhibitor series with low micromolar IC50 values against SARS-CoV-2 Mpro. The docking was validated by 12 co-crystal structures. Together these examples hint at the vast number of covalent inhibitors accessible through our protocol.
|
Jun 2021
|
|
I04-1-Macromolecular Crystallography (fixed wavelength)
I24-Microfocus Macromolecular Crystallography
|
Martin A.
Redhead
,
C. David
Owen
,
Lennart
Brewitz
,
Amelia H.
Collette
,
Petra
Lukacik
,
Claire
Strain-Damerell
,
Sean W.
Robinson
,
Patrick M.
Collins
,
Philipp
Schäfer
,
Mark
Swindells
,
Chris J.
Radoux
,
Iva Navratilova
Hopkins
,
Daren
Fearon
,
Alice
Douangamath
,
Frank
Von Delft
,
Tika R.
Malla
,
Laura
Vangeel
,
Thomas
Vercruysse
,
Jan
Thibaut
,
Pieter
Leyssen
,
Tu-Trinh
Nguyen
,
Mitchell
Hull
,
Anthony
Tumber
,
David J.
Hallett
,
Christopher J.
Schofield
,
David I.
Stuart
,
Andrew L.
Hopkins
,
Martin A.
Walsh
Open Access
Abstract: Effective agents to treat coronavirus infection are urgently required, not only to treat COVID-19, but to prepare for future outbreaks. Repurposed anti-virals such as remdesivir and human anti-inflammatories such as barcitinib have received emergency approval but their overall benefits remain unclear. Vaccines are the most promising prospect for COVID-19, but will need to be redeveloped for any future coronavirus outbreak. Protecting against future outbreaks requires the identification of targets that are conserved between coronavirus strains and amenable to drug discovery. Two such targets are the main protease (Mpro) and the papain-like protease (PLpro) which are essential for the coronavirus replication cycle. We describe the discovery of two non-antiviral therapeutic agents, the caspase-1 inhibitor SDZ 224015 and Tarloxotinib that target Mpro and PLpro, respectively. These were identified through extensive experimental screens of the drug repurposing ReFRAME library of 12,000 therapeutic agents. The caspase-1 inhibitor SDZ 224015, was found to be a potent irreversible inhibitor of Mpro (IC50 30 nM) while Tarloxotinib, a clinical stage epidermal growth factor receptor inhibitor, is a sub micromolar inhibitor of PLpro (IC50 300 nM, Ki 200 nM) and is the first reported PLpro inhibitor with drug-like properties. SDZ 224015 and Tarloxotinib have both undergone safety evaluation in humans and hence are candidates for COVID-19 clinical evaluation.
|
Jun 2021
|
|