25 August 2025

Fragments vs KEAP1: Fragment growing this time

Kelch-like ECH-associated protein 1 (KEAP1) binds to nuclear factor erythroid 2-related factor 2 (NRF2), targeting it for degradation. Blocking this interaction has anti-inflammatory effects, and indeed the approved drugs dimethyl fumarate and omaveloxolone are believed to act in part through this mechanism. But those drugs hit a lot of other targets, and more specific molecules have long been sought; we wrote about one in 2016 and another in 2021. In an open-access paper just published in Angew. Chem. Int. Ed., Anders Bach and an international team of collaborators at University of Copenhagen and elsewhere describe a new chemical series.
 
As in the 2016 paper, the researchers started with a crystallographic screen, in this case using the 768-member DSI-poised library, which we wrote about here. This resulted in 80 hits, all binding in the so-called Kelch pocket, which has previously been targeted. Thirteen of these bound in the central region, and compound 1 showed modest but measurable affinity by SPR.
 

All previously reported non-covalent high-affinity KEAP1 ligands contain at least one acidic moiety to interact with arginine residues in the protein, so the researchers used structure-based design to add carboxylic acids, resulting in compound 4, with low micromolar affinity. This molecule, unlike the initial fragment, could also block the KEAP1-NRF2 interaction in a fluorescence polarization assay.
 
Building into a hydrophobic sub-pocket yielded compound 12, and adding strategically placed hydrogen-bond acceptors led to further improvements in affinity, ultimately leading to compound 28, with low nanomolar activity. Crystallography revealed that these molecules bound in a similar fashion as the initial fragment.
 
Compound 28 and related molecules were tested in a variety of assays. They were selective for KEAP1 over 15 other human Kelch domains in a thermal shift assay. Compound 28 activated NRF-2 regulated cytoprotective genes and decreased inflammatory markers in multiple cell lines. It also displayed RNA expression profiles similar to those of other reported non-covalent KEAP1 inhibitors. Cellular potency in some of these assays was as good as 60 nM.
 
This is a nice fragment-to-lead story, though no ADME or DMPK data are reported, and the combination of relatively high molecular weight, negative charge, and lipophilicity suggest that permeability and oral bioavailability may be challenging. Indeed, the researchers note that no non-covalent KEAP1-NRF2 inhibitors have entered the clinic. Perhaps this target is better suited for covalent inhibitors, preferably ones more selective than dimethyl fumarate. More on those later.

18 August 2025

Hundreds of crystallographic ligands for FABP4 – many not as expected

The ten human fatty-acid binding proteins (FABPs) shuttle lipids around cells. As we noted several years ago, FABP4 and FABP5 are potential drug targets for diabetes and atherosclerosis, but selectivity over FABP3 is needed to avoid cardiotoxicity. Markus Rudolph and colleagues at Hoffmann-La Roche describe progress towards selective molecules in three consecutive open-access Acta. Cryst. D papers. Perhaps more importantly, they gift a massive high quality data set to the scientific community – along with some important caveats about data for protein-ligand structures.
 
The first paper focuses on purification and NMR characterization of FABP4. Recombinant FABPs are normally expressed in E. coli, and they always contain natural fatty acids that copurify with the protein. This can complicate ligand binding studies, since the endogenous fatty acids act as competitors. Indeed, the researchers highlight two structures in the protein data bank (PDB) whose supposed ligands are probably fatty acids.
 
To solve this problem, the researchers denature FABP4, separate the fatty acid, and then refold the protein. This truly apo form of the protein was studied by NMR, revealing that the protein becomes more rigid upon ligand-binding.
 
The second paper is of more general interest. It reports a set of 229 crystal structures of various FABPs, of which 216 have a bound ligand. Of these, 75 have associated IC50 values for at least one FABP, and 50 compounds have IC50 values reported for FABP3, FABP4, and FABP5. Importantly, the structures are solved to high resolution, with a median of 1.12 Å. Two crystal forms are particularly suitable for soaking, and compounds were typically soaked at 60 mM in 30% DMSO overnight.
 
All the crystal structures are deposited in the PDB, and all the binding data are provided in the supporting information. Given FABPs’ predilection for carboxylic acids, the ligands contain a variety of carboxylic acid mimetics. This wealth of high-quality data should be valuable for constructing machine-learning binding models, and the researchers conclude by calling “on other industrial organizations to also make their legacy data available such that prediction models with broader applicability may be developed more quickly.”
 
But it was the third paper that really caught my attention: the researchers summarized it as “what is written on the bottle is not what is in the crystal.” In fact, of the 216 ligands reported, a whopping 33 (15%) do not match the compound registered. These are grouped into several categories and described in detail.
 
Human error is the simplest to explain: the researchers show an example where a 1,2-benzoxazole was registered as a 1,3-benzoxazole. Because the molecules have the same molecular weight, mass spectrometry could not distinguish them. Similarly, the researchers find several cases where the wrong enantiomer or diastereomer was registered. In another case, a racemic mixture led to a single enantiomer bound to FABP4, with the protein acting as a “chiral sponge.”
 
Other cases are more unusual, and include ring closing, ring opening, acyl shifts, hydrolysis, and instances of ligand decomposition or incomplete reactions. The researchers note that small amounts of impurities could be particularly problematic at the high ligand concentrations used for soaking; they calculate that just 0.06% impurity would be equivalent to the total amount of FABP in a crystal. Some fragment screens are done at even higher concentrations, further increasing the risk of enriching impurities.
 
A 15% rate of unexpected ligands is comparable to the numbers we blogged about here, but those were commercial libraries, whereas this set is from Roche, which likely has better internal quality control. One factor that led to the recognition of the problem is the high resolution, where a single atom change could be readily seen. Another is the buried nature of the ligands; ligands bound on the surface of a protein may have more dynamically disordered bits, which would be difficult to distinguish from missing moieties caused by decomposition.
 
Indeed, the researchers examine two other proteins, PDE10 and ATX, for which they have also released ~200 ligand-bound structures but at lower average resolutions. There are some unexpected ligands for these proteins too, but many fewer than for the FABPs – or perhaps we just can’t observe some of them.
 
As we noted back in 2014, up to a quarter of ligand-containing crystal structures in the PDB may contain serious errors, and the researchers cite a study suggesting that 12% are “just bad.” These could have obvious negative consequences for training computational models, and the researchers call on the community to set standards to create a rigorously chosen training set. Perhaps this discussion could be held in parallel with the discussion on how to house fragment screening data, which we wrote about last month.

11 August 2025

Fragments vs CYP125 and CYP142 for M. tuberculosis

Although 2020 and 2021 were baleful exceptions, tuberculosis is normally the world’s deadliest infectious disease. The pathogen Mycobacterium tuberculosis (Mtb) makes its home inside macrophages, the very cells that normally destroy microorganisms. Worse, some strains have become resistant to approved drugs. In a recent open-access J. Med. Chem. paper, Madeline Kavanagh, Kirsty McLean, and collaborators at University of Manchester, University of Cambridge, and elsewhere explore a new mechanism to fight this ancient disease.
 
An important nutrient source Mtb exploits inside human cells is cholesterol, which bacteria oxidize with the cytochrome P450 enzyme CYP125. A second enzyme, CYP142, is also present in some strains and is functionally redundant. Thus, the researchers set out to make a dual inhibitor.
 
Mtb has some 20 CYPs, and the Cambridge researchers have been studying them for a long time: we wrote about their work on CYP121 in 2016 and their work on CYP126 in 2014. All these enzymes contain a heme cofactor, and much is known about targeting the bound iron. However, some ligands are promiscuous, hitting human P450 enzymes, or they are rapidly effluxed out of cells. Thus, the researchers built a fragment library of just 80 likely heme binders but excluded particularly promiscuous moieties, such as imidazoles. The library was screened using UV-vis spectroscopy; ligands that bind to the heme group cause a red-shift in the λmax. Only four hits were found for CYP125, while a dozen were found for CYP142, including three of the four CYP125 hits. Compound 1a had modest affinity for CYP125 and low micromolar affinity for CYP142.
 
Compound 1a was soaked into crystals of CYP142, and interestingly two molecules bound at the active site: one coordinating to the iron atom as expected, the other binding near the entrance of the active site. This suggested a linking or merging strategy, so the researchers made small libraries based on compound 1a and tested these against the two enzymes. Compound 5m was the most potent against both. Crystal structures of this molecule bound to both CYP125 and CYP142 confirmed that the pyridine nitrogen maintained its interaction with the heme iron, while the added bit nicely filled the space previously occupied by the second copy of compound 1a.
 
Functional assays revealed that compound 5m inhibited both enzymes with nanomolar activity, comparable to their affinities. It also inhibited the growth of Mtb grown on media containing cholesterol as the sole source of carbon. More impressively, it even inhibited the growth of Mtb in standard media spiked with just low concentrations of cholesterol. Oddly though, it also inhibited the growth of Mtb grown on media not containing cholesterol, albeit at a higher concentration, suggesting perhaps other targets. But one reason tuberculosis is so hard to treat is that the bacteria persist inside human cells. Encouragingly, compound 5m inhibited the growth of Mtb in human macrophages at low micromolar concentrations, and it  did not show cytotoxicity up to 50 micromolar concentration.
 
Unfortunately, compound 5m did show cytotoxicity to human HepG2 cells, and it also inhibited several human P450 enzymes at high nanomolar concentrations, which could cause drug-drug interactions. Also, selectivity against other MTb P450 enzymes is unclear. Finally, no in vitro ADME data are reported. Nonetheless, this is a nice fragment to lead story, and compound 5m could be used – cautiously – as a chemical probe to study Mtb biology.

04 August 2025

The Chemical Probes Portal turns ten. Use it!

Last week we highlighted a new tool to computationally predict whether a molecule might aggregate, thereby causing false positives. This doesn’t necessarily mean the molecules are bad (after all, some approved drugs aggregate), but it’s all too easy to screen molecules under inappropriate conditions. This brings up the topic of chemical probes, and as it happens the Chemical Probes Portal turns ten years old this year, as celebrated in a Cancer Cell Commentary by Susanne Müller, Domenico Sanfelice, and Paul Workman and a blog post by Ben Kolbington at the Institute of Cancer Research.
 
We first wrote about the Chemical Probes Portal in July 2015, when it contained just 7 compounds. When we returned in 2023 it contained more than 500 compounds, and by the end of last year the number was up to 803. As of today it lists 1174 probes for 622 targets. Nearly a third of the probes also have chemically related inactive controls. These seem like large numbers, but the the human genome conservatively encodes for some 20,000 proteins, and the ambitious Target 2035 initiative seeks chemical probes for all of them.
 
The new paper emphasizes that the standards are in some ways higher for chemical probes than for approved drugs: “whereas probes principally require a high degree of selectivity, drugs need ‘only’ to be safe and effective and may often hit several targets.” Dimethyl fumarate comes to mind as a highly promiscuous covalent modifier that is nonetheless a useful drug for multiple sclerosis and psoriasis.
 
Even when a compound hits a target of interest, that doesn’t mean any biological effects observed are due to the target, particularly when the readout is cell death. The researchers note that TH588 was originally reported as a potent inhibitor of MTH1, but it actually kills cancer cells by binding to tubulin, a fact not always mentioned by chemical suppliers. Another study found that ten clinical compounds were still active in cells even when their putative target was knocked out using CRISPR.
 
The tone of the Commentary is pragmatic, emphasizing that for new or difficult targets, it may be difficult to find good chemical probes. For example, LY294002 is mentioned as a “pathfinder tool” that was useful to explore the biology around the PI3 kinase family but has now been superseded by more selective molecules.
 
Unfortunately, not everyone seems to have gotten the message. Curcumin, which as we noted can aggregate, form nonselective covalent adducts, fluoresce, and generate reactive oxygen species, appears in >2600 PubMed publicationsjust in the past year. What a waste.
 
If you’re exploring the biology of a target, please check the Portal to see whether there are good probes. If you’re reading (or reviewing!) a paper that reports small molecule studies, please check to see whether the probe has been assessed - especially to see if it shows up as one of more than 250 Unsuitables. And if you’re interested in participating, please consider reviewing or even hosting a Probe Hackathon.