Showing posts with label fragment linking. Show all posts
Showing posts with label fragment linking. Show all posts

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.

03 February 2025

Stitching together fragments with Fragmenstein

As we noted just last week, crystallography has unleashed a torrent of protein-ligand complexes, especially fragments. Historically a single structure might be used for fragment growing, but so many structures present an embarrassment of riches, with sometimes dozens of fragments that bind in the same region. Merging or linking these fragments can be done manually, as seen here and here, but how to do so when the binding modes are partially overlapping is not always intuitive. In a new open-access J. Cheminform. paper, Matteo Ferla and colleagues at University of Oxford and elsewhere describe an open-source solution called Fragmenstein.
 
We briefly described Fragmenstein in 2023, where it was used to combine pairs of low-affinity fragments bound to the Nsp3 macrodomain of SARS-CoV-2 to generate sub-micromolar inhibitors. The current paper describes the platform in detail.
 
Fragmenstein starts by taking two (or more) structures of fragments bound to a protein and virtually combining them. This is done by collapsing rings to their individual centroids, stitching these together along with their substituents, and then re-expanding the ring(s) so the substituents will be close to where they were in the initial fragments. This process produces a surprising array of molecules beyond the obvious. For example, if one fragment contains a phenyl ring and the other fragment contains a furan ring, the stitched molecule might just contain the phenyl (if the two rings bind in nearly the same position), a benzofuran (merging the rings), a phenyl ring linked to a furan by one or more atoms, or even a spiro compound if the rings are perpendicular to one another.
 
In silico approaches sometimes suggest molecules that are synthetically challenging to make, but Fragmenstein can also be used to find purchasable analogs.
 
Next, the new molecules are energetically minimized, first by themselves and then while docked into the protein. In contrast to other docking programs, which might allow molecules to sample thousands of different conformations and sites in a protein, Fragmenstein maintains the new molecule in a similar position and orientation to the initial fragments, with the assumption that these have already identified energetically favorable interactions.
 
The researchers successfully applied Fragmenstein retrospectively to several targets. The COVID Moonshot (which we discussed here) crowd-sourced molecule ideas for the SARS-CoV-2 main protease based on structures of bound fragments. Of 87 ligands that had been crystallographically characterized and were designed based on two fragments, Fragmenstein successfully (RMSD < 2 Å) predicted the binding mode for 69%.
 
Fragmenstein can even be used for covalent ligands, as shown for the target NUDT7, which we wrote about here. Merging two fragments led to compound NUDT7-COV-1, and the RMSD between the Fragmenstein model and the crystal structure was an impressive 0.28 Å.
 
Of course, as the researchers acknowledge, the number of possible analogs might be daunting, and deciding which to make or buy is not necessarily straightforward. Also, Fragmenstein assumes that the fragments themselves are making productive interactions with the protein, which may not be the case, as we suggested here. Still, the tool is open-source and worth trying, especially if you are swimming in crystal structures.

16 September 2024

Casting light on target-guided synthesis

Target-guided synthesis, in which a protein templates the formation of its own inhibitor, is a concept first proposed decades ago. There are roughly two flavors. Dynamic combinatorial chemistry (DCC) involves reversible formation of the product, and we wrote in 2017 about some of the challenges. Kinetic target-guided synthesis (KTGS) involves irreversible chemistry, for which the options are limited. The classical click chemistry azide-alkyne cycloaddition is so slow that reactions usually take days, which can be a problem for delicate proteins. A recent (open-access) paper in Angew. Chem. Int. Ed. by Cyrille Sabot et al. describes a bright way to accelerate things.
 
The researchers turned to photochemistry, specifically diazirine chemistry. Illuminating 3-trifluoromethyl-3-phenyldiazirines leads to loss of nitrogen and formation of highly reactive carbenes. The carbenes are so hot that they can react indiscriminately with proteins, as we described here. However, the reaction with thiols is faster than the reaction with other functional groups on proteins, so the researchers reasoned that a library of thiols could out-compete the protein.
 
The carbonic anhydrase bCA-II was chosen as a model protein. Sulfonamide-containing molecules such as compound 5 are known to be good inhibitors. This “anchor” molecule was incubated at 60 µM with seven different diazirines, each at 400 µM, in the presence or absence of 30 µM bCA-II and then irradiated with 365 nM light for a few minutes. Most of the reactions produced similar amounts of product in the presence or absence of bCA-II, but compound 1b yielded about threefold more of compound 2d in the presence of bCA-II, suggesting the reaction was being templated by the protein. 
 

Control experiments lend credence to this hypothesis. First, adding a known competitive bCA-II inhibitor reduced the formation of compound 2d to background levels. Second, other proteins did not cause a similar enhancement in the formation of compound 2d. Finally, conducting the experiment with phenylmethanethiol (ie, a variant of compound 5 lacking the sulfonamide moiety essential for interaction with bCA-II) did not cause an enrichment of the photochemical product in the presence of the enzyme.
 
Chiral HPLC was used to show that compound 2d was slightly enriched for the (R)-enantiomer, with an enantiomeric excess of around 10%, when the reaction was conducted in the presence of bCA-II but not in the absence. The two enantiomers were synthesized and tested, and the (R) form did indeed have slightly better activity (300 nM vs 330 nM).
 
This is a thoughtful, well-conducted investigation. But it makes me even less sanguine about the practicality of KTGS for finding new chemical matter, for several reasons. First, the efficiency of the reaction is poor: the researchers calculate the yield of compound 2d at around 1% of the enzyme concentration, so low that they used single-ion monitoring (SIM) mass spectrometry to detect it. Because of this low efficiency, the concentration of enzyme used needs to be quite high.
 
The most serious strike against KTGS is the fact that all of the diazirines generated potent (sub-micromolar) inhibitors. One of them was even slightly better than compound 2d but did not show enrichment in the presence of bCA-II. False negatives seem to be a major problem, as we’ve written previously.
 
One caveat to my caveats is that compound 2d is only marginally more potent than the starting compound 5. NMR experiments conducted with diazirine 1b suggest binding to the protein, though the affinity was not quantified. Perhaps a different fragment linking system, in which both fragments have measurable affinity for the target, would be better suited to demonstrate the utility of KTGS. For now, this paper does a nice job highlighting its drawbacks.

18 December 2023

Review of 2023 reviews

The annual Practical Fragments look-back on the preceding year may not be the most highly anticipated year-end tradition, but I hope you find something of interest in this twelfth edition.
 
I was fortunate to attend several conferences and wrote about CHI’s Discovery on Target in Boston and Drug Discovery Chemistry in San Diego. As for reviews, Louise Walsh and collaborators at Astex, Vrije Universiteit Amsterdam, Novartis, and Frontier Medicines (me!) published our annual analysis of fragment-to-lead success stories in J. Med. Chem., this one covering the year 2021. Some twenty other reviews of interest to this readership were also published. I’ll cover them thematically below.
 
Methods
Crystallography is the most popular fragment-finding technique, and in Expert Opin. Drug Disc. Wladek Minor and collaborators at University of Virginia and Jagiellonian University examine “the current role and evolution of X-ray crystallography in drug discovery and development.” At the start of 2023 the Protein Data Bank (PDB) contained more than 200,000 structures, which sounds impressive until you learn that the AlphaFold database contains more than 200 million predicted protein structures. But this is not experimentalist vs machine: the researchers note how machine learning approaches can be used to more rapidly refine and improve experimental data with resources such as CheckMyBlob and PDB-REDO.
 
For those wishing to dig deeper, two papers in Methods Enzymol. go into experimental detail. In the first, Natalie Tatum and colleagues at Newcastle University describe “crystallographic fragment screening in academic drug discovery.” May Sharpe and collaborators at the Swiss Light Source and University of Hohenheim describe their fast fragment-screening pipeline in a comprehensive (49 page) guide. The focus is on reproducibility, and there is plenty of practical advice. For example, “the authors have even been successful in flying with crystal plates,” though getting these through airport security may be easier in some countries than others.
 
Protein-detected NMR was the first truly practical fragment-based approach, and another paper in Methods Enzymol. by Brian Volkman, Brian Smith, and colleagues at Medical College of Wisconsin describes “fragment-screening by protein-detected NMR.” This distills eight years of effort building their internal protein-detected NMR fragment screening platform that has been applied to 16 proteins thus far. The chapter is particularly detailed on protein and library preparation and screening.
 
Compared with crystallography and NMR, virtual screening can be dramatically faster; we’ve highlighted multibillion-compound screens. In WIREs Comput. Mol. Sci., Artem Cherkasov, Francesco Gentile, and colleagues at University of British Columbia and University of Ottawa discuss (open access) how computational methods are “keeping pace with the explosive growth of chemical libraries.” They cover brute force methods, fragment-based virtual screening, and machine-learning based methods, all while avoiding hype, and conclude that it will take time for these methods to “have a real impact on practical drug discovery.”
 
Finally, Marianne Fillet and collaborators at University of Liege and University of Namur provide a general review in Trends Anal. Chem. covering multiple methods to detect non-covalent fragments. These include established techniques such as biochemical assays, ligand-observed NMR, crystallography, thermal shifts, and SPR, as well as less common ones such as WAC, microscale thermophoresis, ACE, and DEL. The paper includes several nice tables and even a decision tree to help choose among the various approaches.
 
Covalent fragments
Many techniques to detect noncovalent interactions also apply to reversible covalent inhibitors, the subject of a review in Med. Chem. Res. by Faridoon and collaborators at Genhouse Bio and Olema Oncology. The researchers focus on various warheads including cyanoacrylamides, nitriles, ketones and aldehydes, boronic acids, and others, and provide multiple examples for each.
 
In contrast, an open-access review in Pharmaceuticals by Monique Multeder and collaborators at Leiden University Medical Center discusses methods to detect both reversible as well as irreversible covalent protein-drug adducts. Crystallography is the most informative, but the researchers also delve into various mass-spectrometry techniques including top-down (with intact proteins) and bottom-up (after digestion of modified proteins). Also covered are activity-based protein profiling (ABPP) methods, NMR, and fluorescence-based approaches. The nearly 300 references make a useful compendium.
 
One of the most exciting recent developments is “proteome-wide fragment-based ligand and target discovery,” the subject of an open-access review in Isr. J. Chem. by Ines Forrest and Christopher Parker, both at Scripps. This concise, highly readable account covers a lot of ground, from ABPP to fully functionalized fragments (FFFs) to phenotypic screening.
 
If you’re doing covalent FBLD you’ll need a library of covalent fragments, and if you’re building one, I’d recommend a review in Prog. Med. Chem. by David Mann and colleagues at Imperial College London. The paper nicely summarizes design principles such as choice of warhead and the fact that reactivity can vary considerably even among compounds with the same warhead. Synthetic methods and screening approaches are also well covered, along with methods to distinguish specific binding from nonspecific reactivity.
 
Most covalent fragments target cysteine residues, but there at least nine other potentially reactive amino acids, and these are the subject of an open-access review by György Keserű and colleagues at Budapest University of Technology and Economics in Trends Pharm. Sci. Lysine, serine, threonine, tyrosine, and histidine are the most common targets, though some of the warheads are so reactive that specificity will be challenging, let alone reasonable pharmacokinetic properties. This is especially true for aspartic and glutamic acids, methionine, and tryptophan.
 
Finally, another article in Trends Pharm. Sci. by Carlo Ballatore and colleagues at University of California San Diego describes using covalent strategies to develop stabilizers and inhibitors of protein-protein interactions (PPIs). Site-directed fragment tethering with disulfide and imine chemistry is a focus, particularly in the context of 14-3-3 proteins. Proximity-enabled covalent strategies, in which warheads are grafted onto non-covalent molecules, are also covered. There is also a short section on covalent PROTACs – more on that topic below.
 
Targets
Keeping with the theme of protein-protein interactions, Ge-Fei Hao, Guang-Fu Yang, and collaborators at Central China Normal University and Guizhou University discuss fragment-based approaches against “undruggable” PPIs in Trends Biochem. Sci. After describing why protein-protein interactions can be difficult, the paper presents several successful case studies, including venetoclax, sotorasib, and targeting 14-3-3 proteins.
 
Targeted protein degradation continues to be a major focus for drug discovery, and this is commonly achieved by hijacking E3 ligases to cause them to ubiquitinate a target of interest. Iacovos Michaelides and Gavin Collie (AstraZeneca) describe how FBLD has been used to find ligands against E3s in an open-access J. Med. Chem. paper. There are more than 600 E3s, and because their biology relies on protein-protein interactions they are often tough targets. Fragment hits can be weak and difficult to advance, though the researchers do describe several success stories including against KEAP1 and XIAP/cIAP. Covalent fragments have the potential to permanently reprogram E3 ligases, and these are covered well too.
 
Another difficult type of target is RNA, the topic of two reviews. In an open-access Curr. Opin. Struct. Biol. paper Kevin Weeks and colleagues at University of North Carolina Chapel Hill provide a concise and beautifully illustrated overview of the field. They note that “RNA-targeted FBLD is in its infancy,” but given that the first report dates to 2002 it is a long childhood, and the paper does a good job of describing the challenges.
 
A more extensive treatment of “fragment-based approaches to identify RNA binders” is provided by Matthew Disney and colleagues at UF Scripps in J. Med. Chem. The paper describes many case studies, some of which we’ve covered, and also contains a handy table comparing the pros and cons of a dozen different methods for finding RNA-binding fragments.
 
Tuberculosis kills more than 1.5 million people each year, and fragment-based approaches have been applied against multiple targets within the pathogen, as reviewed by Baptiste Villemagne and colleagues at University Lille in Eur. J. Med. Chem. We’ve covered many of these studies on Practical Fragments, but as the paper notes none have advanced to the clinic. This is attributed in part to cell permeability, and the researchers suggest turning to phenotypic screens (see below).
 
Other
Fragment linking can be difficult but highly effective, especially for difficult targets. An overview of published linkers is provided by Isabelle Krimm and collaborators at Université Claude Bernard Lyon and Université Montpellier in Expert Opin. Drug Disc. The paper includes a table summarizing 40 fragment linking stories, noting that most linkers are short and flexible. Another table summarizes 19 examples of target-guided synthesis, including dynamic combinatorial chemistry. As the paper notes, all of these are small model studies based on known compounds. In silico approaches, the last topic covered, will probably prove more practical.
 
And on the subject of practical, Dean Brown (Jnana Therapeutics) provides an “analysis of successful hit-to-clinical candidate pairs” in J. Med. Chem. This is an update to his 2018 article and captures 156 clinical candidates reported in the journal between 2018 and 2021. Of these, 14 had fragments in their lineage. Most of these drugs appear in our list of fragment-derived clinical candidates (though berotralstat does not – I’ll need to look closer). The paper contains lots of interesting analyses. For example, of the 138 oral drugs, 39 had a molecular weight > 500 Da, 24 had Clog > 5, and 17 had more than 10 hydrogen bond acceptors (HBA). On the other hand, none had more than 5 HBD, emphasizing that you should be parsimonious with hydrogen bond donors.
 
Finally, veteran drug hunter Nicholas Meanwell provides “reflections on a 40-year career in drug design and discovery” (open access) in Med. Chem. Rev. Those of you who saw his talk earlier this year at the CHI DDC meeting will know what to expect, and those of you who didn’t will be in for a treat. A personal and entertaining romp through pharma starting in the early 1980s, the paper is full of surprises, such as the pursuit of minor impurities in a phenotypic screen that ultimately led to the hepatitis C drug daclatasvir. Nicholas notes that “you discover what you screen for, so screen design is of paramount importance.”
 
The paper also reveals a passion for medicinal chemistry: “In a search for inspiration for design concepts, I sat down one Saturday afternoon in early October of 1987 and perused every molecule in the United States Adopted Names (USAN) dictionary.” And, as he notes near the end, “Decision making in drug discovery and development is a delicate balancing act, inherently flawed based on absence of predictive accuracy, and knowing when to conclude a discovery program with grace is also an important trait.” That said, he provides examples of successful programs that were almost killed multiple times – and others that were killed at Bristol Myers Squibb but subsequently succeeded elsewhere. While this is frustrating on one level, Nicholas takes satisfaction in the fact that “the science that we conducted and the molecules and pharmacophores that we defined have been of benefit to mankind.”
 
There are still a couple weeks left in the year, but that’s it for Practical Fragments for 2023. Thanks for reading, and special thanks for commenting. And if you live in one of the 70+ countries with elections in 2024, please vote.

11 September 2023

Fragments vs malarial DHFR

Malaria continues to be a worldwide scourge, with some quarter billion cases last year. A seventy-year-old drug called pyrimethamine targets the dihydrofolate reductase (DHFR) enzyme from Plasmodium falciparum, but resistance mutations have rendered this molecule mostly useless. An analog called P218 was developed to overcome this resistance and completed a handful of phase 1 clinical trials, but unfortunately the human pharmacokinetics were found lacking. In a new RSC Med. Chem. paper, Marie Hoarau and colleagues at the National Center for Genetic Engineering and Biotechnology in Thailand describe their efforts to improve this molecule.
 
The researchers recognized that the phenyl propanoate moiety of P218 was a metabolic liability and sought a replacement. They screened a library of 1163 fragments (from Key Organics) at 1 mM using a thermal shift assay. This resulted in 64 hits, 52 of which confirmed by SPR. Of these, 22 showed some level of inhibition at 0.5 mM against mutant PfDHFR.
 
Among the hits, five were “bi-aromatic carboxylates,” such as compound 136. These were prioritized because, while reminiscent of the phenyl propanoate in P218, they had fewer rotatable bonds. Some of them also showed slow off-rates by SPR, though in my opinion the sensorgrams look suspicious, perhaps due to excessive protein loading on the chip. (For example, the Kd for compound 136 calculated from the on and off rates comes in at 160 nM, unrealistically potent given that it shows only 20% enzymatic inhibition at 0.5 mM. Note – all values here and in the figure are for the mutant form of the enzyme.)
 

SAR by catalog was used to find additional analogs, such as compound AF10, which showed measurable inhibition of the enzyme. Next, the researchers tested hits in the presence of a pyrimidine fragment (L4) derived from P218, known to bind nearby. Compound AF10 showed greater inhibition than would be expected by simple additivity, perhaps suggesting some preorganization of the binding site, as in a different example discussed here.
 
Molecular modeling was used to link the carboxylate fragments with L4, and eight were made and tested. All inhibited both wild type and mutant PfDHFR, and compound 8 showed good selectivity over human DHFR too. A crystal structure confirmed that it bound as predicted. From a fragment-linking perspective, the sub-nanomolar affinity of compound 8 is impressively better than would be expected given the weak affinities of L4 and AF10.
 
Unfortunately, despite similar in vitro potency against the isolated enzymes, compound 8 and the other molecules tested showed “disappointing” activity against Plasmodium falciparum carrying either wild-type or mutant DHFR, roughly 100- to 1000-fold less potent than P218. The researchers suggest solubility may be a factor.
 
This paper is a useful reminder of the dramatic disconnects often seen between enzymatic and cell activity. Nonetheless, it is another good example of using fragment-based methods to replace one portion of an existing molecule.

14 August 2023

Stabilizing protein-protein interactions: part 3 (fragment linking)

Stabilizing protein-protein interactions is becoming increasingly popular, and not just for PROTACs. Nearly three years ago we highlighted the use of crystallographic screening to find fragments that could stabilize interactions between the adapter protein 14-3-3δ and peptides derived from p53, a prominent cancer target. After noting how much work lay ahead, we ended the post with, “expect a part 3!” This has now been published (open access) in Angew. Chem. by Adam Renslo, Luc Brunsveld, Michelle Arkin, Christian Ottmann, and collaborators at UCSF and Eindhoven University of Technology.
 
In addition to crystallographic fragment screening, the researchers had previously performed a disulfide Tethering screen on the 14-3-3δ protein, which we described here. The fragments from the two screens bound next to one another, so the researchers decided to link them. They started by solving the crystal structure of compound 1 disulfide-bonded to 14-3-3δ in the presence of fragments from the crystallographic screen as well as a peptide derived from estrogen receptor alpha (ERα, another anti-cancer target). These co-structures guided the synthesis of new linked molecules, and these were soaked into crystals of 14-3-3δ and the ERα peptide. Compound 6 gave strong electron density and overlayed nicely on the initial fragments.
 
 
To determine whether the linked molecule could stabilize the 14-3-3δ/ERα complex, the researchers developed a fluorescence anisotropy assay with a dye-labeled peptide from ERα. Some of the linked molecules produced an increase in anisotropy, suggesting stabilization of the 14-3-3δ/ERα complex, but when the researchers ran the important control of repeating the experiment in the absence of 14-3-3δ they found that several molecules still increased anisotropy, which could be due to aggregation. (Adam published a nice early paper on aggregation and is thus particularly attuned to the dangers.)
 
Fortunately, some of the molecules passed this control, and with a robust crystallography system the researchers were able to use structure-based design to improve them, ultimately arriving at compound 24, which increased the affinity of the 14-3-3δ/ERα complex by 25-fold. It was also quite specific towards ERα, and did not increase the affinity of nine peptides from from other proteins for 14-3-3δ. The researchers attribute this selectivity to the fact that most other peptides would sterically clash with compound 24. (Not reported was the peptide from p53, which would be interesting.)
 
This is a nice paper on several levels. In addition to selectively stabilizing a therapeutically relevant protein-protein interaction, this is a rare example of starting with a covalent fragment and developing a non-covalent binder. (For another, see here.) Also, this is a good example of fragment linking, which is often challenging.
 
There is still a long way to go. The most potent molecules all contain amidine moieties, whose high polarity is a liability for cell permeability, let alone oral bioavailability. Moreover, the affinity of compound 24 is still quite weak, with a low ligand efficiency.
 
That said, with a wealth of structural and biological understanding I am optimistic further progress can be made, perhaps by rebuilding the covalent linkage to the protein, as was the case of sotorasib or this more recent paper from the UCSF team. I look forward to part 4!

13 March 2023

A very useful list: common linkers and bioisosteric replacements

Last week’s post highlighted an example of fragment linking, which despite being less common than fragment growing can still be effective. But how do you choose the linker? We’ve previously written about the most common rings found in drugs. In a new Bioorg. Med. Chem. paper Peter Ertl and colleagues at Novartis tabulate the most common linkers found in bioactive molecules.
 
The researchers start by defining linkers “as moieties connecting 2 ring systems.” To focus on druglike molecules, linkers could contain no more than eight non-hydrogen atoms total and no more than five consecutive bonds between the two ring systems. This means that para-disubstituted phenyl or 1,4-disubstiuted butyl would both be considered in the analysis, but longer linkers such as this recent example would not.
 
Molecules were extracted from the databases ChEMBL and ZINC, yielding a total of 1686 unique linkers. Various descriptors were calculated for all, which in addition to size and length included the number of heteroatoms and electronic properties. Bioactivity data for molecules in ChEMBL was used to assess which replacements were most frequently tolerated. If one linker could be replaced by another without causing a drop in affinity (or inhibition, etc.), the two linkers were considered to be bioisosteres.
 
So, what are the most common linkers? A single methylene is the most common, followed by an amide bond. I was surprised that, of the 40 most common linkers, only five are rings: para-disubstituted phenyl, 1,4-piperzine, 1,4-piperidine, 1,2,4-oxadiazole, and meta-phenyl, in that order. Not coincidentally, phenyl rings, piperidines, and piperazines are also the most common rings found in drugs, according to an analysis last year.
 
Last year we highlighted a paper from the Ertl group that included a link to a “Ring Replacement Recommender,” which suggests bioisosteric replacements for any ring. Alas, there is no “Linker Replacement Recommender,” but the new paper does provide a “bioisosteric replacement network,” which is a full-page 10 x 15 grid with the 150 most common linkers arranged such that nearby linkers are likely to be bioisosteric. For example, para-phenyl is adjacent to 2,5-thiophene and quite some distance from sulfone. These make sense, but there are also less obvious examples: the table suggests that a 1,4-pyrazole makes a good replacement for a carbamate.
 
The next time you’re doing SAR, it may be worth consulting the bioisosteric replacement network for ideas.

06 March 2023

Fragment linking on the bacterial TPP riboswitch

Last week’s post focused on fragment screening against RNA, and we continue the theme this week with a paper published in Proc. Nat. Acad. USA by Kevin Weeks and collaborators at University of North Carolina Chapel Hill, New York University, and Université de Sherbrooke.
 
The researchers developed a screening technology called SHAPE-MaP (Selective 2’-Hydroxyl Acylation analyzed by Primer Extension and Mutational Profiling). Essentially, RNA in the presence or absence of potential ligands is treated with an acylating agent that reacts with the 2’-hydroxyl group on ribose subunits. This addition requires the hydroxyl groups to be exposed, so ligands that bind in the vicinity may directly block or cause conformational changes to change the patterns of acylation. Conveniently, acylation causes mutations when the modified RNA is sequenced, making modified sites easy to detect. Moreover, by clever uses of “barcodes” in other regions of the RNA, multiple samples can be pooled and analyzed.
 
Because the approach uses sequencing to identify binding sites, long strands of RNA can be tested. In this case, the researchers built an RNA construct containing a ‘pseudoknot’ structure in the dengue virus genome as well as a thiamine pyrophosphate (TPP) riboswitch, which changes conformation when it binds to TPP. (We wrote about a different fragment screen against this riboswitch back in 2014). A set of 1500 rule-of-three compliant fragments from Maybridge was screened, resulting in 41 hits. These were then rescreened in triplicate, which winnowed the field to just eight fragments, of which seven bound TPP and one appeared to be nonspecific. All eight were assessed by isothermal titration calorimetry (ITC), which produced measurable affinities for six.
 
Compound 2 was the most potent hit, and when the researchers tested 16 analogs they found some, such as compound 17, with improved affinity. With an eye towards fragment linking, they took a conceptually similar approach to SAR by NMR by rescreening the original 1500 fragments in the presence of compound 2 to look for ligands that would bind at a second site. This yielded five hits, including compound 28. ITC characterization of a more soluble analog, compound 31, revealed that it had weak but measurably improved affinity in the presence of compound 2. A handful of linked analogs were made, and while most of these had affinity similar or worse than the best initial fragment, compound Z1 bound with sub-micromolar affinity as assessed by ITC.
 

The natural ligand TPP binds to the riboswitch with a Kd of 110 nM, and in doing so blocks in vitro transcription of bound RNA. Despite having a similar affinity as TPP, compound Z1 was much less effective at blocking transcription. Unfortunately, although the researchers were able to obtain crystal structures of several molecules bound to the riboswitch, including compound 17, they were unable to obtain one with compound Z1.
 
This is a rare example of fragment linking on RNA, and although the linked molecule does not show fully additive affinity, it does have reasonable ligand efficiency. But like the example last week, this paper illustrates how difficult discovering RNA binders is likely to be. The confirmed hit rate is less than 0.5%, and this is for an RNA sequence that evolved specifically to bind low molecular weight ligands. As the researchers note, none of the fragments bound the dengue virus pseudoknot. Perhaps most RNA is truly undruggable, at least with small molecules.

09 January 2023

From fragments to inhibitors of the SARS-CoV-2 Nsp3 macrodomain

Two years ago we highlighted what was likely the largest crystallographic fragment screen against any target, the macrodomain (Mac1) of the nonstructural protein 3 (Nsp3) of SARS-CoV-2. Mac1 dampens the cellular immune response to viral infection by removing ADP-ribose from various proteins. Separate mutational studies suggested this enzyme could be a good target for treating COVID-19.
 
The 234 fragment hits identified in 2021 could serve as good starting points. This has proven to be true, as demonstrated in a paper just published (open access) in Proc. Nat. Acad. Sci. USA by Brian Shoichet, James Fraser, and collaborators at University of California San Francisco, University of Oxford, Diamond Light Source, Enamine, and Chemspace. Despite the wealth of fragment hits, none of them were particularly potent; the best had an IC50 value of 180 µM in a homogenous time-resolved fluorescence (HTRF) competition assay. In the new paper, the researchers leveraged computational methods to advance these fragments.
 
First, they explored a fragment-linking approach termed Fragmentstein. This entailed choosing pairs of fragments that bound in close proximity to one another, merging or linking them, docking them to ensure the new molecule would bind in a similar manner to the component fragments, and then searching make-on-demand libraries in Enamine’s REAL database. Four pairs of fragments were evaluated, and 13 of 16 designed compounds were synthesized. Eight of these confirmed crystallographically, and two showed low micromolar activity in the HTRF assay. Interestingly, both of these came from the same fragment pair, ZINC922 and ZINC337835. The best molecule was a mixture of diastereomers, and one of the pure stereoisomers turned out to be submicromolar.
 
The potency of this fragment is all the more impressive given the low affinity of the initial fragments, which could only be crystallographically characterized using PanDDA, a method to find low-occupancy ligands that we wrote about here. Unfortunately, the compound has low cell permeability, likely due to the carboxylic acid moiety.
 
In addition to the linking approach via Fragmentstein, the researchers also conducted two virtual docking campaigns with more than 400 million molecules with molecular weights between 250-350 amu. Of 124 molecules purchased and tested, 47 confirmed crystallographically and 13 confirmed by HTRF, with IC50 values from 42 to 504 µM. (Ten of the 13 HTRF hits were also crystallographic hits. The researchers suggest the difference in confirmation rate is due at least in part to compound concentrations, which were 10-40 higher in the crystallographic screens.) In general the crystal structures confirmed the computationally predicted binding modes, particularly for fragments with measurable activity. Structure-based optimization of some of these fragments led to multiple low micromolar inhibitors, such as LRH-0021. Despite the carboxylic acid, this molecule is cell permeable.

This paper nicely illustrates how even very weak fragments can lead to multiple and very different series of inhibitors. The researchers acknowledge that the molecules are still at an early stage of development; indeed, they note that there are currently no good cellular assays to even assess the effect of Mac1 inhibition. Laudably, all the structures are deposited in the Protein Data Bank, which should provide a useful resource not just for further efforts on this protein but for understanding molecular interactions more generally.

22 December 2022

Review of 2022 reviews

The winter solstice is behind us in the Northern Hemisphere, which means 2022 is rapidly drawing to a close. As we have done for the past decade, Practical Fragments will spend this last post of the year summarizing conferences and reviews.
 
The remarkable progress in vaccines against SARS-CoV-2 allowed the full return of in-person conferences, and it was nice to see folks at CHI’s Discovery on Target in Boston and Drug Discovery Chemistry in San Diego. Nearly twenty reviews of interest to this readership were published, and these are covered thematically.
 
Targets
Several reviews cover the use of FBLD to target antiviral and antibacterial targets. Sangeeta Tiwari and colleagues at University of Texas El Paso cover both in an open access Pharmaceuticals review, focusing on tuberculosis and HIV, which often afflict the same individuals, leading to worse outcomes. The paper includes several tables with chemical structures, though the fragment origins of some molecules are not apparent.
 
Tuberculosis is caused by Mycobacterium tuberculosis, but there are more than 170 known members of the Mycobacteriaceae family. In an open access Int J. Mol. Sci. paper, the Tiwari group describes fragment-based approaches against these bugs. In addition to multiple examples, the review provides summaries of fragment finding methods and some of the challenges the field faces.
 
Another organism, Pseudomonas aeruginosa, infects the lungs of people with cystic fibrosis. In an open access Front. Mol. Biosci. paper, Tom Blundell and collaborators at University of Cambridge summarize fragment-based campaigns against this organism and its enzymes. The authors focus on structure-guided methods and note that the work is “at an early stage” but encouraging.
 
Switching to mammalian targets, Katrin Rittinger and colleagues at The Francis Crick Institute review (open access) applications of FBLD for targeting the ubiquitin system in Front. Mol. Biosci. The paper includes a nice table summarizing 15 examples that includes target, enzyme class, fragment binding mode, detection methods, and chemical structures of the fragment hit and optimized compound where applicable. Many of these are covalent modifiers; more on that topic below.
 
Finally, Tarun Jha, Shovanlal Gayen, and collaborators at Jadavpur University discuss “recent trends in fragment-based anticancer drug design strategies” in Biochem. Pharm. In addition to case studies (with chemical structures) of FBLD approaches against 18 oncology targets, the review covers fragment libraries, screening methods, optimization, and challenges.
 
Methods
Many of the targets above are challenging, and it’s always nice to be able to assess how challenging a project might be at the outset. In Curr. Opin. Struct. Biol., Sandor Vajda and collaborators at Boston University and Stony Brook University discuss (open access) “mapping the binding sites of challenging drug targets.” This is a brief, readable account of computational methods to identify hot spots, including allosteric ones. The authors examine the various small-molecule binding sites on KRAS and conclude that, due to “limited druggability,” the “other G12 oncogenic mutants will be very challenging.” Perhaps, but not impossible, as researchers at Mirati demonstrated earlier this year with the (open access) publication of a low (or sub) nanomolar KRASG12D inhibitor.
 
Among experimental methods used in FBDD, NMR is a mainstay, as demonstrated by Luca Mureddu and Geerten Vuister (University of Leicester) in Front. Mol. Biosci. (open access). The paper covers methods, successes, and challenges, focusing on three compounds that reached the clinic: AZD3839, venetoclax, and S64315.
 
In contrast to NMR, dynamic combinatorial chemistry (DCC) and DNA-encoded libraries (DEL) are used less frequently in FBLD. In RSC Chem. Biol., Xiaoyu Li and collaborators at University of Hong Kong and Jining Medical University discuss “recent advances in DNA-encoded dynamic libraries.” This concise paper covers lots of ground and does not understate the challenges.
 
Libraries
“The importance of high-quality molecule libraries” is emphasized by Justin Bower and colleagues at the Beatson Institute in Mol. Oncol. This highly readable and wide-ranging open access review covers all aspects of library design and use and includes comparisons of some of the major commercial vendors. An important point is that the “hit rate does not define the success of a library as it is more important to identify ligand-efficient and chemically tractable start points.”
 
Thus, even though shapely fragments may have lower hit rates than more planar aromatic fragments, they may still be worth including – if you can make them. In Drug Discov. Today (open access), Peter O’Brien and collaborators at University of York and Vrije Universiteit Amsterdam review synthetic strategies behind 25 “3D” fragment libraries. The tabular summary showing all the scaffolds emphasizes that most of these libraries are modest in size, with the largest being 102 members. Chemists will particularly enjoy the multiple synthetic schemes. The authors note the importance of “fragment sociability” to facilitate SAR and elaboration.
 
Covalent fragments
Special libraries are required for covalent fragment-based drug discovery, the most notable feature being the “warhead” that reacts with the protein target. These are the focus of a chapter in Adv. Chem. Prot. by Péter Ábrányi-Balogh and György Keserű of the Hungarian Research Centre for Natural Sciences. The review includes a table containing more than 100 warheads with associated mechanisms and amino acid selectivity.
 
The “reactivity of covalent fragments and their role in fragment-based drug design” is the focus in an (open access) Pharmaceuticals review by Kirsten McAulay and colleagues at the Beatson Institute. This is a nice overview of the field and contains several case studies. The authors conclude that “striking a balance between reactivity, potency and selectivity is key to identifying potential candidates.”
 
“Advances in covalent drug discovery” are reviewed (open access) by Dan Nomura and colleagues at University of California Berkeley in Nat. Rev. Drug Disc. This is a highly readable and comprehensive overview of the field. The authors differentiate between “ligand-first” approaches, in which a covalent warhead is appended to a known binder (such as here) and “electrophile-first,” in which “the initial discovery process is rooted in finding a covalent ligand from the outset,” such as for KRASG12C inhibitors.
 
Another broad overview of covalent inhibitors is provided by Juswinder Singh (Ankaa Therapeutics) in J. Med. Chem. Jus is a pioneer in the field, having published the first targeted covalent inhibitor in 1997. Of 1673 small molecules approved as drugs by the US FDA, only about 7% are covalent, and it wasn’t until recently that these have been intensively pursued. Part of the reluctance has been concerns over toxicity, but the paper suggests that – at least among kinase inhibitors – covalent drugs may actually be safer, perhaps due to conjugation of glutathione to the warhead and rapid clearance rather than formation of reactive metabolites.
 
Other
Whether covalent or not, thermodynamics plays a fundamental role in protein-ligand interactions, and this is the topic of an (open access) review in Life by Conceição Minetti and David Remeta of the State University of New Jersey. The paper covers a lot of ground, including drug discovery approaches, metrics (such as LE, LLE, etc.), isothermal titration calorimetry, case studies, and more. Importantly, the authors acknowledge the many challenges of applying thermodynamics to drug discovery, some of which we highlighted here.
 
Thermodynamics explains the potency increases longed for when doing fragment-linking, the subject of two reviews. In Chem. Biol. Drug Des. Anthony Coyne and colleagues at University of Cambridge provide a broad overview, starting with the historical theoretical background and newer developments. The bulk of the paper surveys published examples of fragment linking, with structure-based methods (whether X-ray, NMR, or computational) separated from target-guided methods such as DCC.
 
The second review, published in Bioorg. Chem. by Junmei Peng and colleagues at University of South China, is broader in scope, encompassing not just FBLD but also linkers used in PROTACs and even antibody-drug conjugates. The paper is organized by chemical structure of the linker.
 
Finally, in J. Med. Chem., Peter Dragovich, Wolfgang Happ, and colleagues at Genentech and Roche examine “small-molecule lead-finding trends” at their organizations between 2009 and 2020. (Although Genentech is fully owned by Roche, its research organization operates independently.) Fragment-based approaches led to only a small fraction of chemical series at Genentech and none at Roche. The authors note that leads derived from public sources such as patent applications were often found and pursued earlier, and that “purposeful dedication” of resources to fragment approaches may be necessary. Another major source of leads at Genentech is in-licensing, and some of these are fragment-derived.
 
And that’s it for 2022, year three of COVID-19. Thanks for reading and special thanks for commenting. May the coming year bring health, peace, and significant scientific progress.

07 November 2022

Fragments vs IL17A: merging and linking

One of the talks at the Discovery on Target meeting last month described the discovery of small molecule inhibitors of IL17A, a pro-inflammatory cytokine. Antibody-based drugs against this protein are useful for psoriasis and other diseases, but they require regular injections. Also, because the antibodies stick around for a long time and dampen the immune response, they could leave patients less able to combat an infection. An orally available small molecule could solve both these problems, but blocking protein-protein interactions is generally difficult. Early progress towards this goal has been published (open access) in Sci. Reports by Eric Goedken and collaborators at AbbVie.
 
The researchers started by 13C-labeling the methyl groups of isoleucine, leucine, valine, and methionine. They then performed a 2D NMR screen ([1H, 13C]-HSQC) of ~4000 fragments in pools of 12, with each fragment at 1 mM. This yielded multiple hits, including compound 4. Interestingly, the chemical shift perturbations (CSPs) caused by this fragment were distinct from those caused by known previously disclosed binders, suggesting a different binding site. In particular, one of these CSPs could be traced to a methionine residue. The full NMR assignment of the protein was not conducted, but modeling and mutagenesis narrowed the possibility down to a methionine near the C-terminus.
 
Surface plasmon resonance (SPR) experiments revealed that compound 4 had very low affinity, but two rounds of optimization led to compounds 5 and 6. At this point the researchers were able to solve the crystal structures of these molecules bound to IL17A. The protein exists as a homodimer, and the molecules bind symmetrically, with two copies per homodimer. Moreover, the two copies bind close to one another.
 

 
In addition to these fragments, the researchers had also identified compound 7, and a crystal structure revealed that this binds in a similar fashion. Merging compound 7 with compound 6 and linking two copies of the resulting monomer led ultimately to dimeric compound 10, with nanomolar affinity by both SPR and isothermal titration calorimetry (ITC). This molecule inhibited the binding of IL17A with its receptor in a biochemical assay and also showed low micromolar activity in a cellular assay.
 
This is a nice paper that bears some similarity to previous work we highlighted from AbbVie on a different cytokine, TNFα. There too fragment linking was used initially. As in the present case, that effort led to a drop in ligand efficiency and a significant increase in the size of the molecules, resulting in suboptimal pharmaceutical properties. Identifying drug-like small molecules has been an ongoing challenge for IL17A; peptide-based inhibitors and macrocycles have been found that bind to other sites on the protein, but many of these are also well beyond rule-of-five space. As the researchers conclude, “we look forward to seeing which of these sites prove to be the most amenable to producing optimized drug candidates.”

20 September 2021

Chemical couplets inhibit the GAS41 YEATS domain

Practical Fragments has covered bromodomains extensively, most recently just a couple months ago. But these epigenetic readers are not the only proteins that recognize acetylated lysine residues. Two years ago we highlighted fragment hits against one of the four YEATS domain proteins. A paper recently published in Cell Chem. Biol. by Jolanta Grembecka, Tomasz Cierpicki, and colleagues at University of Michigan tackles another member of the family.
 
The researchers were interested in the protein GAS41, which is amplified in multiple forms of cancer. The YEATS domain within this protein binds to acylated lysine residues in histone H3 proteins. However, unlike the deep pockets found in bromodomains, the acyl-lysine binding site in the YEATS domain consists of a partially solvent-exposed channel, making it a more challenging site to drug.
 
Nonetheless, an NMR-based screen of fragments (in pools of 10, each at 500 µM) led to compound 1, which produced multiple chemical shifts in a 1H-15N HSQC experiment. Two different competition assays, fluorescence polarization (FP) and AlphaScreen formats, confirmed that compound 1 could compete with H3-derived peptides. Fragment growing led to compounds 7 and 11. (All IC50 values below are from the FP assay; these are somewhat weaker than those from the AlphaScreen, but they more closely track binding affinities determined by isothermal titration calorimetry.)
 

A crystal structure of a compound closely related to compound 11 revealed that the molecule nearly fills the small binding channel, suggesting that further gains in affinity would be difficult. Indeed, no GAS41 inhibitors have been previously reported. However, the protein is dimeric, so the researchers decided to dimerize their molecule to bind to two YEATS domains simultaneously. This led to nanomolar molecules such as compound 19.
 
Not only was compound 19 potent in biochemical assays, it also disrupted binding of GAS41 to acetylated histone proteins in cells. Moreover, the compound inhibited growth of cancer cell lines with amplified GAS41.
 
This is a nice case study in fragment dimerization, an uncommon but interesting approach. The linking in this case led to a 44-fold improvement in affinity, which though impressive is far from synergistic, and is associated with a considerable loss in ligand efficiency. And although the micromolar potency of compound 19 in cells needs to be improved to generate a chemical probe, let alone a drug lead, these results nonetheless support the notion that targeting GAS41 could be a useful strategy for certain cancers.

02 August 2021

Linking fragments on DNA, revisited

Three years ago we discussed using DNA-encoded libraries to find and link fragments. In a new open-access Bioorg. Med. Chem. article, Nicolas Winssinger and colleagues at University of Geneva report a different version of this approach.
 
Rather than using DNA, the researchers constructed their libraries with peptide nucleic acids (PNAs), which can be assembled using traditional solid-phase peptide synthesis and will also hybridize to DNA. Each PNA is coupled to a different fragment, and the fragment-PNA molecules are then bound to microarrays of DNA such that two fragment-PNA molecules bind to a single DNA strand. In this case the researchers used 250,000 combinations of fragment pairs.
 
Next, a protein of interest (here the anti-apoptotic cancer target BCL-xL) was screened at 50 nM. Binding to specific pairs of fragments was assessed by fluorescent detection of the protein at various spots on the microarray.
 
Trying to figure out which of the fragments are best – and how to link them – is “not trivial,” so the researchers took a combinatorial approach. Based on the first screen, they generated a new library of 10,000 molecules in which 10 sulfonamide-containing fragments were linked to 100 heterocycle-containing fragments using 10 different linkers. These compounds were screened using the same microarray technology, and the best binders were then resynthesized with a biotin tag rather than the PNA.
 
The biotinylated molecules were able to pull down recombinant BCL-xL in solution. Two of them, including compound 80-28, were even able to pull down recombinant protein that was spiked into cell lysate. Importantly, neither fragments 80 nor 28 did this by themselves. The affinity of 80-28 was measured by SPR to be 96 nM. 
 
 
Finally, the researchers tested 80-28 in K562 cells and found that it was cytotoxic with EC50 = 1.7 µM. They compare this favorably to venetoclax, the second fragment-based drug to be approved. However, this is a disingenuous comparison: venetoclax was specifically designed to bind less tightly to BCL-xL than to the related protein BCL-2. A more appropriate comparison would be a molecule such as navitoclax or the specific BCL-xLbinder A-1155463.
 
Like most BCL-family binders, compound 80-28 is also a rather unusual looking molecule, with a high molecular weight of 735. Unlike navitoclax and venetoclax, it also has 7 hydrogen bond donors and many more rotatable bonds. The long floppy linker in particular is something the earlier DNA-based fragment-linking work sought to fix. As we noted then, such linkers may be an inherent liability with the approach. From a technology perspective this is interesting work. But from a drug discovery perspective it still has some way to go to prove itself practical.

05 April 2021

A general fragment-based approach to… targeting RNA?

This is taken from the title of a recent open-access paper by Matthew Disney and collaborators at Scripps Research Institute Jupiter and Florida Atlantic University in Proc. Nat. Acad. Sci. USA. RNA has long been a target of FBLD: Practical Fragments first blogged about it in 2009, and a 2002 paper reported using fragment linking to obtain a low micromolar binder. So how general is the new approach?
 
The researchers describe chemical cross-linking and isolation by pull-down fragment mapping (Chem-CLIP-Frag-Map). This involves using photoaffinity probes that can crosslink to biomolecules such as RNA. The probes also have an alkyne tag that can be used to isolate bound molecules using click chemistry. We’ve written previously about such “fully functionalized fragments” (FFFs).
 
Earlier work had resulted in the identification of compound 1, which binds to a specific site on pre-miR-21, the precursor to a non-coding microRNA linked to cancer. An FFF version of compound 1 was shown to crosslink to pre-miR-21 after irradiation with UV light, and the site of modification could be mapped using a reverse-transcriptase-mediated primer extension, which stalled at the modified bases.
 
Next, the researchers screened 460 FFFs at 100 µM and found 21 that crosslinked to pre-miR-21. They were ultimately looking to link fragments with compound 1, and thus competition studies were used to eliminate fragments that bound at the same site. This left three fragments, and primer extension studies confirmed that these bound near but not at the binding site of compound 1.
 
Next, the researchers attached these three fragments to compound 1, with or without various linkers. Some of the resulting molecules had improved affinity, and compound 9 showed the tightest binding according to microscale thermophoresis (MST). Mutational and competition studies confirmed that the molecule binds to the expected site. Importantly, compound 9 not only bound to pre-miR-21, it also blocked processing by the enzyme Dicer. Moreover, it showed activity in cell models consistent with inhibition of pre-miR-21.
 

This is a nice paper, but there are several limitations. First, compound 9 is still a fairly modest binder with lackluster ligand efficiency. Indeed, while potency can be overrated, I would love to see a fully synthetic low nanomolar RNA binder. Second, while the approach may be general, it is not necessarily easy, and it requires specialized fragments. And as we noted last year, there is no relation between crosslinking efficiency and affinity. I wish the researchers had tried linking some of the non-selected fragments to see whether these were false negatives. Indeed, given the complexity of the approach, I wonder if the researchers would have been better off simply making and testing an anchor library around compound 1, in a similar fashion as described here.
 
But whether or not Chem-CLIP-Frag-Map turns out to be the solution to targeting RNA, I wholeheartedly agree with the conclusion: “It may be time to describe biomolecules that are perceived to be challenging small molecule targets as ‘not yet drugged’ rather than ‘undruggable.’”