Showing posts with label antimicrobial. Show all posts
Showing posts with label antimicrobial. 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.

21 October 2024

Fragments vs LpxC revisited

Back in 2020 we described fragment-derived inhibitors of the highly conserved bacterial enzyme LpxC, which is essential for biosynthesis of the outer membrane in Gram-negative bacteria. In a recent (open access) paper in J. Med. Chem., a different group consisting of Ralph Holl and collaborators at Universität Hamburg and several other academic centers describe a new series.
 
The researchers started with compound 9, a molecule they had previously discovered. The substrate for LpxC is a rather large small molecule called (UDP)-3-O-[(R)-3-hydroxymyristoyl]-N-acetylglucosamine. Compound 9 does not occupy the UDP-binding site, so the researchers initially tried building towards it with a series of simple linkers connected to a phenyl group. The (S) enantiomers tended to be more active than the (R)-enantiomers, and the most potent was compound (S)-13a, which showed sub-micromolar activity against LpxC from E. coli as well as P. aeruginosa in an enzymatic assay. (For simplicity only the E. coli data are shown here.)
 
Seeking to improve affinity, the researchers screened 650 fragments in pools of five against LpxC in the presence of compound 9 using STD NMR and WaterLOGSY. After deconvolution, this led to 97 hits. STD-based epitope mapping, which we wrote about here, was used to prioritize fragments likely to have a single, well-defined binding mode, culling the number to 19. Finally, NMR-ILOE experiments (see here) suggested that nine of this set bound in close proximity to compound 9, while the other ten did not. Four of these fragments, including the simple indole F3, were then linked to compound 9 at various positions. This is akin to SAR by NMR, but with less information about the relative binding modes so more trial and error is necessary.
 

Among the roughly two dozen molecules made, compound (S)-13j was the most potent against LpxC, with low nanomolar activity. This compound (and several others) also showed antibacterial activity against E. coli and several other strains of Gram-negative bacteria. In vitro stability studies of compound (S)-13j were promising, though the researchers noted the need for improvement. And, since the molecule contains a hydroxamic acid moiety potentially capable of binding to multiple metalloproteins, it was tested against a handful of mammalian zinc-dependent enzymes and shown to be nearly inactive.
 
Compound (S)-13j is 15-fold more potent than the simple phenyl analog (S)-13a, and molecular modeling suggested this may be due to a hydrogen bond from the protein to the indole NH. Although one could argue that it would have been possible to arrive at compound (S)-13j using standard medicinal chemistry starting from (S)-13a, this may have taken longer without knowledge of the indole fragment. Whether or not the molecules advance further, this is a nice example of using fragment screening to find a second-site binder to improve affinity of an existing lead.

02 September 2024

Fragments in Brazil

Most of the fragment events we’ve highlighted are in the US, Europe, and Australia, but that does not fully reflect where all the good science is happening. In a recent ACS Med. Chem. Lett. paper, Carolina Horta Andrade, Maria Cristina Nonato, and Flavio da Silva Emery introduce CRAFT: the Center for Research and Advancement in Fragments and molecular Targets.
 
Established in 2021, CRAFT is a collaboration between the University of Saõ Paulo and the Federal University of Goiás. The center is focused on endemic diseases of Brazil. As the researchers note, only one of the 60 or so fragment-derived drugs that have entered the clinic is an anti-infective, so there is clearly significant need. CRAFT also has an educational and training component reminiscent of the European FragNet and the Australian Centre for Fragment-Based Design.
 
One focus of CRAFT is fragment library design, including underexplored heterocyclic systems. Importantly, the researchers are investigating new synthetic methodologies to be able to functionalize different regions of the fragments. They are also exploring fragments similar to or derived from natural products.
 
Targets are of course essential, and CRAFT is investing in protein production and characterization, such as the enzyme DHODH from Leishmania; we’ve written recently about a fragment approach to the mammalian counterpart.
 
Finally, CRAFT is investing in structure-based design, ligand-based design, and phenotypic screening. And in 2024 no venture would be complete without use of machine learning.
 
Academic laboratories often struggle with downstream drug discovery efforts such as drug metabolism and pharmacokinetics. CRAFT recognizes this and has partnered with the Welcome Centre for Anti-Infectives Research to train participants in DMPK.
 
The researchers “invite the global scientific community to collaborate with us in addressing neglected diseases.” I hope they succeed. Five years ago we highlighted the consortium Open Source Antibiotics, but that site seems to be updated infrequently. The COVID Moonshot has been more successful but is arguably less urgent given the billions of dollars of industry money that poured into research on SARS-CoV-2. From an ethical perspective society should invest more on combating tropical diseases. And as the planet warms, these diseases will increasingly move out of the tropics.

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 December 2020

Fragments vs LpxC, two ways

Gram negative bacteria such as Pseudomonas aeruginosa are a continuing threat, and antibacterial drug discovery is not keeping pace. The enzyme UDP-3-O-acyl-N-acetylglucosamine deacetylase (LpxC) is critical for the synthesis of the bacterial cell wall lipopolysaccharide. In a new J. Med. Chem. paper, Yousuke Yamada, Rod Hubbard, and collaborators at Taisho and Vernalis describe progress against this target.
 
LpxC is a zinc hydrolase, and although previous potent inhibitors have been reported against the metalloenzyme, these contained hydroxamate moieties. Unfortunately, hydroxamic acids are rather nonspecific zinc binders, and many of them hit human enzymes such as HDACs and MMPs. Thus, the researchers turned to fragments to find new metallophilic starting points.
 
The 1152 members of the Vernalis fragment library were screened against LpxC using three NMR experiments: STD, WaterLOGSY, and CPMG in pools of six. This yielded a remarkable 252 hits in at least one assay. These were retested individually and for competition with a substrate pocket-binding small molecule, resulting in 28 hits, two of which were advanced.
 
A crystal structure of compound 6 bound to LpxC suggested that adding a hydroxyl group could make additional interactions with the protein, and this was confirmed in the form of compound 10. Further fiddling in this region of the molecule was not successful, and the phenyl ring did not provide good vectors to a hydrophobic tunnel. However, replacing the phenyl with a more shapely piperidine yielded compound 17. Although this molecule had slightly lower affinity, it did provide a better starting point for further optimization, ultimately leading to compound 21, with low nanomolar potency against LpxC. Unfortunately, this and other members of the series showed only weak antibacterial activity.
 


Compound 9 was weaker than the other fragment starting point, but making and testing related compounds led to improved binders such as compound 27. This was the first molecule in this series to be structurally characterized, and crystallography revealed that the imidazole was making a single interaction with the zinc at the heart of the LpxC active site. Adding a hydroxyl led to bidentate chelator 29 (i.e. two interactions with the zinc) that had better activity, and further structure-based design ultimately led to low nanomolar inhibitors such as compound 43. In contrast to the other series, this one did show antibacterial activity, and the researchers eventually discovered molecules with in vivo efficacy. Both series were also selective against a small panel of human metalloproteases.
 
 
This is a nice fragment to lead story (expect it to be included in the next compilation). As the researchers note, it provides two important lessons. First, fragments can provide multiple different starting points for a target. Second, because fragment libraries tend to be small, it can be valuable to take some time to refine a fragment before launching into fragment growing or merging. Indeed, compound 38 (itself fragment-sized) contains only four more atoms than the initial fragment hit, yet has more than a thousand-fold higher affinity. During lead optimization you often need to add molecular weight, lipophilicity, and possibly polar atoms, so it is crucial to get the core binding elements as good as possible.

29 April 2019

Help develop new antibiotics from fragments!

The state of antibiotic drug discovery is – to put it mildly – dangerously poor. Not only do you have all the challenges inherent to drug discovery, you’re dealing with organisms that can mutate more rapidly than even the craftiest cancer cells. And then there’s the commercial challenge: earlier this month the biotech company Achaogen filed for Chapter 11 bankruptcy, less than a year after winning approval for a new antibiotic.

As Douglas Adams’s Golgafrincham learned, complacency about microbial threats is suicidal. But what can any one of us do? Chris Swain, whom we’ve previously highlighted on Practical Fragments, is involved with a consortium of researchers called Open Source Antibiotics. Their mission: “to discover and develop new, inexpensive medicines for bacterial infections.” And they are asking for our help. More on that below.

The researchers initially chose to focus on two essential enzymes necessary for cell wall biosynthesis, MurD and MurE, both of which are highly conserved across bacteria and absent in humans. They conducted a crystallographic fragment screen of both enzymes at XChem, soaking 768 fragments individually at 500 mM concentration. As we’ve written previously, you’ll almost always get hits if you screen crystallographically at a high enough concentration.

For MurD, four hits were found, all of which bind in the same pocket (in separate structures). Interestingly, this pocket is not the active site, but adjacent to it. The binding modes of the fragments are described in detail here, and the researchers suggest that growing the fragments could lead to competitive inhibitors. The fragments also bind near a loop that has been proposed as a target for allosteric inhibitors, so growing towards this region of the protein would also be an interesting strategy.

MurE was even more productive, with fragments bound at 12 separate sites. (Though impressive, that falls short of the record.) Some of these sites are likely artifacts of crystal packing, or so remote from the active site of the enzyme that they are unlikely to have any functional effects. However, some fragments bind more closely to the active site, and would be good candidates for fragment growing.

If this were a typical publication one might say "cool," and hope that someone picks up on the work sometime in the future. But this, dear reader, is different.

The researchers are actively seeking suggestions for how to advance the hits. Perhaps you want to try running some of these fragments through the Fragment Network? Or do you have a platform, such as “growing via merging,” AutoCouple, or this one, that suggests (and perhaps even synthesizes) new molecules? Perhaps you want to use some of the fragments to work out new chemistry? The consortium has a budget to purchase commercial compounds, and will also accept custom-made molecules. In addition to crystallography, they have enzymatic assays, and are building additional downstream capabilities.

The Centers for Disease Control identifies antibiotic resistance as one of the most serious worldwide health threats. Some have called for a global consortium—modeled after the International Panel for Climate Change—to tackle the problem. But in the meantime, you can play a role yourself. If you would like to participate, you can do so here. The bugs are not waiting for us – and they are already ahead.

24 August 2015

Fragment-Based Drug Discovery

This is the straight-to-the-point title of a new book published by the Royal Society of Chemistry, edited by Steven Howard (Astex) and Chris Abell (University of Cambridge). It is the second book on the topic published so far this year, and it is a testimony to the fecundity of the field that the two volumes have very little overlap.

After a brief forward by Harren Jhoti (Astex) and a preface by the editors, the book opens with two personal essays. The first, by me, is something of an apologia for Practical Fragments and the growing role of social media in science (and vice versa). If you’ve ever wondered how this blog got started or why it keeps going, this is where to find out. The second essay is by Martin Drysdale (Beatson Institute). Martin is a long-time practitioner of FBDD, dating back to his early days at Vernalis (when it was RiboTargets) and he tells a fun tale of “adventures and experiences.”

Chapter 1, by Chris Abell and Claudio Dagostin, is entitled “Different Flavours of Fragments.” With a broad overview of the field it makes a good introduction to the book. There are sections on fragment identification, including the idea of a screening cascade, as well as several case studies, some of which we’ve covered on Practical Fragments, including pantothenate synthetase, CYPs, RAD51, and riboswitches.

The next two chapters deal with two of the key fragment-finding methods. Chapter 2, by Tony Giannetti and collaborators at Genentech, GlaxoSmithKline, and SensiQ, covers surface plasmon resonance (SPR). This includes an extensive discussion of data processing and analysis, which is critical for improving the efficiency of the technique. Competition studies are also described, as are advances in hardware, notably those from SensiQ. This is a good complement to Tony's 2011 chapter.

Chapter 3, by Isabelle Krimm (Université de Lyon), provides a thorough description of NMR methods, both ligand-based (STD, WaterLOGSY, ILOE, etc) and protein-based (mostly HSQC). The chapter does a nice job of describing techniques in terms a non-specialist can understand while also providing practical tips on matters such as optimal protein size and concentration.

Chapter 4, by Ian Wall and colleagues at GlaxoSmithKline, provides an overview of FBLD from the viewpoint of computational chemists. The chapter includes some interesting tidbits, such as the observation that fragment hits that yield crystal structures tend to be less lipophilic but also contain a smaller fraction of sp3 atoms and more aromatic rings. The researchers note that the current fashion for “3D” fragments is yet to be experimentally validated. They also include accessible sections on modeling, druggability, and integrating fragment information into a broader medicinal chemistry program.

The remaining chapters focus on specific types of targets. Chapter 5, by Miles Congreve and Robert Cooke (both at Heptares) is devoted to G protein-coupled receptors (GPCRs). This includes descriptions of how to screen fragments against these membrane proteins using SPR, TINS, CE, thermal melts, and competition binding. It also includes a detailed case study of their β1 adrenergic receptor work (summarized here). Congreve and Cooke assert that, although many of the GPCR targets screened to date have been highly ligandable, technical challenges only now being addressed have caused this area of research to lag about a decade behind other targets. They predict a bright future.

Rod Hubbard (Vernalis and University of York) turns to protein-protein interactions in Chapter 6. After describing why these tend to be more challenging than most enzymes and covering some of the methods for finding and advancing fragments, he then presents several case studies, including FKBP (one of the first targets screened using SAR by NMR), Bcl-2 family members (including Bcl-xL and Mcl-1), Ras, and BRCA2/RAD51. He concludes with a nice section on “general lessons,” which boils down to “patience, pragmatism, and integration.” As Teddy recently noted, this can lead to substantial rewards.

Allosteric ligands have potential advantages in terms of selectivity and addressing otherwise challenging targets, and in Chapter 7 Steven Howard (Astex) describes how fragments can play a role here. This includes how to establish functionality of putative allosteric binders, as well as case studies such as HIV-1 RT, FPPS, and HCV NS3. Astex researchers have recently stated that they find on average more than two ligand binding sites per protein, and this chapter includes a table listing these (including 5 binding sites each on bPKA-PKB and PKM2).

The longest chapter, by Christina Spry (Australian National University) and Anthony Coyne (University of Cambridge) describes fragment-based discovery of antibacterial compounds. After discussing some of the challenges, the authors report several in depth case studies including DNA gyrase, DNA ligase, CTX-M, AmpC, CYP121, and pantothenate synthetase, among others. At least one fragment-derived antibacterial agent entered the clinic; hopefully more will follow.

Chapter 9, by Iwan de Esch and colleagues at VU University Amsterdam, focuses on acetylcholine-binding proteins (AChBPs), both as surrogates for membrane-bound acetylcholine receptors and as well-behaved model proteins on which to hone techniques (see for example here, here, and here). Since AChBPs have evolved to bind fragment-sized acetylcholine, these proteins can bind tightly to small ligands; 14-atom epibatidine binds with picomolar affinity, for example, with a ligand efficiency approaching 1 kcal mol-1 atom-1.

And Chapter 10, by Chun-wa Chung and Paul Bamborough at GlaxoSmithKline, concisely covers epigenetics. Bromodomains are well-represented, including a table of ten examples (see for example here, here, here, here, here, and here). Happily, although some of these projects started from similar or identical fragments, the final molecules are quite divergent. However, the authors note that much less has been published on histone-modifying enzymes, such as demethylases and deacetylases, perhaps reflecting the challenges of achieving specificity with what are often metalloenzymes.

Finally, this is the 500th post since Teddy founded Practical Fragments way back in the summer of 2008. Thanks for reading, and special thanks for commenting!

02 February 2015

Fragments vs DsbA: targeting bacterial virulence

The quest for new antibacterial agents can seem quixotic: no sooner have you found a killer molecule than the bugs have developed resistance to it. Evolution is hard to beat, particularly when it comes down to life or death. But what if you could lower the stakes? Many bacteria express virulence factors that are not essential for survival but are important for colonizing their host. Perhaps targeting these would be less prone to generating resistance.

Virulence factors often contain disulfide bonds, and the bacterial protein DsbA is essential for catalyzing their formation. In a paper published recently in Angew. Chem. Int. Ed., Begoña Heras (formerly University of Queensland), Jamie Simpson and Martin Scanlon (Monash University) and collaborators describe a fragment-based approach against the E. coli. version of this target. (See also here for Derek Lowe’s thoughts.)

The researchers started with an STD NMR screen of an 1132 fragment library from Maybridge, with compounds in pools of 3 to 5 (each at 0.3 mM). This yielded 171 hits, 37 of which showed appreciable chemical shift perturbations (CSPs) in a two-dimensional HSQC 15N-1H NMR assay. All of these were relatively weak, with none showing saturation at 1 mM fragment concentration, but they all appeared to be binding in a hydrophobic groove adjacent to the active site.

The 37 hits clustered into eight different structural subclasses, one of which – the phenylthiazoles – is described in detail. The Monash fragment library was designed with SAR-by-catalog in mind, and 22 commercial analogs were purchased and tested in the HSQC assay to assess the SAR. Several of the compounds were soaked into crystals of DsbA, in one case leading to a structure in which two fragments were bound stacked on top of each other in the hydrophobic groove. However, this binding mode was inconsistent with the NMR data, and indeed co-crystallography of the same fragment revealed a 1:1 complex with the protein, also in the hydrophobic groove. (As an aside, this is an interesting case of crystal soaking and co-crystallization giving different results; are readers aware of others?)

The crystal structure was used to inform fragment-growing, ultimately leading to molecules with dissociation constants around 0.2 mM as assessed by surface plasmon resonance and with similar IC50 values in a functional assay. One of these compounds was also tested against E. coli. DsbA is not needed for bacterial growth in rich media but is necessary for motility, and happily the assays showed just this – the compound did not affect growth but did inhibit cell motility.

Although the molecules are still too weak to answer the question of whether targeting DsbA will be a viable antibacterial strategy in vivo, this paper presents promising starting points, along with a wealth of data (including 78 pages of supporting information!) And if you want to learn more, Martin Scanlon is one of the organizers of the FBLD symposium at Pacifichem this December – so you can ask him questions in person in Honolulu!

13 August 2014

Intentionally dirty fragments

Practical Fragments has tried to publicize the dangers of pan-assay interference compounds, or PAINS. These compounds show up as nuisance hits in lots of assays. So what are we to make of a new paper in Curr. Opin. Microbiol. by Pooja Gopal and Thomas Dick, both at the National University of Singapore, entitled “Reactive dirty fragments: implications for tuberculosis drug discovery”?

As the researchers point out, several approved anti-tuberculosis drugs are fragment-sized and hit multiple targets; they are “dirty drugs”. For example, isoniazid (MW 137, 10 heavy atoms), is an acylhydrazide that is metabolically activated and forms an adduct with an essential cofactor, causing havoc to the pathogen. Ethionamide (MW 166, 11 heavy atoms), a thioamide, works similarly. The fact that these molecules are so small probably allows them easier passage through the microbe’s rather impermeable cell membrane, and the fact that they hit multiple targets may make it more difficult for the organism to develop resistance. The researchers conclude:
The success of small dirty drugs and prodrugs suggests that fragment-based whole cell screens should be re-introduced in our current antimycobacterial drug discovery efforts.
While it is true that many antimicrobials do have reactive warheads, and it is also true that there is a huge need for new antibiotics, I worry about this approach. Not only is there an increased risk of toxicity (isoniazid in particular has a long list of nasty side effects), it can be very hard to determine the mechanism of action for these molecules, complicating optimization and development. As evidence, look no further than pyrazinamide (MW 123, 9 heavy atoms). Despite being used clinically for more than 60 years, the mechanism remains uncertain.

Fragment-based lead discovery is typically more mechanistic: find an ideal molecule for a given target. Indeed, much of modern drug discovery takes this view. Gopal and Dick propose a return to a more phenomenological, black-box approach. This may have value in certain cases, but at the risk of murky or worse misleading mechanisms.

If you do decide to put PAINS into your library, you might want to read a new paper in Bioorg. Med. Chem. by Kim Janda and collaborators at Scripps and Takeda. They were interested in inhibitors of the botunlinum neurotoxin serotype A (BoNT/A), which causes botulism.

Since BoNT/A contains an active-site cysteine, the researchers decided to pursue covalent inhibitors, and the warheads they chose, benzoquinones and napthoquinones, are about as PAINful as they get. However, in contrast to other groups, they went into this project with their eyes wide open to the issue of selectivity and examined the reactivity of their molecules towards glutathione. Reaction with this low molecular weight thiol suggests that a compound is not selective for the protein. Not surprisingly, selectivity was generally low, though a few molecules showed some bias toward the protein.

The researchers also tried building off the benzoquinone moiety to target a nearby zinc atom, and although they were able to get low micromolar inhibitors, these no longer reacted with the cysteine; apparently when the ligand binds to zinc, the protein shifts conformation such that the cysteine residue is no longer accessible.

To return to the premise of Gopal and Dick, there can be a therapeutic role for dirty molecules. The fact that dimethyl fumarate is a highly effective blockbuster drug for multiple sclerosis calls for a certain degree of humility. However, if you do decide to pursue PAINS, you should do so in full awareness that your road to a drug – not to mention a mechanism – will likely be much longer and more difficult.

05 May 2014

Biofragments: extracting signal from noise, and the limits of three-dimensionality

What does this protein do? Now that any genome can be sequenced, this question gets raised quite often. In many cases it is possible to give a rough answer based on protein sequence: this protein is a serine protease, that one is a protein tyrosine kinase, but figuring out the specific substrates can be more of a challenge. In a recent paper in ChemBioChem, Chris Abell and collaborators at the University of Cambridge and the University of Manchester attempt to answer this question with fragments.

The bacterium Mycobacterium tuberculosis (Mtb), which causes tuberculosis, has 20 cytochrome P450 proteins (CYPs), heme-containing enzymes that usually oxidize small molecules. Although some are essential for the pathogen, it is not clear what many of them do. The researchers used an approach called “biofragments” to try to pin down the substrate of CYP126.

The biofragments approach starts by selecting a collection of fragments based on known substrates. Of course, the specific substrates are not known, so in this case the researchers started with a set of several dozen natural (ie, non-synthetic) substrates of various other CYPs, both bacterial and eukaryotic. They then computationally screened the ZINC database of commercial molecules for fragments most similar to these substrates and purchased 63 of them. Perhaps not surprisingly given their similarity to natural products, these turned out to be more “three-dimensional” than conventional fragment libraries, as assessed both by the fraction of sp3 hybridized carbons and by principal moment-of-inertia.

Next, the researchers screened their fragments against CYP126 using three different NMR techniques (CPMG, STD, and WaterLOGSY). Since they were primarily interested in hits that bind at the active site, they also used a displacement assay in which the synthetic heme-binding drug ketoconazole was competed against fragments. This exercise yielded 9 hits – a relatively high 14% hit rate.

Strikingly, all of the hits are aromatic, and 7 of them could reasonably be described as planar. In other words, even though the biofragment library was relatively 3-dimensional, the confirmed hits were some of the flattest in the library! The researchers interpreted this to mean that “CYP126 might preferentially recognize aromatic moieties within its catalytic site,” but there could be something more general going on – perhaps aromatics are simply less complex, and thus more promiscuous.

Examining the fragment hits more closely, the researchers found that one of them – a dichlorophenol – produced a spectrophotometric shift similar to that produced by substrates when bound to the enzyme. This led them to look for similar structures among proposed Mtb metabolites. Weirdly, pentachlorophenol came up as a possible hit, and a spectrophotometric shift assay reveals that this molecule does have relatively high affinity for CYP126. Whether this is a biologically relevant substrate for the enzyme remains to be seen.

This is an intriguing approach, but I do have reservations. First, in constructing fragment libraries based on natural products, it is essential to avoid anything too “funky”. The Abell lab is one of the top fragment groups out there, well aware of potential artifacts, and has a long history of studying CYPs, but researchers with less experience could easily populate a library with dubious compounds.

More fundamentally though, I wonder about the basic premise of biofragments. The whole point of fragments is that they have low molecular complexity and are thus likely to bind to many targets, so is it realistic to try to extract selectivity data from them? Indeed, as we’ve seen (here and here), fragment selectivity is not necessarily predictive of larger molecules.

That said, the approach is worth trying. Even if it doesn’t ultimately lead to new insights into proteins’ natural substrates, it could lead to new inhibitors.

23 October 2013

Fragment merging revisited: CYP121

Last year we highlighted a paper from Chris Abell and colleagues at the University of Cambridge in which they applied FBLD to CYP121, a potential anti-tuberculosis target. Several fragments with different binding modes were identified, and while some could be successfully merged to produce higher affinity binders, others couldn’t. In a new paper in ChemMedChem, the researchers take a closer look at why some of their initial attempts at fragment merging failed, and figure out how to succeed.

In the original paper, fragment 1 was particularly interesting for two reasons. First, crystallography revealed that it did not make direct interactions with the enzyme’s heme iron, as do most inhibitors of CYPs, suggesting that higher specificity might be achievable. Moreover, the co-crystal structure revealed that fragment 1 could bind in two nearly overlapping orientations, practically begging to be merged. Unfortunately, the resulting merged compound 4 actually bound worse than the initial fragment.


Computational modeling suggested that a primary reason for this disappointing result is the steric clash between two hydrogen atoms on the two phenyl rings of compound 4. These are forced into an unfavorable configuration when the molecule binds the protein. To fix this, the researchers sought to introduce a new interaction with the protein that would allow the molecule to relax into a lower energy conformation, alleviating the steric clash. This led to compound 5, with a satisfying increase in affinity. But lest folks become too cocky, an attempt to pick up an additional hydrogen bond to the protein (compound 8) actually led to a decrease in affinity despite the presence of the designed hydrogen bond, as assessed by crystallography. More successfully, building into a cavity led to the most potent compound 9. (Geeky aside: the aminopyrazole versions of fragment 1 had similar affinities as fragment 1, suggesting that the aminopyrazole moiety per se only gives a boost in potency in the context of the merged molecule.)

High-resolution co-crystal structures were solved for several of the molecules; the figure below uses color-coded carbons to show the overlay of the two different binding modes of fragment 1 (green), compound 4 (cyan) and compound 5 (magenta). What’s striking is how closely all the molecules superimpose, despite their very different affinities.


This is a nice case study in fragment merging that emphasizes just how difficult the strategy can be, even when it looks like it should be easy. And while Practical Fragments has not always looked kindly on computational methods, this is a beautiful example of how modeling can be used to understand why things that look good on paper don’t work, as well as how to fix them.

14 September 2012

Fragments vs CYPs – on purpose

The cytochrome P450 enzymes, or CYPs, are a huge class of oxidizing enzymes found across all kingdoms of life. In humans these enzymes metabolize many drugs, and to avoid drug-drug interactions, drug hunters generally shun or re-engineer molecules that inhibit CYPs. But microorganisms such as Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, also contain CYPs, and targeting these could lead to a sorely needed new treatment for this disease. A team led by Chris Abell at the University of Cambridge has published just such a strategy in Angew. Chem. Int. Ed.

The researchers were interested in CYP121, which is unique to Mtb and important for its viability. They used a thermal-shift assay to screen 665 commercial fragments at 5 mM, of which 66 increased the melting point by at least 0.8 ˚C. 56 of these were further characterized by STD and WaterLOGSY NMR, and 26 showed interactions with the protein and could also be competed with a known substrate. Eight of the most soluble of these were then soaked into crystals of CYP121, leading to four high-resolution structures, three of which are shown here. Isothermal titration calorimetry was used to determine dissociation constants.


 Interestingly, two of these fragments (1 and 2 in blue and red, respectively) coordinate to the heme iron, while the other two do not. Fragment 4 (green in figure above) showed two binding modes in the crystal structure, leading the researchers to make molecules that merge both binding modes. Although the resulting molecules bind in a similar fashion as compound 4 as judged crystallographically, they show at best marginal improvements in affinity and sizable losses in ligand efficiency. Quantum mechanical calculations suggested that this lack of improvement was due to conformational strain within the molecules.

Happily, merging compounds 1 and 2 was much more successful, leading to compound 14, which maintained ligand efficiency and improved affinity. A crystal structure revealed that, as designed, compound 14 binds in a very similar manner as the initial fragments. The molecule was also selective for CYP121 over a different CYP from Mtb as well as several human CYPs.


This is a nice paper not only because it reports a successful example of fragment merging on a new class of targets, but because it also describes several approaches that didn’t work. Fragment merging and fragment linking probably fail more often than they succeed, and this report really digs into the SAR and addresses why merging can be so challenging.

Of course, what would be really cool would be to link compound 14 with compound 4 (ie, link all of the fragments in the top figure), and the paper ends with the statement that this is currently ongoing. It will be fun to see the results.

24 August 2012

ACS Fall Meeting 2012


I recently returned from Philadelphia, where the American Chemical Society held its 244th national fall meeting. As always this was a massive affair, but fragments were well-represented, particularly in a nice session organized by Percy Carter, Debbie Loughney, and Romyr Dominique.

I opened the session by giving an introduction to fragment-screening, as well as an overview of some of the work we’re doing at Carmot. Andrew Good had perhaps the best title (“Fragment fat wobbles too”), and discussed some of the work done at Genzyme on Pim-1 kinase. Eric Manas next described some of the computational tools being used at GlaxoSmithKline, in particular strategies to deal with water. He also discussed the utility of looking for fragment analogs early in a project. In the last talk before the intermission, Chris Abell from the University of Cambridge described a number of projects from his group, starting with antimicrobial targets (such as this one); we’ll cover another in a separate post. Chris is unabashedly going after difficult targets, not just protein-protein interactions, but oligonucleotides – specifically riboswitches. There is only limited precedent for targeting RNA with fragments, so it will be fun to see how this progresses.

Francisco Talamas next described a nice example from Roche using FBLD to discover hepatitis C NS5B Palm I allosteric inhibitors. An HTS campaign of around 900,000 molecules yielded just 3 hits, none of which were advanced. A fragment screen of about 2700 fragments gave a better hit rate (5.9%), but of the 29 co-crystal structures attempted only a single structure was obtained. However, by combining the information from this crystal structure with information from other crystal structures, both proprietary and public, the researchers put together a set of rules to design a de novo fragment library tailored to this protein. This effort ultimately yielded compounds that were optimized to a clinical candidate.

Next, Nick Wurtz from Bristol-Myers Squibb described his company’s approach to discover neutral Factor VIIa inhibitors. The researchers used a combination of computational, functional, and biophysical approaches to find uncharged fragments that would bind in the P1 pocket, leading to a couple dozen crystal structures. Despite the low affinities of these fragments (typically mM), many of them could successfully be merged onto an existing series, replacing a positively charged moiety to yield potent molecules with better permeability. This is the first time I’ve seen a fragment story out of BMS, so I'm glad to see that they’re active in this area. This is also a prime example of what has been described as fragment-assisted drug discovery.

Finally, Prabha Ibrahim of Plexxikon gave a lovely overview of the discovery and development of vemurafenib, including a more detailed description of the SAR than has been presented in their earlier papers.

In addition to this dedicated session, there was a scattering of other talks and posters, including a notable poster from Timothy Rooney at the University of Oxford using fragment-based approaches to discover bromodomain inhibitors, a target class we’ve previously discussed.

A session entitled “A medicinal chemist’s toolbox” ranged over several topics of interest. Ernesto Freire of Johns Hopkins gave a great overview of thermodynamics in drug discovery, a topic we’ve previously covered. Most readers are probably familiar with the concept of enthalpy-entropy compensation, in which (for example) an added hydrogen bond fails to achieve the desired boost in potency due to unfavorable entropy. Recognizing this, he suggested that one should target groups in proteins that are already well-structured, so you don’t have to pay the cost of structuring a disordered part of the protein. He also suggested that if you introduce one hydrogen bond, you might be better off introducing a second one too, as the incremental entropic cost is likely to be low.

György Keserű of Gedeon Richter discussed the importance of avoiding lipophilicity by using tools such as LELP, which we’ve covered here and here. Continuing this theme, Kevin Freeman-Cook of Pfizer described two examples of using LLE in lead discovery programs, in particular calculating LLE values before making compounds. Although this may seem obvious, what was quite striking was the dramatic effect subtle changes in structure could make to ClogP values.

Of course, these are just a few of thousands of presentations. Please feel free to point out any that caught your eye, or expand on some of those mentioned above. And just a reminder, it’s only 4 weeks to FBLD 2012 in San Francisco – the biggest fragment event of the year!

08 January 2009

Ligand efficiency for antibiotics

Back in October of last year we highlighted a paper in Science that disclosed a new antibiotic targeting the bacterial protein FtsZ. The compound was derived through fragment-based techniques, though at the time no details were provided. A new paper in BMCL now provides some of the early medicinal chemistry, and also introduces an interesting new tool for evaluating antibiotics.

As mentioned in the Science paper, the researchers (led by Prolysis but with a number of contributors from Evotec and Key Organics) started with the fragment-like (MW = 151, 11 heavy atoms) 3-methoxybenzamide. An initial survey of “SAR by catalog” soon moved to the synthesis of analogs that could be assembled in up to four steps from commercially available compounds. This study found that the amide was essential, and only limited substitutions around the aromatic ring were tolerated. Turning to the alkoxy group, the authors took the classic “methyl, ethyl, butyl” approach, but kept going all the way to dodecyl. Intriguingly, a nonyloxy substituent proved to be optimal, better than either 8 or 10 carbon chains. Adding two fluorine atoms to the aromatic ring improved the potency further. Although the paper does not describe the final push to PC190723, the authors do describe the desire to replace the long alkyl chain and its likely attendant problems.



The paper also defines an interesting variation of ligand efficiency:

Antibacterial efficiency = -ln (MIC) / N, where
MIC = minimum inhibitory concentration (mg/ml) and
N = non-hydrogen atoms

Although the metric has a few quirks (for example, low-efficiency compounds can actually have negative numbers), “good” values correspond roughly to good LE values; clinically approved low molecular weight antibiotics have antibacterial efficiencies in the 0.26-0.32 mg/ml/atom range.

So for all you folks working on antibiotics, not only are fragments a viable starting point, you now have a new way to evaluate progress.