22 March 2009

Fragments in silico, confirmed by X-ray

I’ve always been something of an empiricist, and have therefore been wary of computational fragment screening. It’s not that I think it’s impossible, just that the algorithms and parameters developed to date have not often shown themselves up to the task. A paper just published in Nature Chemical Biology from Brian Shoichet’s group at UCSF has caused me to reconsider my skepticism.

Shoichet and Yu Chen used the program DOCK to screen 67,489 commercially available fragment-sized molecules contained in the database ZINC against the active site of the beta lactamase CTX-M, a bacterial enzyme responsible for resistance to penicillin and cephalosporin. Of 69 top hits, 10 actually inhibited the enzyme when tested experimentally. In contrast, of 37 high-scoring hits from a similar computational screen of 1,147,326 larger lead-like molecules, none showed any inhibition up to the limit of their solubilities.

Interestingly, each of the ten active fragments contained an anionic group: 3 carboxylates, 2 sulfates, and 5 tetrazoles among the set. A reexamination of the docked lead-like molecules revealed a relatively high-scoring tetrazole, which exhibited an experimental Ki value of 21 micromolar (see figure). Although this was an in silico hit, it was swamped by the number of (inactive) hits and so had not been selected for experimental follow-up until the fragment results revealed tetrazoles to be privileged pharmacophores. Additional similarity searching of the lead-like molecules led to two additional low micromolar inhibitors.



Five of the inhibitory fragments and one of the lead-like molecules were characterized crystallographically, and the results were remarkable: all of them bound in a similar manner to that predicted by docking.

Chen and Shoichet also investigated the specificity of the fragments compared to the lead-like compounds, and the results agreed well with those predicted by Hann and colleagues (as discussed on our sister blog FBDD-Lit here). Namely, while the fragments had relatively low specificity against a mechanistically distinct beta lactamase (AmpC), the lead-like molecule exhibited roughly 100-fold tighter inhibition of CTX-M. In other words, fragments likely have a higher hit rate (and correspondingly lower specificity) due in part to their simplicity, but as fragments are elaborated, specificity can be readily built into the molecules.

So does this mean the era of computational fragment-based screening has arrived? While these results are impressive, it is important to keep them in perspective. CTX-M has a relatively rigid active site, while many proteins of interest show a level of flexibility that confounds modeling. Moreover, Chen and Shoichet were working with an ultra-high resolution (0.88-Angstrom) crystal structure of CTX-M in which they could actually see density for hydrogen atoms on some polar groups. Needless to say, this is atypical. Still, the paper does give hope that the computational tools are ready, as long as they are applied to appropriate systems.

No comments: