Sunday 2 August 2020

Why fragments?


Paramin panorama

Crystallographic fragment screens have been run recently against the main protease (at Diamond) and the Nsp3 macrodomain (at UCSF and Diamond) of SARS-Cov-2 and I thought that it might be of interest to take a closer look at why we screen fragments. Fragment-based lead discovery (FBLD) actually has origins in both crystallography [V1992 | A1996] and computational chemistry [M1991 | B1992 | E1994]. Measurement of affinity is important in fragment-to-lead work because it allows fragment-based structure-activity relationships to be established prior to structural elaboration. Affinity measurement is typically challenging when fragment binding has been detected using crystallography although affinity can be estimated by observation of the response of occupancy to concentration (the ∆G° value of −3.1 kcal/mol reported for binding of pyrazole to protein kinase B was derived in this manner).

Although fragment-based approaches to lead discovery are widely used, it is less clear why fragment-based lead discovery works as well as it appears to. While it has been stated that “fragment hits form high-quality interactions with the target”, the concept of interaction quality is not sufficiently well-defined to be useful in design. I ran a poll which asked about the strongest rationale for screening fragments.  The 65 votes were distributed as follows: ‘high ligand efficiency’ (23.1%), ‘enthalpy-driven binding’ (16.9%), ‘low molecular complexity’ (26.2%) and ‘God loves fragments’ (33.8%). I did not vote.

The belief is that fragments are especially ligand-efficient has many adherents in the drug discovery field and it has been asserted that “fragment hits typically possess high ‘ligand efficiency’ (binding affinity per heavy atom) and so are highly suitable for optimization into clinical candidates with good drug-like properties”. The fundamental problem with ligand efficiency (LE), as conventionally calculated, is that perception of efficiency varies with the arbitrary concentration unit in which affinity is expressed (have you ever wondered why Kd , Ki or IC50 has to be expressed in mole/litre for calculation of LE?). This would appear to be an rather undesirable characteristic for a design metric and LE evangelists might consider trying to explain why it’s not a problem rather than dismissing it as a “limitation” of the metric or trying to shift the burden of proof is onto the skeptics to show that the evangelists’ choice of concentration unit for calculation of LE is not useful.

The problems associated with the arbitrary nature of the concentration unit used to express affinity were first identified in 2009 and further discussed in 2014 and 2019. Specifically, it was noted that LE has a nontrivial dependency on the concentration,  C°, used to define the standard state. If you want to do solution thermodynamics with concentrations defined then you do need to specify a standard concentration. However, it is important to remember that the choice of standard concentration is necessarily arbitrary if the thermodynamic analysis is to be valid. If your conclusions change when you use a different definition of the standard state then you’ll no longer be doing thermodynamics and, as Pauli might have observed, you’ll not even be wrong. You probably don't know it, but when you use the LE metric, you’re making the sweeping assumption that all values of Kd, Ki and IC50 tend to a value of 1 M in the limit of zero molecular size. Recalling the conventional criticism of homeopathy, is there really a difference between a solute that is infinitely small and a solute that is infinitely dilute?

I think that’s enough flogging of inanimate equines for one blog post so let’s take a look at enthalpy-driven binding. My view of thermodynamic signature characterization in drug discovery is that it’s, in essence, a solution that’s desperately seeking a problem. In particular, there does not appear to be any physical basis for claims that the thermodynamic signature is a measure of interaction quality.  In case you’re thinking that I’m an unrepentant Luddite, I will concede that thermodynamic signatures could prove useful for validating physics-based models of molecular recognition and in, in specific cases, they may point to differences in binding mode within congeneric series. I should also stress that the modern isothermal calorimeter is an engineering marvel and I'd always want this option for label-free, affinity measurement in any project.

It is common to see statements in the thermodynamic signature literature to the effect that binding is ‘enthalpy-driven’ or ‘entropy-driven’ although it was noted in 2009 (coincidentally, in the same article that highlighted the nontrivial dependence of LE on C°) that these terms are not particularly meaningful. The problems start when you make comparisons between the numerical values of ∆H (which is independent of C°) and T∆S° (which depends on C°). If I’d presented such a comparison in physics class at high school (I was taught by the Holy Ghost Fathers in Port of Spain), I would have been caned with a ferocity reserved for those who’d dozed off in catechism class.  I’ll point you toward an article which asserts that, “when compared with many traditional druglike compounds, fragments bind more enthalpically to their protein targets”. I have a number of issues with this article although this is not the place for a comprehensive review (although I’ll probably pick it up in ‘The Nature of Lipophilic Efficiency’ when that gets written).

While I don’t believe that the authors have actually demonstrated that fragments bind more enthalpically than ligands of greater molecular size, I wouldn’t be surprised to discover that gains in affinity over the course of a fragment-to-lead (F2L) campaign had come more from entropy than enthalpy. First, the lost translation entropy (the component of ∆S° that endows it with its dependence on C°) is shared over greater number of intermolecular contacts for structurally-elaborated compounds and this article is relevant to the discussion. Second, I’d expect the entropy of any water molecule to increase when it is moved to bulk solvent from contact with molecular surface of ligand or target (regardless of polarity of the molecular surface at the point of contact). Nevertheless, this is something that you can test easily by examining the response of (∆H + T∆S°) to ∆G° (best to not to aggregate data for different targets and/or temperatures when analyzing isothermal titration calorimetry data in this manner). But even if F2L affinity gains were shown generally to come more from entropy than enthalpy, would that be a strong rationale for screening fragments?

This gets us onto molecular complexity and this article by Mike Hann and GSK colleagues should be considered essential reading for anybody thinking about selecting of compounds for screening. The Hann model is a conceptual framework for molecular complexity but it doesn’t provide much practical guidance as to how to measure complexity (this is not a criticism since the thought process should be more about frameworks and less about metrics). I don’t believe that it will prove possible to quantify molecular complexity in an objective manner that is useful for designing compound libraries (I will be delighted to be proven wrong on this point). The approach to handling molecular complexity that I’ve used in screening library design is to restrict extent of substitution (and other substructural features that can be considered to be associated with molecular complexity) and this is closer to ‘needle screening’ as described by Roche scientists in 2000 than to the Hann model.

Had I voted in the poll, ‘low molecular complexity’ would have got my vote.  Here’s what I said in NoLE (it’s got an entire section on fragment-based design and a practical suggestion for redefining ligand efficiency so that perception does not change with C°):

"I would argue that the rationale for screening fragments against targets of interest is actually based on two conjectures. First, chemical space can be covered most effectively by fragments because compounds of low molecular complexity [18, 21, 22] allow TIP [target interaction potential] to be explored [70,71,72,73,74] more efficiently and accurately. Second, a fragment that has been observed to bind to a target may be a better starting point for design than a higher affinity ligand whose greater molecular complexity prevents it from presenting molecular recognition elements to the target in an optimal manner."

To be fair, those who advocate the use of LE and thermodynamic signatures in fragment-based design do not deny the importance of molecular complexity. Let’s assume for the sake of argument that interaction quality can actually be defined and is quantified by the LE value and/or the thermodynamic signature for binding of compound to target. While these are massive assumptions, LE values and thermodynamic signatures are still effects rather than causes.

The last option for poll was ‘God loves fragments’ and more respondents (33.8%) voted for this than any of the first three options. I would interpret a vote for ‘God loves fragments’ in three ways. First, the respondent doesn’t consider any one of the first three options to be a stronger rationale for screening fragments than the other two. Second, the respondent doesn’t consider any of the first three options to be a valid rationale for screening fragments. Third, the respondent considers fragment-based approaches to have been over-sold.

This is a good place to wrap up. While I remain an enthusiast for fragment-based approaches to lead discovery, I do also believe that they have been somewhat oversold. The sensitivity of LE evangelists to criticism of their metric may stem from the use of LE to sell fragment-based methods to venture capitalists and, internally, to skeptical management. A shared (and serious) deficiency in the conventional ways in which LE and thermodynamic signature are quantified is that perception changes when the arbitrary concentration,  C°, that defines the standard state is changed. While there are ways in which this deficiency can be addressed for analysis, it is important that the deficiency be acknowledged if we are to move forward. Drug design is difficult and if we, as drug designers, embrace shaky science and flawed data analysis then those who fund our activities may conclude that the difficulties that we face are of our own making.     

4 comments:

Editor Chemrj said...

Very Nice Information and Very good blog on Molecular design.

Editor
Chemistry Research Journal

Dan Erlanson said...

Hi Pete,

I confess I didn't see the poll while it was running, but in my opinion there are at least two other good answers to the question "why fragments." The first, which is related but not identical to molecular complexity, is greater coverage of chemical space; this has been an argument for fragments from the field's earliest days. The second is that FBDD has been effective at putting dozens of drugs into the clinic, even for historically difficult targets. Nothing like success to popularize a technique!

Dan Erlanson said...

Hi Pete,

I confess I didn't see the poll while it was running, but in my opinion there are at least two other good answers to the question "why fragments." The first, which is related but not identical to molecular complexity, is greater coverage of chemical space; this has been an argument for fragments from the field's earliest days. The second is that FBDD has been effective at putting dozens of drugs into the clinic, even for historically difficult targets. Nothing like success to popularize a technique!

Peter Kenny said...

Hi Dan,

Apologies for my tardiness in approving your comments (I’ve retained the duplicates because to delete one might get it flagged as spam). The objective of the poll was very much to ask why fragment-based methods work rather than to ask whether fragment-based methods work. My view is that coverage is a much more important consideration than diversity in screening library design and that structurally-prototypical compounds of low molecular complexity allow chemical space to be covered most effectively. I agree that low molecular weight compounds are typically of low molecular complexity. Nevertheless, some compounds that are fragments according to Ro3 are too adorned with features to be of practical value in screening (I would make this criticism of some of the ‘3D fragments’ that I’ve seen in the literature and in Practical Fragments posts).