Category Archives: Small Molecules

RSC Fragments 2024

I attended RSC Fragments 2024 (Hinxton, 4–5 March 2024), a conference dedicated to fragment-based drug discovery. The various talks were really good, because they gave overviews of projects involving teams across long stretches of time. As a result there were no slides discussing wet lab protocol optimisations and not a single Western blot was seen. The focus was primarily either illustrating a discovery platform or recounting a declassified campaign. The latter were interesting, although I’d admit I wish there had been more talk of organic chemistry —there was not a single moan/gloat about a yield. This top-down focus was nice as topics kept overlapping, namely:

  • Target choice,
  • covalents,
  • molecular glues,
  • whether to escape Flatland,
  • thermodynamics, and
  • cryptic pockets
Continue reading

Taking Equivariance in deep learning for a spin?

I recently went to Sheh Zaidi‘s brilliant introduction to Equivariance and Spherical Harmonics and I thought it would be useful to cement my understanding of it with a practical example. In this blog post I’m going to start with serotonin in two coordinate frames, and build a small equivariant neural network that featurises it.

Continue reading

Finding and testing a reaction SMARTS pattern for any reaction

Have you ever needed to find a reaction SMARTS pattern for a certain reaction but don’t have it already written out? Do you have a reaction SMARTS pattern but need to test it on a set of reactants and products to make sure it transforms them correctly and doesn’t allow for odd reactants to work? I recently did and I spent some time developing functions that can:

  1. Generate a reaction SMARTS for a reaction given two reactants, a product, and a reaction name.
  2. Check the reaction SMARTS on a list of reactants and products that have the same reaction name.
Continue reading

The workings of Fragmenstein’s RDKit neighbour-aware minimisation

Fragmenstein is a Python module that combine hits or position a derivative following given templates by being very strict in obeying them. This is done by creating a “monster”, a compound that has the atomic positions of the templates, which then reanimated by very strict energy minimisation. This is done in two steps, first in RDKit with an extracted frozen neighbourhood and then in PyRosetta within a flexible protein. The mapping for both combinations and placements are complicated, but I will focus here on a particular step the minimisation, primarily in answer to an enquiry, namely how does the RDKit minimisation work.

Continue reading

Demystifying the thermodynamics of ligand binding

Chemoinformatics uses a curious jumble of terms from thermodynamics, wet-lab techniques and statistical terminology, which is at its most jarring, it could be argued, in machine learning. In some datasets one often sees pIC50, pEC50, pKi and pKD, in discussion sections a medchemist may talk casually of entropy, whereas in the world of molecular mechanics everything is internal energy. Herein I hope to address some common misconceptions and unify these concepts.

Continue reading

Conference feedback: AI in Chemistry 2023

Last month, a drift of OPIGlets attended the royal society of chemistry’s annual AI in chemistry conference. Co-organised by the group’s very own Garrett Morris and hosted in Churchill College, Cambridge, during a heatwave (!), the two days of conference featured aspects of artificial intelligence and deep machine learning methods to applications in chemistry. The programme included a mixture of keynote talks, panel discussion, oral presentations, flash presentations, posters and opportunities for open debate, networking and discussion amongst participants from academia and industry alike. 

Continue reading

A Simple Way to Quantify the Similarity Between Two Sets of Molecules

When designing machine learning algorithms with the aim of accelerating the discovery of novel and more effective therapeutics, we often care deeply about their ability to generalise to new regions of chemical space and accurately predict the properties of molecules that are structurally or functionally dissimilar to the ones we have already explored. To evaluate the performance of algorithms in such an out-of-distribution setting, it is essential that we are able to quantify the data shift that is induced by the train-test splits that we rely on to decide which model to deploy in production.

For our recent ICML 2023 paper Drug Discovery under Covariate Shift with Domain-Informed Prior Distributions over Functions, we chose to quantify the distributional similarity between two sets of molecules through the Maximum Mean Discrepancy (MMD).

Continue reading

9th Joint Sheffield Conference on Cheminformatics

Over the next few days, researchers from around the world will be gathering in Sheffield for the 9th Joint Sheffield Conference on Cheminformatics. As one of the organizers (wearing my Molecular Graphics and Modeling Society ‘hat’), I can say we have an exciting array of speakers and sessions:

  • De Novo Design
  • Open Science
  • Chemical Space
  • Physics-based Modelling
  • Machine Learning
  • Property Prediction
  • Virtual Screening
  • Case Studies
  • Molecular Representations

It has traditionally taken place every three years, but despite the global pandemic it is returning this year, once again in person in the excellent conference facilities at The Edge. You can download the full programme in iCal format, and here is the conference calendar:

Continue reading

Customising MCS mapping in RDKit

Finding the parts in common between two molecules appears to be a straightforward, but actually is a maze of layers. The task, maximum common substructure (MCS) searching, in RDKit is done by Chem.rdFMCS.FindMCS, which is highly customisable with lots of presets. What if one wanted to control in minute detail if a given atom X and is a match for atom Y? There is a way and this is how.

Continue reading