Category Archives: Antibodies

Conference Summary: MGMS Adaptive Immune Receptors Meeting 2024

On 5th April 2024, over 60 researchers braved the train strikes and gusty weather to gather at Lady Margaret Hall in Oxford and engage in a day full of scientific talks, posters and discussions on the topic of adaptive immune receptor (AIR) analysis!

Continue reading

The Antibody Dictionary

Similar to getting lost in a language when moving country, you might encounter a language barrier when moving research fields. This dictionary will guide you in the complex world of immunoinformatics, with a focus on antibodies. Whether your main research will be in this field, you want to apply your machine learning model on antibodies, or you just want to understand the research performed in OPIG, this dictionary will get you started.

The Antibody Dictionary:

Affinity maturation: The optimisation process of naive antibodies to memory antibodies such that the antibody is optimised for a specific antigen. 

Antibody: (immunoglobulin) a Y-shaped molecule important in the adaptive immune system. A canonical antibody consists of two identical heavy chains and two identical smaller light chains. 

Continue reading

Let your library design blosum

During the lead optimisation stage of the drug discovery pipeline, we might wish to make mutations to an initially identified binding antibody to improve properties such as developability, immunogenicity, and affinity.

There are many ways we could go about suggesting these mutations including using Large Language Models e.g. ESM and AbLang, or Inverse Folding methods e.g. ProteinMPNN and AntiFold. However, some of our recent work (soon to be pre-printed) has shown that classical non-Machine Learning approaches, such as BLOSUM, could also be worth considering at this stage.

Continue reading

What can you do with the OPIG Immunoinformatics Suite? v3.0

OPIG’s growing immunoinformatics team continues to develop and openly distribute a wide variety of databases and software packages for antibody/nanobody/T-cell receptor analysis. Below is a summary of all the latest updates (follows on from v1.0 and v2.0).

Continue reading

Exploring the Observed Antibody Space (OAS)

The Observed Antibody Space (OAS) [1,2] is an amazing resource for investigating observed antibodies or as a resource for training antibody specific models, however; its size (over 2.4 billion unpaired and 1.5 million paired antibody sequences as of June 2023) can make it painful to work with. Additionally, OAS is extremely information rich, having nearly 100 columns for each antibody heavy or light chain, further complicating how to handle the data. 

From spending a lot of time working with OAS, I wanted to share a few tricks and insights, which I hope will reduce the pain and increase the joy of working with OAS!

Continue reading

Experience at a Keystone Symposium

From 19th-22nd February I was fortunate enough to participate in the joint Keystone Symposium on Next-Generation Antibody Therapeutics and Multispecific Immune Cell Engagers, held in Banff, Canada. Now in their 51st year, the Keystone Symposia are a comprehensive programme of scientific conferences spanning the full range of topics relating to human health, from studies on fundamental bodily processes through to drug discovery.

Continue reading

SUMO wrestling with developability

When engineering antibodies into effective biotherapeutics, ideally, factors such as affinity, specificity, chemical stability and solubility should all be optimised. In practice, we know that it’s often not feasible to co-optimise all of these, and so compromises are made, but identifying these developability issues early on in the antibody drug discovery process could save costs and reduce attrition rates. For example, we could avoid choosing a candidate that expresses poorly, which would make it expensive to manufacture as a drug, or one with a high risk of aggregation that would drive unwanted immunogenicity.

On this theme, I was interested to read recently a paper by the Computational Chemistry & Biologics group at Merck (Evers et al., 2022 https://www.biorxiv.org/content/10.1101/2022.11.19.517175v1). They have developed a pipeline called SUMO (In Silico Sequence Assessment Using Multiple Optimization Parameters), that brings together publicly-available software for in silico developability assessment and creates an overall developability profile as a starting point for antibody or VHH optimisation.

Read more: SUMO wrestling with developability

For each sequence assessed, they report factors such as sequence liabilities (residues liable to chemical modifications that can alter properties such as binding affinity or aggregation propensity), surface hydrophobicity, sequence identity compared to most similar human germline and predicted immunogenicity (based on MHC-II binding). Also provided are an annotated sequence viewer and 3D visualisation of calculated properties. Profiles are annotated with a red-yellow-green colour-coding system to indicate which sequences have favourable properties.

Overall, this approach is a useful way to discriminate between candidates and steer away from those with major developability issues prior to the optimisation stage. Given that the thresholds for their colour-coding system are based on data from marketed therapeutic antibodies, and that the software used has primarily been designed for use on antibody datasets, I would be interested to see whether the particular descriptors chosen for SUMO translate well to VHHs, or whether there are other properties that are stronger indicators of nanobody developability.