Machine Learning for Protein Engineering – Part 1 banner

The two-part Machine Learning for Protein Engineering conference at PEGS Europe explores the informatics technologies and strategies driving the improvement of quality, precision and developability of biotherapeutics via the application of artificial intelligence and machine learning. The 2023 meeting will focus on discovery and optimization campaigns via a critical examination of use cases, experimental validation procedures, integrations with legacy discovery platforms and data creation/curation. These programs will offer the PEGS Europe community a forum in which to debate successes to date in adopting this nascent family of technologies – and how research organizations can realize the potential in their significant investments in this space. This conference will demonstrate the considerable value of these techniques and emphasize the practical applications over the purely theoretical exercises. The format will be educational and interactive.

Recommended Short Course*
Monday, 13 November, 14:00 – 17:00
SC1: Machine Learning Tools for Protein Engineering
*Separate registration required. See short courses page for details. All short courses take place in-person only.

Wednesday, 15 November

Registration Open and Morning Coffee07:30

DE NOVO DESIGN USE CASES

08:25

Chairperson’s Opening Remarks

Tushar Jain, PhD, Principal Scientist, Computational Biology, Adimab LLC

08:30

Lab-in-the-Loop, an ML-Driven Platform for Automated Molecular Discovery and Design

Nathan Frey, PhD, Senior Machine Learning Scientist, Prescient Design, a Genentech Company

We will discuss the “Lab-in-the-loop” system, a collaboration between Prescient Design and Antibody Engineering at Genentech, to build and integrate state-of-the-art machine learning methods with large molecule design and discovery capabilities. Lab-in-the-loop encompasses generative models, pseudo-oracles, physics-based modeling, large language models, wet-lab assays, and active learning to fundamentally change early-stage drug discovery.

09:00

Generation and Experimental Validation of Novel de novo Abs with Unique Functionalities

Yanay Ofran, PhD, Founder, CEO, Biolojic Design Ltd.

Most therapeutic antibodies are simple antagonists. However, like all proteins, antibodies can be sophisticated nano-machines. Biolojic Design uses AI to program antibodies to become dynamic functional switches affecting biology in new ways. I will describe our AI-design process, and share clinical data from the first AI-designed therapeutic antibody. I will also show preclinical data on multi-specific antibodies illustrating their potential to improve outcome in cancer and autoimmune diseases.

09:30 Selecting Optimal Antibodies for IND-Enabling Studies with an Integrated High-Throughput & Lead Assessment Platform

Lucas Kraft, Senior Research Scientist, Translational, AbCellera

Session Break to Transition into Plenary Keynote10:00

PLENARY KEYNOTE SESSION

10:10

Plenary Keynote Introduction

Enkelejda Miho, PhD, Professor, University of Applied Sciences and Arts Northwestern Switzerland, and Managing Director, aiNET

10:15

Benchmarking the Impact of AI Biologics Discovery and Optimisation for Pharma

Rebecca Croasdale-Wood, PhD, Director, Augmented Biologics Discovery & Design, Biologics Engineering, Oncology, AstraZeneca

The biologics landscape is rapidly changing with the number of AI-enabled biologics in pre-clinical and clinical stages estimated to be 50-60 (1). This change is driven by the increase in enterprise software solutions to capture and store data, augmented discovery workflows, improvements in machine learning technology, and advances in computing power. Augmented biologics discovery has the potential to revolutionize biologics discovery, yet information of how in silico technologies perform, versus traditional discovery platforms is scarce. At PEGS Europe, we will present current in silico biologics design and optimisation technologies, with a focus on our internal efforts to benchmark the impact of combining novel in silico technologies with our existing biologics discovery platforms.

10:45

Keynote Chat 

Rebecca Croasdale-Wood, PhD, Director, Augmented Biologics Discovery & Design, Biologics Engineering, Oncology, AstraZeneca

Interviewed By:

Enkelejda Miho, PhD, Professor, University of Applied Sciences and Arts Northwestern Switzerland, and Managing Director, aiNET

Coffee Break in the Exhibit Hall with Poster Viewing11:00

11:45

Accelerating the Discovery Pipeline with ML: From Library Design to Discovery and Optimization, and Early Developability Screening

Tushar Jain, PhD, Principal Scientist, Computational Biology, Adimab LLC

Incorporating predictive modeling into experimental workflows holds great promise for accelerating discovery, guiding optimization, and prioritizing leads. Here we discuss application of ML to design of synthetic libraries for discovery, hybrid experimental-modeling approaches for selection of functionally diverse antibodies, and developability predictions that decrease resource-intensive experiments. Our integration of ML into a larger informatics/data platform enables predictions to inform candidate selection throughout the discovery and lead optimization process.

12:15

KEYNOTE PRESENTATION: Antibody Structure and Dynamics in Solution

Klaus R. Liedl, PhD, Professor, Head, General, Inorganic, & Theoretical Chemistry, University of Innsbruck

Antibodies are highly flexible molecules, due to the hinge regions, the elbow linkers, the interdomain interfaces between Ig-fold domain pairs and the loops in the paratope. We demonstrated that the binding competent structure is normally the dominant structure in solution, even though it is often not the structure found for unbound antibodies. The resulting opportunities and challenges for AI-driven antibody structure prediction are discussed in the light of these findings.

12:45 Innovative Antibody Discovery Workflow Leveraging Artificial Intelligence to Prioritize Leads

Crystal Richardson, Ph.D, Business Partnership Manager, Azenta Life Sciences

Azenta now offers an innovative end-to-end antibody screening solution that guides your discovery program to more diverse leads while reducing liabilities for antibody development. Utilizing next generation sequencing of your in-vivo samples (i.E. B-cells, pbmcs) or in-vitro libraries (i.E. Phage display), a bioinformatics platform, and gene synthesis, antibodies are produced with promising biophysical profiles for commercialization. 

Session Break13:15

13:20 LUNCHEON PRESENTATION:Smart People Solve Problems. AI Geniuses Avoid Them

Patrick Doonan, Ph.D., Director of Antibody Engineering, Antibody Discovery, XtalPi

XtalPi’s comprehensive end-to-end antibody discovery pipeline utilizes an array of unbiased AI checkpoints across every phase of the process.  Generative AI and in silico prescreening reduces the burden of wet-lab experimentation thereby expediting the time to clinic.  Developability concerns are minimized early in the discovery process to avoid problematic leads.  Our AI tools are used to further improve quality hits resulting in highly developable antibodies with exceptional therapeutic potential.

Session Break14:20

IMPLEMENTATION CHALLENGES AND SOLUTIONS

14:30

Chairperson’s Remarks

Jeffrey Ruffolo, PhD, Machine Learning Scientist, Profluent Bio

14:35

Developing Internal AI Capabilities via External Collaborations and Internal Resources

Hubert Kettenberger, PhD, Head, Computational Protein Engineering, Roche

AI applications have become increasingly powerful, and play an increasing role in today's research and development. At the same time, there is no consensus yet regarding AI methodologies, and how to best integrate them in the discovery and development process. Building internal capabilities and establishing external collaborations can help navigate through this exciting new augmentation of biologics drug development.

EMERGING MODELS AND PLATFORMS

15:05

Implementation of CamSol Using Machine Learning

Marc Oeller, PhD, Postdoctoral Fellow, Mann Group, Max Planck Institute of Biochemistry

In 2015, we introduced the CamSol method, which enables accurate prediction of protein solubility solely by analysing the physico-chemical properties of their sequences. We recently extended the method to enable accurate prediction of non-natural amino acids such as chemically or posttranslationally modified ones. In this talk, I will highlight how CamSol can be used in drug development pipelines and explore the possibilities to extend CamSol using machine learning.

15:35 AI Driven de novo Antibody Discovery

Satoshi Tamaki, PhD, CSO, MOLCURE Inc.

Considering the challenges of antibody development, MOLCURE has designed a platform that integrates AI, robotics, and molecular biology experiments. Our platform has generated >1 billion data points to train AI models. We would like to introduce the showcase of the discovery of pM-order affinity VHH antibody from a single phage display experiment and discuss the future that generative AI opens up.

 

 

Refreshment Break in the Exhibit Hall with Poster Viewing16:05

17:00

FEATURED PRESENTATION: Generative Antibody Modelling

Charlotte M. Deane, PhD, Professor, Structural Bioinformatics, Statistics, University of Oxford; Chief Scientist, Biologics AI, Exscientia

Here we show that by optimising an inverse folding model specifically for antibody structures, we are able to outperform generic protein models on sequence recovery and structure robustness, with notable improvement on the hypervariable CDR-H3 loop.We also demonstrate the applications of our model to drug-discovery and binder design and evaluate the quality of proposed sequences.

17:30

Integration of Machine Learning, Structural Biology, and Wet Lab Data to Augment Drug Discovery for Autoimmune Diseases

Nathan Higginson-Scott, PhD, CTO, Seismic Therapeutic

We will discuss how Seismic Therapeutic is using its IMPACT platform to integrate machine learning, structural biology, protein engineering and translational Immunology to accelerate the discovery and development of therapeutics for autoimmune diseases, caused by a dysregulated adaptive immune system.

18:00

Machine Learning for Biomolecule Engineering

Anna Puszkarska, PhD, Senior Machine Learning Scientist, Biologics Engineering, AstraZeneca

Engineering new biological molecules with desired activity profiles requires time consuming and expensive cycles of design-make-test-analyse (DMTA) work. Even for short protein sequences, the available exploration space is intractable for traditional methods of experimental biology. In this talk, I will discuss how machine learning can be used to augment the design of peptide therapeutics. Specifically, I will present our recent studies focussed on de novo design and optimisation of GPCR ligands.

18:30

Generative Modeling for Functional Protein Design

Jeffrey Ruffolo, PhD, Machine Learning Scientist, Profluent Bio

Generative language models trained on protein sequences have proven incredibly powerful for protein sequence design. In this talk, we will demonstrate how protein language models enable discovery of diverse proteins, which often function on par with natural counterparts- despite significant deviation in sequence space. Beyond generation of sequences, protein language models are effective zero-shot predictors of fitness, enabling direct optimization of function.

Close of Machine Learning for Protein Engineering – Part 1 Conference19:00