Optimisation and Developability track banner

The 12th Annual Optimisation & Developability conference at PEGS Europe reviews innovative approaches, methods and models that scientists use to develop strategies for candidate selection, lead optimisation, liability mitigation and developability prediction, for existing molecules as well as novel modalities. This conference will also explore the applications of artificial intelligence and machine learning (AIML) and in silico approaches to predict stability and optimise biophysical properties.

Pre-Conference Virtual Short Course
14:00 Recommended Pre-Conference Virtual Short Course*
SC1: Developability of Bispecific Antibodies: Formats and Applications

*Separate registration required. See conference website short course page for details.

Tuesday, 2 November

07:00 Registration and Morning Coffee

STRATEGIES TO OPTIMISE BIOPHYSICAL PROPERTIES AND PREDICT DEVELOPABILITY OF BIOTHERAPEUTICS

08:25

Chairperson's Opening Remarks

Pietro Sormanni, PhD, Group Leader, Royal Society University Research Fellow, Chemistry of Health, Yusuf Hamied Department of Chemistry, University of Cambridge
08:30

To Sub 10 pM Affinity and Beyond! Discovery of a Next-Generation Highly Potent Long-Acting Anti IL-5 Antibody

Martin A. Orecchia, PhD, Team Leader, Antibody Discovery, GlaxoSmithKline

In the case study presented here, a therapeutic grade highly potent anti-IL-5 antibody was significantly improved through combining a yeast-based antibody selection platform with a highly efficient affinity maturation method, using structure based as well as unbiased CDR targeted diversification. CDR-targeted mutagenesis libraries were constructed using the Adimab Yeast Platform and antibodies with improved affinity for IL-5 were selected by fluorescence activated cell sorting. This combined approach enabled the identification of sub-pM affinity range antibodies using a single rapid maturation cycle.

  • NEW DATA - This Presentation Contains New Data
  • NEW TALK - This Presentation Will be Given for the First Time
09:00

Validation of huFcRn Transgenic Mouse Model to Screen Novel Fc-Engineered Monoclonal and Multi-Specific Antibodies

Delphine Valente, PhD, Head, Pharmacokinetics, Modeling and Simulations, Sanofi

First PK profiling of multispecific antibodies with various backbones and Fc mutations in huFcRn transgenic mice compared to Non-Human Primates. Tg32 mouse model can be used to distinguish PK differences between antibodies containing Fc-backbone mutations in the same range of order as observed in NHP. This model may serve as a convenient, cost effective surrogate for in vivo prediction of antibodies t1/2z, Vss and Clearance facilitating the screening strategy to select antibodies with the most favorable PK. By extension, a first estimation of PK parameters in human can be directly deduced from this mouse model.

09:30

Comparison of CH1-CL and CH3-CH3 Interfaces – Lessons for the Design of Bispecific Antibodies

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

Typically, immunoglobulin G antibodies depend on homodimerization of the fragment crystallizable regions of two identical heavy chains. By modifying the CH3-CH3 interface, with different mutations on each domain, the engineered Fc fragments form rather heterodimers than homodimers. Here, we use classical molecular dynamics simulations to identify key interactions in the CH3-CH3 and CH1-CL interfaces, that contribute to their stability and tendency to heterodimerize. Additionally, we compare CH3-CH3 domains, to the structurally similar CH1-CL interfaces in antigen-binding fragments (Fabs). The CH1-CL domain shares a very similar fold and interdomain orientation with the CH3-CH3 dimer. Thus, numerous well established optimisation efforts for CH3-CH3 interfaces, have also been applied to CH1-CL dimers to reduce the number of mispairings in the Fabs. By comparing all CH3-CH3 and CH1-CL dimers with each other we find similar interaction patterns. However, we also observe pairing specific interface interactions and show that also different combinations of heavy chains with k and l-light chains result in characteristic interface contacts. Additionally, we present exemplary dissociation mechanisms for both CH3-CH3 and CH1-CL dimers and identify key interactions that contribute to stability and their tendency to heterodimerize. Thus, this study has broad implications for improving the yield of bispecifics as it provides a structural and mechanistical understanding of CH3-CH3 and CH1-CL interfaces and thereby presents a crucial aspect for the development and optimisation of bispecific antibodies.

10:00 Coffee Break in the Exhibit Hall with Poster Viewing
10:45

Application of Molecular Dynamics Simulations for Developability Assessment and Formulation Development of Biologics

Carolin Berner, Chair of Pharmaceutical Technology & Biopharmaceutics, Pharmaceutics, University of Munich

Computational methods can accelerate the selection of therapeutic protein candidates with suitable biophysical properties and guide formulation development. Here, we will present approaches for the selection of aggregation-resistant proteins and the identification of new stabilizing excipients in silico. Furthermore, we will show how molecular dynamics simulations can provide mechanistic explanations of buffer effects and stabilizing protein-excipient interactions. We will conclude by discussing the opportunities and limitations of the presented approaches.

  • NEW TALK - This Presentation Will be Given for the First Time
11:15

Combining Random Mutagenesis, Structure-Guided Design, Next-Generation Sequencing and in silico Prediction to Optimize Polyreactivity and Other Biophysical Properties in Therapeutic Antibodies

James R. Apgar, PhD, Associate Research Fellow, BioMedicine Design, Pfizer Inc.

We will discuss the challenges in developing a neutralizing anti-IL-21R antibody, that can effectively compete with IL-21 for its highly negatively-charged paratope, while maintaining favorable biophysical properties. To overcome the limitations in charge-based in vitro deselection methods, we utilized a combination of structure-guided rational library design, next-generation sequencing of library outputs and machine learning methods to identify a high-affinity antibody with desirable stability and biophysical properties. We have expanded upon these methods to develop additional predictive tools to improve the biophysical properties of antibodies.

Andrew Bradbury, MB BS, PhD, Chief Scientific Officer, Specifica

The Specifica Generation 3 Library Platform is based on highly developable clinical scaffolds, into which natural CDRs purged of sequence liabilities have been embedded. The platform uses phage+yeast display to directly yield highly diverse (100-1000 clusters differing by Levenshtein distance 30-40), high affinity (20% subnanomolar), extremely developable (>80% lack biophysical liabilities), drug-like antibodies, which in a recent Covid campaign were as potent as antibodies from immune sources.

12:15 Session Break
Ariel Gilert, PhD, Manager, Research and Development, R&D, Lonza AG

An efficient solution for the correct assembly of IgG-like bispecific antibodies is a sought-after component for the manufacturing of a growing number of therapeutic antibodies entering the clinic.

Using a panel of bispecific antibodies we will present a  comprehensive evaluation of the “Lonza bispecific platform”, showing that this technology provides an excellent, easy-to implement solution for the manufacturing of bispecific IgG-like antibodies composed of four different chains.

12:55 Session Break

USING AIML TO ACCELERATE DISCOVERY AND DEVELOPABILITY

13:45

Chairperson's Remarks

Klaus R. Liedl, PhD, Professor & Head, General, Inorganic & Theoretical Chemistry, University of Innsbruck
13:50 KEYNOTE PRESENTATION:

Third-Generation Methods of Antibody Discovery and Optimisation: In silico Rational Design

Pietro Sormanni, PhD, Group Leader, Royal Society University Research Fellow, Chemistry of Health, Yusuf Hamied Department of Chemistry, University of Cambridge

De novo design methods promise a cheaper and faster route to antibody discovery, while enabling the targeting of predetermined epitopes and, at least in principle, the screening of multiple biophysical properties. I will present some recent advances in this area, including a data-driven strategy to design antibodies targeting structured epitopes and rapid and accurate solubility predictions. Our experimental validations demonstrate that in silico approaches are becoming increasingly competitive, and can be applied in synergy with established laboratory-based procedures to streamline antibody development.

  • NEW DATA - This Presentation Contains New Data
14:20

Computational Prediction of Non-Specific Interactions of Antibodies

Michele Vendruscolo, PhD, Professor, Chemistry, University of Cambridge

There is an increasing interest in developing computational methods to predict the developability of antibodies in order to reduce the cost of antibody discovery programmes as well as late stage failures. Among the major causes of attrition in these programmes, poor specificity stands out, in part because it is challenging to assess it experimentally in a high-throughput manner. The availability of large databases of antibody sequences and data about antibody specificity is making it possible to develop computational methods to predict the specificity of antibodies from their amino acid sequences. I will present a novel method to achieve this goal.

14:50

Predicting Antibody Developability through Experimental and in silico Approaches

Marc Bailly, PhD, Principal Scientist, Merck Research Labs

The current race to develop better drugs faster has led biopharmaceutical companies into optimizing all drug discovery and development processes. As part of this effort, machine learning algorithms are being developed to identify correlations between amino acid sequences and physicochemical properties observed during drug development. Here, we describe our ongoing efforts aiming at benchmarking such machine learning algorithms to facilitate our drug discovery process. The audience will learn about Merck MSD current strategy in order to enable in silico approaches for developability assessments. They will learn about useful experimental assays endpoints for in silico approaches, data collection, curation and usage as well as the new platform Merck MSD is currently developing to serve as the interface bewteen the Scientists and machine learning algorythms for in silico developability assessments.

  • NEW DATA - This Presentation Contains New Data
  • NEW TALK - This Presentation Will be Given for the First Time
Raymond Miller, Senior Global Product Manager, GenScript Biotech Corp.

GenScript precision mutant libraries by electrochemical semiconductor DNA synthesis technology allow user-defined codon with precise ratios, saving valuable time and effort during the screening and characterization process, speeding up the engineering workflow, and reducing the overall cost of downstream expenses. We showcase a case study of our precision mutant libraries in affinity maturation of a monoclonal antibody with a  ~10,000 fold improved affinity via site saturation and combinatorial mutant library.

15:50 Refreshment Break in the Exhibit Hall with Poster Viewing
16:35

Deep Neural Networks in Protein Engineering – From Predicting Stabilizing Sequences to Refining 3D Structures

Abhishek Mukhopadhyay, PhD, Principal Scientist and Group Lead, Zymeworks, Inc.

Advances in artificial intelligence have demonstrated its ability to drive and accelerate new scientific discoveries. We show that the complex sequence structure relation can be learnt efficiently using deep neural networks trained on crystallized protein structures. We attempt addressing two protein modelling problems here: protein side-chain geometry prediction and predicting stabilizing protein sequences. These models achieve improved efficiency when compared against the state-of-the-art protein packing and protein design tools respectively.

  • NEW TALK - This Presentation will be given for the First Time
17:05

Intrinsic Physiochemical Profile of Biological Medicines

Sandeep Kumar, PhD, Senior Research Fellow, Computational Biochemistry and Bioinformatics, Boehringer Ingelheim Pharmaceuticals

We recently collected amino acid sequences of currently marketed antibody-based biologic medicines and used these to derive their homology-based structural models. Availability of both sequence and structural models of these biotherapeutics afforded us an opportunity to analyze their physicochemical attributes from a perspective of developability. These analyses have resulted in a profile that can be used to estimate ‘medicine-likeness’ of the biologic drug candidates currently in discovery and development.

Oren Beske, Dr, Amalgamator of Business and Biology, ATUM

Since the Leap-In Transposase platform was launched for biotherapeutic cell line development three years ago, the market adoption has been robust, and it has rapidly become well accepted in the industry.  This talk will highlight key milestones from these past few years and will focus on case studies around our COVID response, chain ratio balancing for complex molecules and how the technology can be used to reduce target gene expression.

18:05 Welcome Reception in the Exhibit Hall with Poster Viewing

Explore new products and services in our Exhibit Hall, engage with poster presenters, schedule 1-on-1 meetings, and build your research community during this open networking period.

19:05 Close of Optimisation & Developability