Emerging Targets and Therapeutic Approaches banner

Developing technologies, increased understanding and the proliferation of engineering approaches has accelerated the pace of exploring novel targets against cancer, autoimmune, inflammatory disease, and a range of other indications. The Emerging Targets and Therapeutic Approaches Conference will highlight success with new approaches, improved targeting and innovative solutions to address challenges that remain. Join us this November in Lisbon for a dynamic look at the incredible progress and myriad of solutions that are being investigated to create first-in-class biotherapeutics.

Wednesday, 15 November

Registration Open and Morning Coffee07:30

INNOVATIVE APPROACHES TO THE CHALLENGES OF SOLID TUMOURS

08:25

Chairperson's Opening Remarks

Kerry Chester, PhD, Professor, Molecular Medicine, University College London

08:30

Tumour Specific Immunogene Therapy to Deliver Protein Therapeutics to Solid Tumour Microenvironments

Samantha Bailey-Bucktrout, PhD, Senior Vice President, Akamis Bio

The tumour-specific immunogene platform (T-SIGn) has been designed to deliver protein therapeutics selectively to tumor microenvironments following systemic dosing and is moving into Phase 2 clinical testing. Novel antibody fragments have been expressed and validated, including single, bispecific, and biparatopic ScFv, and single and tandem VHH. The functionality of immune checkpoint inhibitors and immune inhibitors will be presented. Perspectives on the expression of combination therapeutics with fragment antibodies and other immune modulatory proteins within the solid tumour microenvironment will be discussed.

09:00

Local Secretion of Immune Active Proteins for Direct and Bystander Tumour Killing

Jonathan Fisher, PhD, Group Leader, University College London

Whilst striking success has been seen using CAR T cells to treat leukaemia and other cancers, the majority of solutions are autologous leading to high production costs and logistical challenges. Vg9Vd2-gdT cells are a versatile, non-alloreactive chassis for cellular immunotherapy, possessing potent antibody-dependent cellular cytotoxicity (ADCC) capacity, a tissue-tropic homing profile, and a range of innate tumour-sensing receptors minimizing the likelihood of tumour escape. OPS-gd is a cell therapy platform harnessing these properties, secreting tumour-targeting opsonins and armouring cytokines. OPS-gd cells show equivalent cytotoxicity but superior exhaustion phenotype to CAR-abT cells, recruitment of ADCC-competent bystanders, and in vivo efficacy against patient-derived osteosarcoma.

09:30 Transgenic Llama Mice - a Fast and Flexible Single Domain Discovery Tool

Alessa Schaffrath, Doctoral Student, UKE Hamburg

Looking to rapidly generate a diverse pool of single-domain antibodies? Learn how Genovac partnered with UKE to establish a unique discovery platform combining Genovac’s single cell screening tools with UKE’s transgenic Llama mice, LaMice. UKE’s transgenic LaMice are the latest addition to Genovac’s class leading host species options which now includes transgenic and wild type mice and rats, rabbits, llamas and convalescent patient human cells.

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

INNOVATIVE APPROACHES TO THE CHALLENGES OF SOLID TUMOURS

11:45 KEYNOTE PRESENTATION:

AI-Based Target and Antibody Discovery from Patient Tumour Profiles

Xiaole Shirley Liu, PhD, CEO, GV20 Therapeutics

Patient tumours contain anti-tumour antibodies but might not have sufficient abundance and correct Fc to cure the tumours. GV20 uses bioinformatics and AI to decode natural B-cell responses from large cohorts of patient tumours to uncover novel targets and antibodies simultaneously. It already brought a first-in-class antibody against a novel innate checkpoint to Ph1 clinical trial in the US.

12:15

The Influence of DM1, MMAE, and MMAF on Biodistribution and Preclinical Therapeutic Efficacy of Affibody-Based Drug Conjugates

Torbjörn Gräslund, PhD, Professor, Department of Protein Science, KTH Royal Institute of Technology

Affibody molecules are small engineered alternative scaffold affinity proteins that can be site-specifically loaded with cytotoxic drugs to create homogenous conjugates with a desired drug-to-carrier ratio. The presentation will explore the targeting of HER2 and HER3 with affibody-based drug conjugates. It will also describe the impact on biodistribution and in vivo cytotoxic efficacy of drug conjugates loaded with auristatin and maytansine-derived payloads.

12:45 Cells meet biosensor – Automated determination of binding kinetics on living cells

Nena Matscheko, Dr., Team Lead R&D Cells and Antibodies, R&D, Dynamic Biosensors GmbH

Real-time binding kinetics are a pivotal element of drug candidate characterization. Dynamic Biosensors’ innovative Real-Time Interaction Cytometry (RT-IC) technology enables automated time-resolved measurements of antibodies, small molecules, or proteins (e.g. pMHC) interacting directly with cells immobilized in the heliXcyto biosensor. This renders cumbersome membrane protein purification superfluous and kinetic results (kon, koff, Kd) include effects of native co-interactions, avidity, and PTMs for highest relevance.

Presentation to be Announced13:00

Session Break13:15

13:20 LUNCHEON PRESENTATION I:Expression and Characterization of Novel GCN-Related N-acetyltransferases by Nuclera's Rapid Protein Access System

Hans Gerstmans, Postdoctoral Researcher, Laboratory for Biomolecular Discovery & Engineering, KU Leuven, VIB

Wai Long Tam, Head of Technology Watch at VIB, Technology Watch, VIB

GCN5-related N-acetyltransferases (GNATs) play a role in the acetylation of various proteins. GNATs are relevant to antibiotic resistance as integral enzymes that bacteria employ to modify antibiotics evading their effects and contributing to the development of resistance. Despite the importance of these enzymes, they have not been expressed and published on. Using Nuclera’s eProtein Discovery system, we were able to find the expression conditions and obtain GNATs.

 

 

Session Break14:20

14:30

Chairperson’s Remarks

Kerry Chester, PhD, Professor, Molecular Medicine, University College London

14:35

Broadening Specificity of T-Cell Receptors for HLA-A*03:01/A*11:01 Alloselectivity

Vijaykumar Karuppiah, PhD, Associate Director, Protein Engineering, Immunocore

The recent approval of the first TCR-based drug represents an exciting advance for cancer immunotherapy. While HLA restriction may potentially limit patient coverage, this may be overcome when the same peptide antigen is presented on multiple HLA alleles. Here, we show that a TCR targeted towards a peptide presented by an HLA allele can be optimised by structure-guided mutations to also bind a second allele with similar affinity and potency.

ADVANCES IN DISCOVERY AND ENGINEERING OF ANTIBODIES FOR NON-CANCER TARGETS

15:05

Discovery of Broadly-Neutralizing Antibodies against Coronaviruses

Joshua Tan, PhD, Chief, Antibody Biology Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health

The potential for future coronavirus outbreaks and ongoing mutations in SARS-CoV-2 highlight the need to broadly target this group of pathogens. Here, we used an epitope-agnostic approach to identify two groups of monoclonal antibodies that target distinct regions of the spike protein and broadly neutralize diverse coronaviruses. Top neutralizers from both antibody classes neutralized all SARS-CoV-2 variants of concern tested including the Omicron subvariant XBB.1.5, inhibited fusion mediated by SARS-CoV-2 spike, and limited disease caused by SARS-CoV-2 in a Syrian hamster model. This two-step approach of isolating rare neutralizing mAbs against cryptic targets is highly relevant for pandemic prevention and the development of therapeutic tools against emerging pathogens.

15:35 Antibody Discovery Dead Ends and New Approaches

Anthony Stajduhar, Director of Global Business Development, Business Development, Rapid Novor

Antibody discovery remains one of the most challenging aspects in antibody therapeutic development.  Novel proteomics-based approaches to antibody discovery offers a promising strategy to overcome roadblocks often associated with other discovery technologies. With REpAb polyclonal sequencing, antibody discovery with mass spectrometry enables the exploration of the natural immune repertoire with unparalleled antibody diversity and creates a pathway for the discovery of novel human antibodies.

Refreshment Break in the Exhibit Hall with Poster Viewing16:05

17:00

A Patient-First Approach to Discover First-in-Class Antibody Therapeutics

Jorge Dias, PhD, Principal Scientist, Alchemab Therapeutics Ltd.

Identifying genuinely novel targets that have immense therapeutic potential is an increasing challenge across central nervous system diseases. Alchemab addresses this challenge by truly disrupting the target discovery process — simply put, we let the patient select the best targets. We have discovered novel disease-relevant targets through our proprietary platform that searches for protective auto-antibodies in patients who are resistant to disease.

17:30

Advancing Snake Envenomation Treatment: Designing the Next Generation of Antivenoms

Sandra Ergueta-Carballo, PhD, Project Coordinator, University of Cambridge

Snake envenomation kills over 100,000 people annually and cripples three times as many. Current treatment relies on animal-derived serum, which, although life-saving presents many drawbacks, including lack of standardisation, a low proportion of neutralising antibodies, and serum sickness from injecting grams of animal protein. Therefore, it is crucial to modernise snakebite treatment. This presentation will show the ongoing efforts in developing the new generation of antivenoms and their associated challenges.

18:00

Advancing Brain Shuttle-Enabled Therapeutics for Efficient Delivery to CNS —Translation to Primates

Pawel Stocki, PhD, Vice President Research, Ossianix

Brain delivery across blood-brain barrier (BBB) is a major hurdle in the development of biological therapeutics for CNS disorders. We developed TXP1 brain shuttle based on anti-TfR1 antibody that can be fused to any therapeutic payload. In monkeys, TXP1 demonstrated ~35-fold improved brain penetration over the control, reaching up to 7.5% brain/plasma ratio. TXP1 showed to be safe and provided brain-specific delivery with no accumulation in other organs and had long-lasting brain PK, essential for efficacious CNS therapies.

18:30

Development of Autoregulating FVIII-Mimetic Bispecific Antibodies to Reduce Risk of Prothrombotic Events in Treatment of Haemophilia A

Vincent Muczynski, PhD, Director, NovalGen

Engineered proteins are powerful medicines, but may carry a risk of life-threatening toxicities. We have developed a broadly applicable approach of autoregulatable drugs by harnessing treatment-related biological signals to trigger inactivation of the drug at a set threshold through a negative feedback loop to prevent/attenuate high-grade adverse events. Using emicizumab, a marketed bispecific antibody for haemophilia A, we successfully demonstrated that autoregulation reduces frequency of thrombotic events.

Close of Emerging Targets and Therapeutic Approaches Conference19:00