Main Conference Day 3 - European Time GMT+1
- Sai Reddy, PhD - Associate Professor of Systems and Synthetic Immunology, ETH Zurich
We present Germinal, a model for performing epitope-targeted design of de novo nanobodies and scFvs against a wide range of epitopes, including the ability to design specificity or breadth across multiple targets.
- Brian Hie - Assistant Professor of Chemical Engineering, Stanford University
Generative AI is redefining what’s possible in biologics discovery — enabling scientists to explore protein sequence space more efficiently, design higher-performing molecules, and accelerate the path from concept to candidate. At Cradle, we are developing intuitive software tools that allow protein engineers to directly leverage generative AI models within their existing workflows. This presentation will showcase how Cradle’s technology enables scalable and reproducible protein design, combining advanced AI architectures with user-centered design to reduce cycle times and ultimately create better biologics.
- Eli Bixby - Co-Founder and ML Lead, Cradle
We ran an open protein design competition in where 100+ participants from industry and academia submitted binders against Nipah virus. Testing 1000 sequences experimentally in our lab revealed which computational methods translate to experimental success, with hit rates varying 10-fold between approaches. We'll share lessons on benchmarking design pipelines, the value of negative data, and how open competitions can accelerate the field when paired with standardized experimental validation.
- Julian Englert - CEO and Co-Founder, Adaptyv Bio
- Janine Schuurman, Ph.D. - Biotech Consultant, Lust for Life Science
- James Ernst, Ph.D. - Vice President, Development Sciences; Head of Protein Sciences & Technology, Xencor
- Darko Skegro, PhD - Director and Site Head, Biotherapeutic Engineering and Conjugation, Novartis
Ficerafusp alfa, is a first-in-class EGFR-TGF-b bifunctional antibody. TGF-β signaling within the tumor microenvironment (TME) creates fibrotic, immunosuppressive barriers that impede tumor penetration of immune cells. Mechanistic insights from paired biopsies and preclinical studies reveal the importance of tumor targeted inhibition of TGF- b in breaking down barriers to tumor penetration. Ultimately, ficerafusp alfa’s tumor penetration has the potential to drive deep and durable responses that lead to long-term clinical benefits.
- Brenda O'Connell - Senior Director, Translational Science, Bicara Therapeutics
Stellabody® is a single point mutation in the CH3 region that facilitates “on-target assembly” of immune biologics that transforms killing or agonistic potency in multiple immune protein formats i.e. mAbs, bispecific antibodies, Fc-fusions and novel scaffolds. Stellabody biologics mediate greatly (10-100x) enhanced potency in head-to-head comparisons with the equivalent standard biologic including standard-of-care mAbs in oncology on primary patient-derived clinical samples and targets in infection and immunology.
- Vijaya Pattabiraman, PhD - Co-founder and CTO, Bright Peak Therapeutics
- Minami Maeda - President, Rakuten Medical
- Hung-En Hsia, PhD - Bioanalytical Scientist, Roche Innovation Center Munich
Ability Biotherapeutics presents AbiLeap™ – a proprietary AI-enabled platform to systematically generate conditionally active antibodies. Trained on one of the largest therapeutic antibody databases, AbiLeap achieves unprecedented mutational reach (up to 25 amino acid changes) while retaining epitope specificity and therapeutic-ready humanness. Using AbiLeap we have generated leads against 4 targets and are pursuing IND-enabling studies by 2027.
- Jean-Philippe Bürckert, Ph.D. - Chief Technology Officer, Ability Biotherapeutics
- Pierce Ogden, PhD - Co-Founder, CTO and Board Director, Manifold Bio
- Basile Wicky, PhD - Assistant Professor, Department of Biosystems Science and Engineering, ETH Zürich
- Simone Oostindie - Director, Research and Discovery, Gyes BV
- Alexey Lugovskoy, PhD - President and CEO, Diagonal Therapeutics
Adeno-associated virus (AAV) gene therapies face challenges in tissue-specific delivery and safety. We have developed bispecific Altibody for precise AAV retargeting to muscle and CNS. This innovative "plug-and-play" approach enhances on-target transduction efficacy and safety in mice, marking a milestone in AAV retargeting strategies for gene therapy applications.
- Yang Shen, PhD - Executive Director, Antibody Engineering & Bispecifics, Regeneron
- Karen Silence, PhD - Head Preclinical Product Development, Argenx
- Katherine Harris, PhD - Chief Development Officer, Rondo Therapeutics
- Sophia Hober, PhD - Professor of Molecular Biotechnology, KTH Royal Institute of Technology
Systemic toxicities from target expression on healthy tissues limit cancer therapy. The PrimeBody platform employs protease-cleavable linkers and affinity-tuned masking to generate tumor-activated biologics with exceptional systemic stability and efficient tumor activation. VOR-101, a masked, Fc-enhanced CD47 blocker, remains inert in circulation but rapidly activates in tumors, achieving >700-fold selectivity. It delivers ~100-fold higher exposure than conventional agents without toxicity, driving durable tumor regressions and a markedly improved therapeutic index.
- Ugur Eskiocak, Ph.D - CEO, Voro Therapeutics
T cell engagers (TCEs) show promise in solid tumors but are constrained by cytokine release and on-target, off-tumor toxicity. Amberstone’s T-MATE™ platform overcomes these barriers with a pH-gated, target-selective mechanism that adapts to tumor pH heterogeneity while maintaining an optimal therapeutic profile. This conditional strategy enables a new generation of TCEs that combines both safety and efficacy with broad applicability across diverse solid tumor indications.
- Aude Segaliny, PhD - Senior Director, Head of Research & Early Development, Amberstone Biosciences
PRO-XTEN™ masked T cell engagers (TCEs) are conditionally activated immune therapeutics designed to exploit dysregulated protease activity within the tumor microenvironment. This approach enables tumor-selective TCE activation while minimizing systemic toxicity. The XTEN mask serves a dual function: masking TCE activity and extending the half-life of the masked molecule, while the proteolytically activated TCE exhibits a short half-life, creating conditional pharmacokinetics that enhance tumor exposure while limiting systemic active drug circulation. Preclinical studies demonstrated: (1) robust masking with ~10,000-fold reduction in cytotoxicity in vitro; (2) potent anti-tumor efficacy in vivo at doses comparable to unmasked TCE controls; and (3) >100-fold improvement in maximum tolerated dose (MTD) in non-human primates. These findings support a significantly enhanced therapeutic window through tumor-specific activation and conditional PK. Clinical trials evaluating PRO-XTEN™ TCEs targeting HER2, PSMA, and EGFR are currently underway, with early data demonstrating the translational potential of this protease-activated platform.
- Davide Corti - SVP, Antibody Research, Vir Biotechnology
- Uli Bialucha, Ph.D - Chief Scientific Officer, Xilio Therapeutics
- Marco Preuss, Ph.D - Project Leader, Process Development, Bioconjugate, Lonza
