In-Person Interactive Discussions

Engage in in-depth discussions with industry experts and your peers about the progress, trends and challenges you face in your research! Interactive discussion groups play an integral role in networking with potential collaborators, provide an opportunity to share examples from your work, and be part of a group problem-solving endeavor.

These will take place IN-PERSON ONLY.

Wednesday, May 14: 1:40 – 2:25 PM

Engineering Antibodies

TABLE 1: The Impact of Artificial Intelligence (AI) on Biologics Discovery and Optimization
Moderator: Christopher R. Corbeil, PhD, Research Officer, Human Health Therapeutics, National Research Council Canada

  • Identifying the highest priority opportunities for AI in biologics discovery, alongside the primary blockers and risks that currently stifle progress
  • Strategies for enhancing data diversity, access, preparation and management to increase the accuracy of AI models
  • Speculating on the next significant breakthroughs that could revolutionize biologics discovery.
  • Assessing the impact of partnerships and collaborative projects on the advancement of AI in biologics discovery, including the hurdles these collaborations face

Advancing Bispecific Antibodies and Combination Therapy to the Clinic

TABLE 2: How Model-Guided Design of Multi-Specific Modalities Can Benefit Your Immune-Oncology Program
Moderator: Michael G. Zager, PhD, Associate Research Fellow, Discovery and Early Development, Pfizer

  • Designing multi-specific modalities can take significantly more time and resource than standard monoclonal antibodies, and often the project team is still left with the question whether their clinical candidate is optimal for the targets and intended pharmacology
  • Mechanistic and systems-level models of pharmacokinetics, target binding and resulting pharmacology (often referred to as quantitative systems pharmacology, or QSP) provides a platform that holistically integrates disparate data that are otherwise difficult to contextualize quantitatively to make critical design decisions. Data such as affinities, crosslinking rates, target expression and internalization, pharmacokinetic properties of the antibody, distribution to the site of action and downstream pharmacology can all be considered together via computational simulations.
  • These computational methods can lead to data-informed decisions that can significantly cut time and resource and can provide a powerful complement to structural design. For example, simulations may demonstrate that further affinity maturation of current leads will not result in increased pharmacology.
  • However, these models require data and take time to create. Therefore, planning for their development, including data generation, well in advance is key for them to provide impact on designs. This is one of the most significant challenges these efforts typically face.
  • In this breakout session, we plan to discuss the utility of these models in modality design, whether we as developers of these models are asking and answering the right questions in the eyes of the protein engineers, and openly discuss what we need to focus on to improve their usefulness.

Cell-Based Immunotherapies

TABLE 3: Protein Engineering of Multi-Specifics for Tailored immunotherapies
Moderator: Hilmar Ebersbach, PhD, SVP Antibody Development & Protein Engineering, ImmunOs Therapeutics AG

  • What are design principles?
  • What are challenges?
  • What is impact of IgG subclass for Fc-fusion proteins?
  • How to profile multispecifics?
  • What are developability considerations and characterization?

Optimizing Protein Expression

TABLE 4: Accelerating Preclinical Discovery and Development:  Getting Your Favorite Protein Quickly and Cleanly
Moderator: David W. Wood, PhD, Professor, Chemical & Biomolecular Engineering, The Ohio State University

  • Deciding the best expression strategy for new proteins: shotgun or informed guess?
  • Balancing the best host: slow and reliable or quick and messy?
  • How pure is pure enough?  Do we always need tags?
  • What happens when you just can’t get what you need out of your strategy?  What next?

Biophysical Methods

TABLE 5: Challenges and Advancements on AAV Drug Product Formulation Development
Moderator: Marilia Barros, PhD, Principal Scientist, Regeneron Pharmaceuticals

  • AAV formulation development common challenges and strategies to support long-term stability. Lessons learned from different AAV serotypes
  • Is it possible to develop a platform formulation that can be applied across all different AAV serotypes? Discuss novel and critical excipients, lyo vs liquid formulation
  • Stability-indicating and extended characterization methods to characterize AAV drug product critical quality attributes (aggregation, full-empty ratio, genome titer etc)
  • High-throughput and low-volume methods to support formulation screening and guide optimal formulation development

mRNA Therapeutics

TABLE 6: Bioanalysis and Immunogenicity of mRNA Therapeutics
Moderator: Xiaobin Zhang, PhD, Principal Scientist, Takeda Pharmaceuticals

  • Challenge issues in the bioanalysis of mRNA therapeutics with different technologies: CE, qRT-PCR, LC-MS et al.
  • Impurity characterization for risk Assessment: impurity levels, criteria, and justification.
  • Fit-for-purpose bioanalytical approaches to meet the needs of pharmacokinetics, pharmacodynamics, and immunogenicity assessment with mRNA therapeutics
  • Product-related analysis to evaluate the immunological effects with mRNA therapeutics: protein expression from mRNA, infectious disease mRNA vaccine, and cancer mRNA vaccine.​

Friday, May 17: 7:30 – 8:25 AM

Machine Learning Approaches for Protein Engineering

TABLE 1: Engineering Novel Cytokine Functions through Experimental and Computational Approaches
Moderator: Jamie B. Spangler, PhD, Assistant Professor, Biomedical Engineering and Chemical & Biomolecular Engineering, Johns Hopkins University

  • Design parameters and objectives for cytokine engineering
  • Tailoring engineering strategies to various cytokine systems
  • Considerations for selecting the appropriate evolutionary and/or rational design strategies
  • Key factors in choosing a computational workflow for cytokine engineering

TABLE 2: De novo peptide sequencing: Applications and opportunities
Moderator: Timothy Patrick Jenkins, PhD, Assistant Professor & Head, Data Science, DTU Bioengineering

  • What tools are out there and how good are they?
  • What applications benefit most from de novo peptide sequencing?
  • What are current limitations and opportunities for further development?

Engineering Bispecific Antibodies

TABLE 3: TRACeR: A Platform for Multimodal Antigen-Focused Targeting of MHC
Moderator: Possu Huang, PhD, Assistant Professor, Bioengineering, Stanford University

Maximizing Protein Production Workflows

TABLE 4: R&D with the End-in-Mind
Moderator: Bjørn Voldborg, MSc, Head, National Biologics Facility, DTU Bioengineering, Technical University of Denmark

  • Determine what your end-product could look like
  • Consider the full pipeline of your candidate as early as possible
  • Select suitable expression systems early on
  • Use regulatory acceptable hosts as soon as possible

TABLE 5: Biotherapeutic Expression & Production Pipeline – Week in Review
Moderators:
Richard Altman, MS, Field Application Scientist, Life Science Solutions, Thermo Fisher Scientific
Henry C. Chiou, PhD, Senior Director General Manager, Biosciences, Thermo Fisher Scientific
Dominic Esposito, PhD, Director, Protein Sciences, Frederick National Laboratory

  • Highlights from the week’s presentations
  • New technologies and strategies
  • With these new technologies, how do we increase not only the efficiency and throughput of recombinant protein expression, but of the entire protein production process?
  • Challenges and future trends

Characterization for Novel Biotherapeutics

TABLE 6: Characterization, Screening and Design of Bispecific Antibodies
Moderator: Dennis Åsberg, PhD, Project Manager, Global Research Technologies, Novo Nordisk A/S

  • Which specific challenges are seen when screening for developability of bispecific antibodies?
  • How can the characterization of the parent antibodies inform on the properties of the bispecific antibody?
  • How to assess and define purity of multivalent-antibody or multi-specific formats?
  • Can a developability profile and/or formulation stability on par with standard antibodies normally be reached for multivalent or multi-specific formats?

Register Now

View By:


Premier Sponsors

   FairJourneyBiologics Integral-Molecular_NEWLONZAOmniAb  Samsung_Biologics  UnchainedLabs