Cambridge Healthtech Institute’s 24th Annual

Engineering Antibodies

Strategies and Science for Engineering Next-Generation Biotherapeutics

May 16 - 17, 2023 ALL TIMES EDT

The field of protein engineering is at an exciting point in its development, with new generations of biotherapeutics now progressing through development and into the market, major technology advances, and a body of discovery stage and clinical evidence that can be used to inform the development of safe, highly effective therapies for unmet medical needs. The 24th PEGS Engineering Antibodies conference offers protein engineers an important annual forum for discussing new technologies and strategies for overcoming complex engineering challenges including membrane-bound targets, agonism, delivery and specificity, and support of discovery stage applications of ML/AI in refining lead selections. The track also explores the ongoing efforts of pharma companies to accelerate this stage of R&D and resolve bottlenecks.

Sunday, May 14

- 5:00 pm Main Conference Registration1:00 pm

Recommended Pre-Conference Short Course2:00 pm

SC1: Antibody Drug Discovery: From Target to Lead

*Separate registration required. See short courses page for details.


Tuesday, May 16

Dessert Break in the Exhibit Hall with Poster Viewing1:40 pm

ENGINEERING FOR IMPROVED FC FUNCTION

2:15 pm

Chairperson’s Remarks

Katherine A. Vallis, PhD, Group Leader, Oxford Institute for Radiation Oncology; Professor, Experimental Radiotherapeutics, University of Oxford

2:20 pm

Engineering the Antibody Fc for Conditional Activity in the Solid Tumor Microenvironment

Jennifer A. Maynard, PhD, Henry Beckman Professor, McKetta Department of Chemical Engineering, Cockrell School of Engineering, University of Texas Austin

Antibody-based therapeutics enjoy considerable successes as cancer treatments, but can cause serious toxicities due to recognition of tumor-associated antigens in non-cancerous tissues. We will discuss recent efforts to develop advanced antibody therapeutics with Fc-mediated activities that are restricted to the acidic solid tumor microenvironment. With the intent of decreasing toxicities and expanding therapeutic windows, protein engineering strategies can render antibody activity sensitive to multiple tumor-specific characteristics.

2:50 pm

The Role of the Fc Domain in Immunity and Disease Outcomes

Bronwyn M. Gunn, PhD, Assistant Professor, Washington State University

The antibody Fc domain shapes protective immunity against many different viral pathogens. We will present a Systems Serology approach to define antibody Fc features associated with protection from Ebola virus and describe how we have used antibody engineering approach to validate and mechanistically dissect the role of distinct Fc-mediated functions in protection. Further, we will discuss how to translate these approaches to other pathogens, such as SARS-CoV-2.

3:20 pm Structure-Based Charge Calculations for Predicting Properties and Profiling Antibody Therapeutics

Nels Thorsteinson, Director of Biologics, Chemical Computing Group

We present a method for modeling antibodies and performing pH-dependent conformational sampling, which can enhance property calculations. Structure-based charge descriptors are evaluated for their predictive performance on recently published antibody pI, viscosity, and clearance data. From this, we devised four rules for therapeutic antibody profiling which address developability issues arising from hydrophobicity and charged-based solution behavior, PK, and the ability to enrich for those that are approved by the FDA.

Refreshment Break in the Exhibit Hall with Poster Viewing3:50 pm

CHALLENGING TARGETS

4:30 pm

Identification of Rare Binders to Challenging Targets from a Phage Display Library Using Flow Cytometry and Biolayer Interferometry

Dong hee Chung, PhD, Postdoctoral Researcher, University of California, San Francisco

In vitro biopanning platforms have expanded the field of antibody identification beyond immunization. However, applying these strategies to identifying binders against challenging targets remains a challenge. Here, we present a new pipeline, RAPID (Rare Antibody Phage Isolation and Discrimination), for the identification of rare high-affinity antibodies against challenging targets. RAPID is enabled by fluorescent labeled phage displayed antibody libraries, and fluorescent activated sorting allows for the isolation of promising binder populations. A Biolayer Interferometry method allows for candidate clones to be discriminatorily screened. We introduce this method with a well characterized Antigen-Antibody pair, and applications of RAPID biopanning are presented.

5:00 pm

Activating and Modulating Antibody Therapeutics for Difficult Membrane Protein Targets

Richard C. Yu, PhD, Co-Founder & CEO, Abalone Bio, Inc.

Antagonist antibodies for membrane protein targets like GPCRs and ion channels are difficult to discover. Agonist and modulating antibodies are even more challenging. Here I will discuss the challenges in the field and strategies for discovering and developing antibodies that access the full spectrum of activities against membrane protein targets.

5:30 pm

Multi-Scale Simulations Reveal Antibody Reach and Energetics of Binding

Daniel Nissley, PhD, Florence Nightingale Bicentenary Research Fellow and Tutor in Bioinformatics, Department of Statistics, University of Oxford

Molecular simulation techniques can help solve the inverse problem of what antibody structures give rise to an experimental observable and provide valuable insight into the biophysical origin of antibody properties. Here, we use coarse-grain and all-atom molecular dynamics to accurately model how far antibodies can reach to bind antigens at both arms and to reveal differences in how TCR-mimetic antibodies bind pMHC targets in comparison to TCRs.

Close of Day6:00 pm

Dinner Short Course Registration6:00 pm

Recommended Dinner Short Course6:30 pm

SC6: Developability of Bispecific Antibodies

*Separate registration required. See short courses page for details.

Wednesday, May 17

Registration and Morning Coffee7:30 am

ENGINEERING FOR TARGETED DELIVERY AND IMPROVED SPECIFICITY

8:25 am

Chairperson’s Remarks

Danielle DiCara, PhD, Principal Scientific Researcher, Antibody Engineering, Genentech, Inc.

8:30 am

Intracellular Delivery of Antibodies for Cancer Applications

Katherine A. Vallis, PhD, Group Leader, Oxford Institute for Radiation Oncology; Professor, Experimental Radiotherapeutics, University of Oxford

A significant barrier to cancer treatment is that although small molecule drugs easily enter cells, many intracellular oncoproteins lack suitable binding pockets, and so are unresponsive to them. Antibodies can act across a broad surface, so inhibiting protein-protein interactions, but unfortunately do not naturally enter cells. We have developed multimeric, cyclised cell-penetrating peptides that transfer functional antibodies efficiently across the cell membrane, allowing the targeting of previously “undruggable” intracellular molecules.

9:00 am

Development of T Cell Engagers Selective for Cells Co-Expressing Two Antigens

Danielle DiCara, PhD, Principal Scientific Researcher, Antibody Engineering, Genentech, Inc.

For the development of safe and effective T Cell Engagers (TCEs) for solid tumors, it is highly desirable to expand the number of available target antigens and also to increase the precision with which a TCE can differentiate tumors from healthy tissue. I will present a strategy to reduce on-target, off-tumor TCE activity by targeting co-expression of two tumor-associated antigens and describe engineering of trispecific antibodies with this selectivity.

9:30 am

KEYNOTE PRESENTATION: Inhibition of Key Intracellular Targets via the Cytosolic Delivery of Antibodies and Proteins

Andrew Tsourkas, PhD, Co-Director, Center for Targeted Therapeutics and Translational Nanomedicine; Professor, Bioengineering, University of Pennsylvania

A limitation of biologics is their inability to cross the cell membrane. Conversely, small molecules readily cross cell membranes, but many intracellular proteins lack pockets for small molecule binding. We developed a method to deliver antibodies into the cytosol, which enabled us to inhibit cancer-associated proteins. We also delivered small, protein scaffolds intracellularly for therapeutic inhibition of conventionally-undruggable targets.  Major themes to be covered include: a) modification of antibodies and proteins with anionic polypeptides for complexation with cationic lipids and efficient intracellular delivery; b) photoreactive antibody binding domains for efficient, site-specific labeling of antibodies; and c) antibody/protein-mediated inhibition of "undruggable" intracellular targets.

10:00 am Isolation of selective antibodies targeting GLUT-1 transporter to inhibit cancer metabolism

Siret Tahk, PhD, Senior scientist, Icosagen Therapeutics

Multi-pass integral membrane proteins compose the largest therapeutically relevant group of proteins that have so far not been effectively targeted with antibody-based molecules. Icosagen has developed a wide array of expertise and technologies in protein production, antibody development, protein engineering and analytics over the past 10 years and here we present the integration of these technologies by launching our therapeutic antibody development pipeline to specifically target those proteins.

Coffee Break in the Exhibit Hall with Poster Viewing10:30 am

Transition to Plenary Keynote Session11:10 am

PLENARY KEYNOTE SESSION

11:20 am

Plenary Keynote Introduction

Maria Wendt, PhD, Global Head and Vice President, Digital and Biologics Strategy and Innovation, Sanofi

11:30 am

Advancing Innovative Biologics Modalities from Research to Clinical Application – Novel Platforms, Automation, and Computation

Rebecca A. Sendak, PhD, Head, Global Large Molecules Research Platform, Sanofi

Addressing disease biology in the clinic with protein therapeutics has become increasingly complex. Turning to innovative and novel scaffolds offers opportunities to tailor therapeutics not previously possible due to advances in host cell engineering and protein design approaches. Designing and developing these modalities requires a next-generation approach as we exploit increased potential design space and also growing data sources to leverage as we invent the next wave of therapeutics.

YOUNG SCIENTIST KEYNOTE

12:15 pm

Engineering Prime Editor Proteins for Therapeutic Applications

Andrew V. Anzalone, MD, PhD, Director & Head, Prime Editing Platform, Scientific Co-Founder, Prime Medicine, Inc.

Precision gene editing technologies have the potential to address a wide range of genetic diseases. Prime Editing is a recently developed “search-and-replace” gene editing approach that can precisely perform a wide variety of DNA sequence edits at programmed target sites in human genomes without requiring double-strand DNA breaks or donor DNA templates. I will describe advances to prime editing technology that improve its efficiency, specificity, and capabilities for therapeutic applications.

Session Break1:00 pm

1:10 pm LUNCHEON PRESENTATION I:Building Better BioTherapeutics using Machine Learning and Synthetic Biology

Claes Gustafsson, PhD, Chief Commercial Officer & Co-Founder, ATUM

 ATUM's integrated pipeline enables Machine Learning algorithms in conjunction with the ability to synthesize sets of systematically varied protein therapeutics to search very large space (>10^15) with just a few hundred protein variants, enabling multidimensional fitness optimization of 'hard-to-measure' functions. We integrate this engineering process directly into Leap-In transposon-mediated stable cell lines for rapid generation of gram quantities of target protein therapeutics.

1:40 pm LUNCHEON PRESENTATION II:Take Back Control of Your Timelines and Budget through Automated Oligo Synthesis in Your Lab

Raymond DiDonato, PhD, Senior Territory Account Manager, DNA Script

Engineered proteins, therapeutic antibodies, and mRNA vaccine candidates are constructed via gene assembly or mutagenesis and cloned into a vector for expression and scale up. R&D groups either construct sequences of shorter oligos synthesized with phosphoramadite chemistry, or order chemically synthesized fragments from third-party gene synthesis companies.  Both approaches create a bottleneck and delays identifying lead candidates which diminish R&D budget for vaccine and therapeutics development. Learn how to synthesize enzymatic oligos and assemble 1-2 KB genes in a day.

 

INTERACTIVE DISCUSSIONS

2:10 pmFind Your Table and Meet Your Moderator
2:15 pmInteractive Discussions

Interactive Discussions are informal, moderated discussions, allowing participants to exchange ideas and experiences and develop future collaborations around a focused topic. Each discussion will be led by a facilitator who keeps the discussion on track and the group engaged. To get the most out of this format, please come prepared to share examples from your work, be a part of a collective, problem-solving session, and participate in active idea sharing. Please visit the Interactive Discussions page on the conference website for a complete listing of topics and descriptions.

TABLE 1: Implementation Challenges for Machine Learning as a Tool for Antibody Discovery - IN-PERSON ONLY

Christopher R. Corbeil, PhD, Research Officer, Human Health Therapeutics, National Research Council Canada

  • Current successes
  • Experimental validation and POC
  • Bottlenecks and challenges
  • Needs from IT and solution providers​
BREAKOUT DISCUSSION:

TABLE 2: Would Increasing In Vivo Data Generation Increase Probability of Clinical Success? - IN-PERSON ONLY

Pierce J Ogden, PhD, Co Founder & CSO, Manifold Biotechnologies Inc.

  • What preclinical in vivo data are most often predictive of clinical success
  • What are some ways we could increase in vivo throughput
  • How can AI and machine learning be utilized in conjunction with in vivo data
  • How to improve the in vitro to in vivo drug development process by supplementing with early in vivo discovery workflows
  • Unbiased in vivo based therapeutic discovery and the required in vivo throughput​

ACCELERATING AND OPTIMIZING PROTEIN ENGINEERING

3:00 pm

Chairperson’s Remarks

Daniel R. Woldring, PhD, Assistant Professor, Chemical Engineering & Materials Science, Michigan State University

3:05 pm

Computational Methods to Complement, Enrich, and Accelerate Antibody Discovery Programs

Daphne Truan, PhD, Associate Director, Protein Design and Informatics, GlaxoSmithKline

In the eternal race to make better biotherapeutics faster, GSK Biopharm discovery pipelines are continuously revamped by a tight collaboration among different departments. This presentation will focus on the computational methods that help select better targets, design and enrich screening libraries, and accelerate the multiobjective optimization of lead candidates through the analysis of NGS data with developability criteria.

3:35 pm

Multiplexed In Vivo Drug Discovery Using a Novel Protein Barcoding Technology

Pierce J Ogden, PhD, Co Founder & CSO, Manifold Biotechnologies Inc.

When engineering therapeutics, in vivo validation remains the primary bottleneck to program advancement. We invented a novel method that enables multiplexed quantification of 100s of protein therapeutics in vivo. We have leveraged our in vivo data with machine learning to perform in vivo molecule design. Further, we show that in vitro assay results are often poorly predictive of downstream in vivo results, highlighting the importance of increased in vivo throughput.

4:05 pm Fit-for-Purpose Strategies for Discovery of Diverse Antibody Panels Against Challenging Targets Using Alivamab Mouse

Jane Seagal, PhD, Vice President, Antibody Discovery, AlivaMab Discovery Services

Success in antibody discovery requires flexibility and risk mitigation. ADS’ fit-for-purpose antibody discovery strategies are empowered by the combinatorial and somatic diversity produced by the suite of AlivaMab® Mouse strains and enhanced through ADS’ unique capabilities for recovering and interrogating an immune repertoire. Case studies featuring ADS strategies for rapidly identifying diverse antibody panels against challenging targets from AlivaMab Mice will be presented.

Ice Cream Break in the Exhibit Hall with Poster Viewing4:35 pm

PEGS BOSTON COMMON: SPEED-NETWORKING

4:45 pm

How Many New Contacts Can You Make? - IN-PERSON ONLY

Mary Ann Brown, Executive Director, Conferences, Cambridge Healthtech Institute

Bring yourself, your business cards or e-cards, and be prepared to share and summarize the key elements of your research in a minute. PEGS-Boston will provide a location, timer, and fellow attendees to facilitate the introductions.

STRUCTURAL MODELING IN ANTIBODY DISCOVERY AND ENGINEERING

5:10 pm

GYST Platform: A Computational Sherpa for Augmented Exploration of Antibody Landscapes in Discovery and Engineering

Seth F. Harris, PhD, Director, Structural Biology, Genentech, Inc.

Effective navigation and distillation of large structural datasets necessitates programmatic approaches. We describe the custom structural informatics platform (GYST) we are developing and its application to study thousands of fab interfaces for opportunities to engineer novel polyvalent fab formats. This platform allows expansion to other protein families within and beyond large molecule drug discovery. We also discuss how the amassed, pre-computed annotations are suited for training deep learning models.

5:40 pm

VHH CDR-H3 Conformation Correlates with Germline Usage: Implications for VHH Ontogeny and Engineering

Patrick Koenig, PhD, Senior Scientist, Antibody and Protein Engineering, 23andMe Inc

VHHs or nanobodies are single antigen binding domains originating from camelid heavy-chain antibodies. Analyzing variable domain structures from llama and alpaca we found that VHHs can be classified into structural clusters based on their CDR-H3 conformation. The clusters have distinct functional properties in how they interact with the antigen. VHHs from the two clusters originate from different VH germlines, demonstrating a previously undescribed impact of germline usage on CDR3 conformation.

6:10 pm

Modeling with Rosetta to Guide Library Design of Antibodies

Daniel R. Woldring, PhD, Assistant Professor, Chemical Engineering & Materials Science, Michigan State University

Therapeutic antibodies have played a critical role in healthcare for over three decades, yet the rate of discovering novel antibodies remains outpaced by the need for treatment options. This is particularly true for challenging glycan targets such as tumor-associated carbohydrate antigens (TACAs). In this work, we use Rosetta software to optimize affinity and specificity among a large collection of natively-paired, immune-evolved antibodies.

Networking Reception in the Exhibit Hall with Poster Viewing6:40 pm

PEGS BOSTON COMMON: WOMEN IN SCIENCE MEET UP

6:50 pm

Women in Science Meet Up - IN-PERSON ONLY

Janice M. Reichert, PhD, COO, The Antibody Society

Rebecca A. Sendak, PhD, Head, Global Large Molecules Research Platform, Sanofi

The Women in Science Meet Up celebrates women trailblazers who are setting their own course in science. We invite women and men to come celebrate the successes of these women in breaking down barriers and inspiring future generations of female leaders. Come join fellow scientists and share your personal and professional journey.​

  • Who or What inspires you to explore a career in science?
  • What fuels your imagination and spirit when you’re faced with challenges?
  • What is your proudest moment?
  • What can each of us do to improve things further?​​

    Close of Engineering Antibodies Conference7:40 pm






    Register Now

    View By:


    Premier Sponsors

       FairJourneyBiologics Integral-Molecular_NEWLONZAOmniAb  Samsung_Biologics TWIST-Bioscience UnchainedLabs