Display of Biologics
Imagining the Next Generation of Biologics
5/11/2026 - May 12, 2026 ALL TIMES EDT
Phage, yeast, and mammalian displays have been instrumental for generating therapeutic molecules against a wide range of diseases. Cambridge Healthtech Institute's 28th annual Display of Biologics conference is the cornerstone of PEGS Boston and brings together leaders to showcase emerging technologies and approaches that are creating novel formats and functionalities. Understanding the role of machine learning and AI to build and navigate repertoires and libraries is at the forefront of the field and accelerating the pace of discovery.

Sunday, May 10

Recommended Pre-Conference Short Course

SC3: AI-Driven Predictive Preclinical Models: Rethinking the Role of Animal Testing

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

Monday, May 11

Registration and Morning Coffee

Organizer's Opening Remarks

NEW TECHNOLOGIES

Chairperson's Remarks

Photo of Andrew R.M. Bradbury, MD, PhD, CSO, Specifica, an IQVIA business , CSO , Specifica, Inc.
Andrew R.M. Bradbury, MD, PhD, CSO, Specifica, an IQVIA business , CSO , Specifica, Inc.

KEYNOTE PRESENTATION: History of Checkpoint Blockade Therapy for Cancer

Photo of Nils Lonberg, PhD, CEO, Tripeaks Therapeutics; Executive in Residence, Canaan Partners , CEO , Tripeaks Therapeutics
Nils Lonberg, PhD, CEO, Tripeaks Therapeutics; Executive in Residence, Canaan Partners , CEO , Tripeaks Therapeutics

The recent emergence of immunotherapy as a new pillar of cancer treatment is largely due to the success of immune checkpoint blockade (ICB) drugs, which block receptors such as, CTLA4 and PD1, and ligands, such as PDL1, involved in pathways that attenuate T cell activation to prevent, or reverse, acquired peripheral tolerance to tumor antigens. Improvement over the first generation of these drugs was frustratingly slow over the last decade; however, exciting new drugs and combinations are now beginning to appear. The history of ICB therapy and recent progress will be discussed.

New Insights into Antibody-Mediated Immunity

Photo of Arturo Casadevall, PhD, Professor & Chair, Molecular Microbiology & Immunology, Johns Hopkins University , Prof & Chair , Molecular Microbiology & Immunology , Johns Hopkins Univ
Arturo Casadevall, PhD, Professor & Chair, Molecular Microbiology & Immunology, Johns Hopkins University , Prof & Chair , Molecular Microbiology & Immunology , Johns Hopkins Univ

While many believe that antibody-mediated immunity is well understood, a variety of observations in recent decades implies the existence of major unresolved fundamental questions regarding the relationship between antibody structure and function. The observation that the constant region can affect specificity and affinity suggests an explanation for variable region restriction in antibody responses based on avoiding autoimmunity.  Antibodies have now been shown to modulate microbial function and to digest microbial antigens through catalytic activity. These findings suggest new ways to harness antibody-mediated immunity in the design of therapeutic immunoglobulins and protective vaccines.

One Antibody to Rule Them All: Consensus Antigen Design for Discovering Broadly Neutralizing Antibodies

Photo of Esperanza Rivera de Torre, PhD, Assistant Professor, Center for Antibody Technologies, Department of Bioengineering, Technical University of Denmark , Assistant Professor , Section for Biologics Engineering , Technical Univ of Denmark
Esperanza Rivera de Torre, PhD, Assistant Professor, Center for Antibody Technologies, Department of Bioengineering, Technical University of Denmark , Assistant Professor , Section for Biologics Engineering , Technical Univ of Denmark

Antibody specificity and affinity are critical determinants of effective target recognition and neutralization. Here, we present a consensus antigen design strategy aimed at discovering broadly neutralizing, cross-reactive antibodies. Our technology generates consensus antigens that represent the average sequence and three-dimensional structure of a family of structurally related proteins, thereby enriching for antibodies with inherent cross-reactivity against the original targets. We applied this approach to spider, scorpion, and snake venoms, where broad reactivity is essential for developing economically feasible, region-specific universal antivenoms. Using a naïve phage display library, we isolated antibodies capable of binding multiple venom toxins and demonstrated in vivo toxicity neutralization, highlighting the translational potential of this strategy.

Networking Coffee Break

KEYNOTE PRESENTATION: Fifty Years of Monoclonals: From Hybridomas to Next-Generation Antibody Therapeutics

Photo of Paul J. Carter, PhD, Genentech Fellow, Antibody Engineering, Genentech , Genentech Fellow , Antibody Engineering , Genentech
Paul J. Carter, PhD, Genentech Fellow, Antibody Engineering, Genentech , Genentech Fellow , Antibody Engineering , Genentech

The invention of hybridoma technology by Köhler and Milstein in 1975 ultimately led to over 200 antibody therapeutics, bringing benefit to millions of patients. This keynote will trace the remarkable rise of antibody therapeutics including bispecifics, antibody-drug conjugates, and CAR T cells. Future progress with antibody therapeutics will surely be accelerated by artificial intelligence, including multi-parameter optimization. On-going advances, including in conditional antigen binding and targeted tissue delivery, bode well for expanding the reach of antibody therapeutics into previously undruggable targets and diseases.

A Universal Monoallelic Human Leukocyte Antigen Class II Immunopeptidomic Platform for Defining Therapeutic Protein Immunogenicity Potential

Photo of Robert Siegel, PhD, Vice President, Laboratory for Experimental Medicine, Eli Lilly and Company , Vice President , Eli Lilly & Co
Robert Siegel, PhD, Vice President, Laboratory for Experimental Medicine, Eli Lilly and Company , Vice President , Eli Lilly & Co

Defining the exact sequences presented by human leudocyte antigen (HLA) Class II molecules is essential for understanding the immunogenicity potential of biotherapeutics. HLA heterozygosity complicates efforts to define both the precise sequences presented by various HLA alleles and the percentage of patients with potential to mount anti-drug responses that can negate clinical benefit and/or result in adverse events. This presentation will describe a diverse, robust, and reproducible monoallelic HLA-DRB1 system in professional antigen presenting cells capable of examining the immunogenic potential of any human IgG. A case study with adalimumab will be discussed.

Session Break

Session Break

NAVIGATING EPISTASIS IN PROTEIN ENGINEERING

Chairperson's Remarks

Photo of K. Dane Wittrup, PhD, C.P. Dubbs Professor, Chemical Engineering & Bioengineering, Massachusetts Institute of Technology , CP Dubbs Professor , Chemical Engineering & Bioengineering , Massachusetts Institute of Technology
K. Dane Wittrup, PhD, C.P. Dubbs Professor, Chemical Engineering & Bioengineering, Massachusetts Institute of Technology , CP Dubbs Professor , Chemical Engineering & Bioengineering , Massachusetts Institute of Technology

Advancements in Machine Learning-Assisted Protein Fitness Optimization

Photo of Jason Yang, PhD Candidate, Chemical Engineering, California Institute of Technology , Ph.D. Candidate , Caltech
Jason Yang, PhD Candidate, Chemical Engineering, California Institute of Technology , Ph.D. Candidate , Caltech

Directed evolution is a powerful tool to optimize protein fitness for a specific application, but it can be inefficient when mutations exhibit non-additive, or epistatic, behavior. Machine learning (ML) methods have the potential to address this limitation by learning from assay-labeled data and suggesting ideal combinations of mutations to navigate the protein fitness landscape more efficiently. Here, I present frameworks for iterative, ML-assisted protein optimization: Active Learning-assisted Directed Evolution (ALDE) and Steered Generation for Protein Optimization (SGPO). Besides showing strong in silico performance, three rounds of wet-lab experimentation with ALDE enabled rapid optimization of five epistatic residues in the active site of an enzyme, yielding an ideal variant with a non-obvious (non-additive) combination of mutants. Overall, these ML-assisted methods are practical and broadly applicable strategies to unlock improved protein engineering outcomes.

The Cause and Consequence of Epistasis in Protein Evolution

Photo of Nobuhiko Tokuriki, PhD, Professor, Michael Smith Laboratories, University of British Columbia , Assoc Prof , Biochemistry & Molecular Biology , Univ of British Columbia
Nobuhiko Tokuriki, PhD, Professor, Michael Smith Laboratories, University of British Columbia , Assoc Prof , Biochemistry & Molecular Biology , Univ of British Columbia

Over the last decades of research on protein evolution and engineering, we have recognized that the success of protein engineering campaigns is overly depending on the systems. What are molecular determinants to dictate the evolution of new protein functions? How can we choose the right strategies to engineer new proteins?I will discuss key molecular properties that can be associated with evolvability of proteins, the ability of proteins to promptly evolve a new function. Especially, I will discuss the causes and consequences of mutational epistasis, interactions between mutational effects that affect the pathways and outcomes of evolution.

AI-Generated Protein-Function Prediction with Therapeutic Applications

Photo of Lucy J. Colwell, PhD, Research Scientist, Google UK Ltd. , Research Scientist , Google
Lucy J. Colwell, PhD, Research Scientist, Google UK Ltd. , Research Scientist , Google

Networking Coffee & Refreshment Break

Transition to Plenary Keynote Session

PLENARY KEYNOTE

Plenary Keynote Introduction

Photo of Mahiuddin Ahmed, PhD, President and CSO, VITRUVIAE , President and CSO , VITRUVIAE
Mahiuddin Ahmed, PhD, President and CSO, VITRUVIAE , President and CSO , VITRUVIAE

CARs 2026: New Models and New Runways

Photo of Michel Sadelain, MD, PhD, Director, Columbia University Initiative in Cell Engineering and Therapy (CICET); Director, Cell Therapy Initiative, Herbert Irving Comprehensive Cancer Center; Professor of Medicine, Columbia University Irving Medical Center , Stephen & Barbara Friedman Chair & Director , Center for Cell Engineering , Memorial Sloan Kettering Cancer Centre
Michel Sadelain, MD, PhD, Director, Columbia University Initiative in Cell Engineering and Therapy (CICET); Director, Cell Therapy Initiative, Herbert Irving Comprehensive Cancer Center; Professor of Medicine, Columbia University Irving Medical Center , Stephen & Barbara Friedman Chair & Director , Center for Cell Engineering , Memorial Sloan Kettering Cancer Centre

T cell engineering holds great promise for the treatment of cancers and other pathologies. The original chimeric antigen receptor (CAR) prototypes targeting CD19 are now giving way to further refined receptors endowed with greater sensitivity and combinatorial possibilities. Emerging new targets and engineering tools augur favorably for broadening the use of CAR therapies.

YOUNG SCIENTIST KEYNOTE

Deep Learning-Based Binder Design to Probe Biology

Photo of Martin Pacesa, PhD, Assistant Professor, Pharmacology, University of Zurich , Assistant Professor , Department of Pharmacology , University of Zurich
Martin Pacesa, PhD, Assistant Professor, Pharmacology, University of Zurich , Assistant Professor , Department of Pharmacology , University of Zurich

Protein-protein interactions are central to biology and drug discovery, yet traditional antibody generation is slow and costly. BindCraft is an open-source, automated computational pipeline for de novo protein binder design that routinely yields nanomolar binders with 10-100% experimental success, without high-throughput screening or maturation. We illustrate applications to peptides, cell-surface receptors, allergens, and gene editors, and outline how deep learning workflows can accelerate next-generation therapeutics, diagnostics, and bioprocessing.


  • What are the advantages/drawbacks of minibinders?
  • Are there "unbindable" protein sites?
  • Are natural amino acid building blocks enough for drug development?
  • What therapeutic properties should deep learning models account for?

Welcome Reception in the Exhibit Hall with Poster Viewing

Close of Day

Tuesday, May 12

Registration and Morning Coffee

ENGINEERING FOR RADIOPHARM AND CHEMOTHERAPY

Chairperson's Remarks

Photo of Jennifer R. Cochran, PhD, Senior Associate Vice Provost for Research and Macovski Professor of Bioengineering, Stanford University , Shriram Chair & Professor , Bioengineering & Chemical Engineering , Stanford University
Jennifer R. Cochran, PhD, Senior Associate Vice Provost for Research and Macovski Professor of Bioengineering, Stanford University , Shriram Chair & Professor , Bioengineering & Chemical Engineering , Stanford University

Engineering Cyclotides as Orally Bioavailable Inflammatory Cytokine Antagonists

Photo of K. Dane Wittrup, PhD, C.P. Dubbs Professor, Chemical Engineering & Bioengineering, Massachusetts Institute of Technology , CP Dubbs Professor , Chemical Engineering & Bioengineering , Massachusetts Institute of Technology
K. Dane Wittrup, PhD, C.P. Dubbs Professor, Chemical Engineering & Bioengineering, Massachusetts Institute of Technology , CP Dubbs Professor , Chemical Engineering & Bioengineering , Massachusetts Institute of Technology

The first naturally occurring cyclotide, a circularized disulfide-rich protein, was discovered as the active ingredient of a folk medicine brewed as a tea. These small cyclic proteins exhibit remarkable oral bioavailability. Combining this property with antibody-like recognition of arbitrary targets would enable new oral therapeutic modalities for interrupting inflammatory cytokine cascades. We will present our progress in designing pre-immune cyclotide repertoires, minimizing polyspecificity, and accelerating lead optimization.

Radiolabeled Camelids against FAP Discovered with Immunization and Phage Display

Photo of Sam Massa, PhD, Head, Protein R&D and CMC, Precirix , Head of Protein R&D and CMC , Precirix
Sam Massa, PhD, Head, Protein R&D and CMC, Precirix , Head of Protein R&D and CMC , Precirix

Single domain antibodies (sdAbs or VHHs) are the smallest antibody-derived fragments with beneficial pharmacokinetic properties for molecular imaging and targeted radionuclide therapy. This talk will focus on the generation, selection, and characterization of sdAb 4AH29 targeting FAP. After conjugation to DOTA, the resulting drug products [68Ga]Ga-DOTA-4AH29 and [225Ac]Ac-DOTA-4AH29 showed the potential of radiolabeled sdAb 4AH29 as a radiotheranostic agent for FAP-positive cancers.

Discovery and Development of ECM-Specific Nanobodies for Targeted Radioligand Therapy

Photo of Noor Jailkhani, PhD, CEO & Co-Founder, Matrisome Bio , CEO & Co-founder , Matrisome Bio
Noor Jailkhani, PhD, CEO & Co-Founder, Matrisome Bio , CEO & Co-founder , Matrisome Bio

Metastatic solid tumors represent a major unmet need, underscoring the limitations of therapies that focus solely on cancer cells. At Matrisome Bio, we are targeting the disease-associated extracellular matrix or ECM within tumors and metastases, which offers a compelling new therapeutic avenue. This talk will highlight the discovery of high-affinity nanobodies (via phage-display) against ECM proteins and their subsequent engineering as radioisotope carriers, enabling the precise delivery of highly differentiated radioligand therapies.

Coffee Break in the Exhibit Hall with Poster Viewing

EVERYTHING BUT VANILLA IgGs: AN OVERVIEW OF THE RISING THERAPEUTIC POTENTIAL OF PEPTIDES AND VHH

Chairperson's Remarks

Photo of Maria Groves, PhD, Senior Director, AstraZeneca , Senior Director , Biologics Engineering R&D , AstraZeneca
Maria Groves, PhD, Senior Director, AstraZeneca , Senior Director , Biologics Engineering R&D , AstraZeneca

From Libraries to Leads: Expanding the Biologics Horizon with de novo Peptide Discovery

Photo of Thomas Murray, PhD, Director, Biologics Engineering, AstraZeneca , Director , Biologics Engineering , AstraZeneca
Thomas Murray, PhD, Director, Biologics Engineering, AstraZeneca , Director , Biologics Engineering , AstraZeneca

Anti-Von Willebrand Factor NANOBODY Compound Cablivi Story from Conception to Commercialization

Photo of Beneditke Serruys, PhD, Global Head Biologics Innovation, Large Molecule Research Platform, Sanofi , Head Biologics Innovation , Pharmacology , Sanofi
Beneditke Serruys, PhD, Global Head Biologics Innovation, Large Molecule Research Platform, Sanofi , Head Biologics Innovation , Pharmacology , Sanofi

Caplacizumab, a revolutionary anti-von Willebrand Factor NANOBODY compound, emerged from two decades of innovation at Sanofi. This presentation traces the entire story from discovery through clinical development to commercial success, chronicling the complete development journey from llama immunization in 2003 to first-in-class therapeutic approval, starting with EMA (2018) and FDA (2019). Clinical development demonstrated the successful translation of this innovative biologic into an effective therapy for acquired thrombotic thrombocytopenic purpura (aTTP). As the first approved NANOBODY therapeutic, caplacizumab's mechanism of targeting pathophysiological platelet adhesion has established a new treatment paradigm for this rare blood disorder.

Session Break

Close of Display of Biologics Conference

Recommended Dinner Short Course

SC7: Targeting the Target: Aligning Target and Biologic Format Biology to Achieve Desired Outcomes

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


For more details on the conference, please contact:

Christina Lingham

Executive Director, Conferences and Fellow

Cambridge Healthtech Institute

Phone: 508-813-7570

Email: clingham@healthtech.com

 

For sponsorship information, please contact:

Companies A-K

Jason Gerardi

Sr. Manager, Business Development

Cambridge Healthtech Institute

Phone: 781-972-5452

Email: jgerardi@healthtech.com

 

Companies L-Z

Ashley Parsons

Manager, Business Development

Cambridge Healthtech Institute

Phone: 781-972-1340

Email: ashleyparsons@healthtech.com


Register

View By:


Premier Sponsors

FairJourneyBiologics GenScript-CRO Integral-Molecular_NEW   Nona_Biosciences_NEW