Cambridge Healthtech Institute's 19th Annual

Difficult-to-Express Proteins

Overcoming Expression, Purification, and Production of Challenging Proteins

May 13 - 14, 2024 ALL TIMES EST

Structural and functional characterization of proteins as well as the design of targeted drugs heavily rely on recombinant protein expression and purification. But what makes certain recombinant proteins challenging to express, produce, and purify? Numerous obstacles come into play, encompassing factors like intricate folding patterns, potential host toxicity, and purification complexities, among others. The 19th Annual Difficult-to-Express Proteins conference, hosted by the Cambridge Healthtech Institute, delves into the myriad of difficulties faced and solutions provided by researchers in their pursuit of achieving higher-yield production for these "difficult-to-express" proteins (DTEPs). This event sheds light on the innovative strategies and cutting-edge technologies that have demonstrated effectiveness in surmounting these obstacles to unlock the potential of these unique and engineered proteins.

Sunday, May 12

Main Conference Registration1:00 pm

Recommended Pre-Conference Short Course2:00 pm

SC2: How to Use and Improve Microbial Expression Systems for Recombinant Protein Production

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

Monday, May 13

Registration and Morning Coffee7:00 am

EXPRESSION AND PRODUCTION OF MEMBRANE PROTEINS

8:20 am

Chairperson's Opening Remarks

Ethan Dunn, Manager, Protein Sciences, Moderna, Inc.

8:30 am

Development and Characterization of Functional Antibodies Targeting NMDA Receptors

Nami Tajima, PhD, Assistant Professor, Physiology & Biophysics, Case Western Reserve University

N-methyl-D-aspartate (NMDA) receptors are ligand-gated ion channels which mediate the excitatory neurotransmission, and are associated with cancer and brain diseases, including epilepsy, schizophrenia, and Alzheimer’s disease. Thus, targeting NMDA receptors is a promising strategy for treating the diseases. Due to the instability and complexity of those large multi-oligomeric membrane protein complexes expressed in eukaryotic species, it remains challenging to yield properly assembled proteins. Here, I present the developed method to improve protein production, electrophysiological and cryo-EM studies of NMDA receptor complexes, and functional antibodies. Our work contributes to a better understanding of molecular mechanisms and strategies to target NMDA receptors.

9:00 am

Production of a Challenging Human Single-Pass ER Membrane Protein: Expression in Multiple Hosts and Screening of Novel Fusion Constructs for Solubility in Expi293

Ethan Dunn, Manager, Protein Sciences, Moderna, Inc.

Moderna is developing multiple mRNA vaccines and therapeutics. Since target protein expression from mRNA drugs is required for their activity, Moderna needs to produce lab-scale quantities of targeted proteins for various studies. Here, we expressed a single-pass transmembrane protein in multiple hosts and screened over 30 fusion constructs for solubility in Expi293, enabling production of the protein in soluble form with successful cleavage of the fusion partners/purification tags.

9:30 am

Rapid Production of Coatomer WD40 Domains: Lessons from COVID-19 Research

S. Saif Hasan, PhD, Assistant Professor, Biochemistry & Molecular Biology, Center for Biomolecular Therapeutics, University of Maryland

The endoplasmic reticulum (ER) is where approximately one-third of the proteome, including hormones, growth factors, and antibodies, is synthesized. The secretory export of these proteins risks leakage of the ER-resident proteome, which causes cell stress and interferes with biosynthesis. We have developed cost-effective strategies for enhanced prokaryotic and eukaryotic production of the WD40 domains of the COPI complex, crucial for retaining the ER-resident proteome during secretory trafficking. These strategies have broad applications since WD40 domains play key roles in the secretory trafficking of medically significant proteins, such as the SARS-CoV-2 spike protein produced from genetic vaccines.

10:00 am A Scalable Protein Production Technology to Accelerate Biotherapeutic Development

Ricarda Finnern, PhD, Chief Scientific Officer, LenioBio

Our presentation will showcase the capabilities of the ALiCE® cell-free protein development platform for the development of complex proteins from discovery to lead optimization. Detailed case studies showcase ALiCE®’s potential to accelerate vaccine (VLPs) and biotherapeutic development (monoclonal antibodies and multispecifics). ALiCE® is the go-to platform from screening to protein optimization and beyond.

Networking Coffee Break10:30 am

11:00 am

Systematic Structural Evaluation of Polycystin Variants by CryoEM

Orhi Esarte Palomero, PhD, Postdoctoral Fellow, Pharmacology, Northwestern University

Mutations in polycystin ion channel subunits cause Polycystic Kidney Disease (PKD), a genetic disorder that afflicts 1:1000 individuals worldwide with perfuse kidney cysts. We developed a mammalian protein expression and purification pipeline to produce recombinant polycystin ion channel variants. We applied this methodology in combination with CryoEM to evaluate the structural changes caused by missense PKD causing mutations in specific domains of the PKD2 polycystin homotetrameric channel assembly.

11:30 am

Reigning in Therapeutic Protein Expression with Pichia pastoris

Adam Nylen, Senior Scientist, Merck & Co Inc

Pichia pastoris is a proven expression host that can be utilized for the production of difficult-to-express proteins. The GlycoFi platform humanized the Pichia glycosylation pathway which allows it to express glycoproteins with a human-like glycan profile. This platform gives the opportunity to select for the optimal glycan structure through genetic knockouts and over-expressions, while simultaneously tackling the production of many challenging-to-express proteins.

Session Break12:00 pm

12:05 pm Luncheon Presentation I:Efficient Therapeutic Development Using The Pfenex Expression Technology® Platform

Elizabeth Orchard, PhD, Director of Process Development, Primrose Bio

The Pfenex Expression Technology® is a commercially validated P. fluorescens based platform used for recombinant protein production. Case studies are discussed demonstrating how the Pfenex toolbox of genetic elements and host strains enabled rapid exploration of expression strategies for challenging protein scaffolds, including proteins engineered for site-specific chemical modification to enable the development of products such as antibody drug conjugates for use as human therapeutics.

12:35 pm LUNCHEON PRESENTATION:Optimizing Protein Expression: Leveraging a Robust CLD Platform and Scalable Process Development Framework

Sojeong Lee, PhD, Lead Scientist, Associate Director of Cell Line Development, Cell Line Development, Samsung Biologics

Biologics development organizations face the challenge of accelerating development while enhancing quality and optimizing processes. A robust CLD process is a pivotal technology driver. Leveraging this robust cell line through scalable process development is crucial for success. Samsung Biologics offers proprietary platforms like S-CHOice® and optimized tech transfer processes, providing a seamless workflow that expedites preclinical development with ensured quality and efficiency.

Session Break1:05 pm

EXPANDING CELL FREE EXPRESSION

1:10 pm

Chairperson's Remarks

Frank Bernhard, PhD, Lab Leader, Institute of Biophysical Chemistry, Goethe University

1:15 pm

FEATURED YOUNG SCIENTIST: Harnessing Cell-free Protein Synthesis to Synthesize and Repurpose Viral Fusion Machinery

Ekaterina Selivanovitch, PhD, Postdoctoral Researcher, Cornell Smith School of Chemical and Biomolecular Engineering, Cornell University

Viruses present one of the most efficient mechanisms for intracellular cargo (i.e. viral genome) delivery. Harnessing the efficiency of this ability would drastically improve cellular uptake of therapeutic and bioactive cargo, currently a major obstacle in pharmaceutical scientific communities. In this work, viral proteins are repurposed for delivering user-defined cargo to cells containing the appropriate receptors. Additionally, by developing adapted cell-free protein synthesis techniques, we construct user-defined proteoliposomes that can be either used towards furthering our understanding of virus ligand-host cell interactions or repurposed for targeted therapies.

1:45 pm

Cell-Free Expression Systems: Methods of Production and Use

Matthew W. Lux, Research Biologist, BioSciences, Edgewood Chemical Biological Center

Cell-free expression systems reconstitute the core cellular functionalities of transcription, translation, and metabolism outside of the confines of the cell. This format enables experimentation simply by adding DNA or other components to a reaction, offering rapid experimental cycles compared to cells and higher control over conditions. Here we describe our work to leverage the high throughput capacity of this technology towards biomanufacturing and sensing applications.

2:15 pm

Molecular Structures of GPCR Complexes Obtained by Rationalized Cell-Free Production Pipelines

Frank Bernhard, PhD, Lab Leader, Institute of Biophysical Chemistry, Goethe University

We present a fast and cost-effective modular cell-free expression platform for the generation of high-resolution cryo-EM structures of GPCR/G protein complexes. We show with the full-length human ß1-adrenergic receptor and with the histamine-2 receptor the first molecular structures of GPCR/G protein complexes obtained from cell-free generated samples. Key advantages are (i) direct cotranslational GPCR insertion into tailored membranes of nanoparticles, (ii) no contacts to detergents, (iii) synthesis of full-length, non-engineered GPCRs without deletions or fusions, (iv) suitable for a wide array of GPCRs, oGPCRs, and other membrane proteins, and (v) free choice of ligands and lipid environment.

2:45 pm

A Cell-Free High Throughput Protein Production Platform for Evaluating AI-designed Antibody-based Therapeutics’ Specificity Against Thousands of Targets Simultaneously

Lydia Gushgari, PhD, Chief of Staff, SPOC an INanoBio Co

Artificial intelligence-enabled drug design produces a large number of possible therapeutic candidates that need to be down-selected for biological validation. High-throughput antibody specificity (on- and off-target) assays are often restricted to a limited subset of specific toxicity targets and lack customizability, which is even more critical for antibody-drug conjugates (ADCs) that may interact with intracellular targets. Sensor-integrated proteome on chip (SPOC) is a novel proteomic biosensor that provides qualitative, quantitative, and kinetic binding affinity data simultaneously against thousands of extracellular domains and intracellular protein targets on a single 1.5-sq cm surface plasmon resonance (SPR) biosensor chip. Real-time kinetic SPR analysis provides insight into transient interactions to identify off-targets earlier in the discovery pipeline for lead candidate identification, and to permit design of tunable on- and off-rates for binding interactions.

Networking Refreshment Break3:15 pm

Transition to Plenary Keynote Session4:15 pm

PLENARY KEYNOTE SESSION

4:25 pm

Plenary Keynote Introduction

Laszlo G. Radvanyi, PhD, President & Scientific Director, Ontario Institute for Cancer Research

4:35 pm

Driving New CAR T Cells

Marcela V. Maus, MD, PhD, Associate Professor, Medicine; Director, Cellular Immunotherapy, Massachusetts General Hospital

We will talk about various roads and challenges in driving new CAR T cells toward the clinic, and learnings from clinical experience.

YOUNG SCIENTIST KEYNOTE

5:20 pm

High-Throughput Discovery of Protein Folding Stability and Dynamics

Gabriel J. Rocklin, PhD, Assistant Professor, Pharmacology, Northwestern University

Every protein has its own conformational energy landscape that governs its folding stability and dynamics. These varied landscapes are rarely predictable in protein engineering but strongly influence function, aggregation, immunogenicity, and more. Our lab develops new large-scale methods to measure stability and dynamics. I will share lessons from stability measurements of >750,000 protein domains and dynamics measurements of >5,000 domains, highlighting the potential to rationally engineer stability and dynamics.

Welcome Reception in the Exhibit Hall with Poster Viewing6:05 pm

YOUNG SCIENTIST MEET-UP

7:00 pm

Facilitators of Young Scientist Meet Up: IN-PERSON ONLY

Orhi Esarte Palomero, PhD, Postdoctoral Fellow, Pharmacology, Northwestern University

Alexandros Karyolaimos, PhD, Researcher, Department of Biochemistry & Biophysics, Stockholm University

Shakiba Nikfarjam, PhD, Postdoc, Lawrence Livermore National Lab

Network, Inspire Others and Connect

The young scientist meet-up is an opportunity for scientists entering the field to develop connections across institutions, and for established leaders to come build relationships with the next generation of scientists. The meet-up will pave the way for mentorships, professional opportunities, and scientific discovery.

  • Get to know fellow peers and colleagues
  • Make connections and network with other institutions
  • Inspire others and be inspired!​​​

Close of Day7:30 pm

Tuesday, May 14

Registration and Morning Coffee7:30 am

APPLYING ANALYTICS TO OPTIMIZE EXPRESSION AND SCREENING

7:50 am

Chairperson's Remarks

Bjørn Voldborg, MSc, Head, National Biologics Facility, DTU Bioengineering, Technical University of Denmark

7:55 am

FEATURED PRESENTATION: Addressing Difficult-to-Produce Protein Challenges in Discovery Research

Erik Vernet, PhD, Senior Director, External Partnerships, Digital Science & Innovation, Novo Nordisk

High-quality biological reagents are a prerequisite for pharmacological research. I will discuss trends from thousands of expression constructs screened in mammalian and bacterial hosts, as well as case studies utilizing stable cell line generation and choice of fusion protein for higher yield and quality of difficult-to-produce proteins.

8:20 am

Utilizing Deep Learning Protein Representations to Predict Recombinant Expression

Sébastien Ouellet, MSc, Machine Learning Developer, Individual Researcher

Cysteine-dense peptides (CDPs) are an attractive pharmaceutical scaffold that display extreme biochemical properties, low immunogenicity, and the ability to bind targets with high affinity and selectivity. Identifying CDPs that can be expressed in mammalian cells is crucial in predicting their compatibility with gene therapy and mRNA therapy. We developed CysPresso—a novel machine learning approach combining time series-inspired methods and embeddings—that predicts recombinant expression of CDPs based on their primary sequence. Our study showcases the applicability of deep learning-based protein representations, such as those provided by AlphaFold2, in tasks beyond structure prediction.

8:45 am

Using Streamlined HTS, High Density Analysis and Data Mining to Optimize DNA Construct Design

Jennifer M Shipman, Associate Principal Scientist, Protein & Structural Chemistry, Merck & Co Inc

The inherent challenge of protein structural enablement is to obtain the target protein of sufficient quality and quantity to support various structural techniques. Our goal is to compile large datasets from a semi-automated, small-scale, data-rich characterization workflow into a minable database, allowing us to build correlations between construct designs and favorable outcomes and ultimately to predict successful designs. This workflow leads to fewer design iterations, greater probabilities of success and reduction in resource demand.

Coffee Break in the Exhibit Hall with Poster Viewing9:10 am

PLENARY KEYNOTE SESSION

10:00 amTransition to Plenary Keynote Session
10:10 am

Plenary Keynote Introduction 

Jennifer R. Cochran, PhD, Senior Associate Vice Provost for Research, Macovski Professor of Bioengineering, Stanford University

10:15 am

Laboratory Evolution of Genome Editing Proteins for Precise Gene Correction and Targeted Gene Integration in Mammalian Cells 

David R. Liu, PhD, Richard Merkin Professor and Director, Merkin Institute of Transformative Technologies in Healthcare; Core Institute Member and Vice-Chair of the Faculty, Broad Institute; Director, Chemical Biology and Therapeutic Sciences Program; Investigator, Howard Hughes Medical Institute; Thomas Dudley Cabot Professor of the Natural Sciences and Professor of Chemistry and Chemical Biology, Harvard University

In this lecture I will describe the use of protein evolution and protein engineering to develop precision genome editing technologies. These technologies include base editors, prime editors, recombinases, and CRISPR-associated transposases (CASTs). Base editors and prime editors have been used by many laboratories around the world to correct pathogenic mutations, resulting in ex vivo and in vivo one-time treatments that rescue disease phenotypes in many animal models of devastating genetic disorders. At least nine base editing clinical trials have begun, with positive clinical readouts from at least three of these trials, and the first prime editing clinical trial was recently cleared by FDA to begin in the U.S. I will also describe the use of phage-assisted continuous evolution (PACE) to evolve prime editors, recombinases, and CASTs to enable efficient targeted gene-sized integration in human cells, addressing a longstanding challenge in the genome editing field. These engineered and evolved proteins enable precise target gene correction, disruption, or insertion in a wide range of organisms with broad implications for the life sciences and therapeutics.

Celebrating 20 Years in the Exhibit Hall with Poster Viewing11:00 am

OPTIMIZING CHO CELLS FOR PRODUCTION OF CHALLENGING BIOTHERAPEUTICS

11:45 am

Establishment of O-Glycoengineered CHO Cell Line Platform for Producing Better Glycoprotein Drugs

Tongzhong Ju, MD, PhD, Principal Investigator, FDA CDER

While N-glycosylation is a known critical quality attribute (CQA), the impact of O-glycosylation on therapeutic protein quality remains elusive. My presentation describes the establishment of an O-glycoengineered CHO cell line platform to modulate O-glycosylation and provides evidence that O-glycosylation of therapeutic glycoproteins, such as etanercept, impacts the physicochemical properties and biological activity. This O-glycoengineered production platform is valuable to identify O-glycosylation as a CQA for glycoprotein drugs to improve quality.

12:10 pm

Screening Biotherapeutics with Bespoke Glycoforms Using Glycoengineered CHO Cells

Bjørn Voldborg, MSc, Head, National Biologics Facility, DTU Bioengineering, Technical University of Denmark

Through gene-specific cell engineering, we have developed a panel of glycoengineered CHO cells, (geCHO) enabling rapid production of bespoke glycoforms of therapeutic proteins. With this platform, we have produced and screened multiple therapeutic drug candidates (vaccines and enzymes) in vitro and in vivo to determine their optimal glycoform with respect to antigenicity, activity, and stability, etc.

12:35 pm

Repressing Expression of Difficult-to-Express Recombinant Proteins during the Selection Process Increases Productivity of CHO Stable Pools

Jean-Sebastien Maltais, PhD, Research Officer, Human Health Therapeutics, National Research Council Canada

Many next-generation therapeutics remain intrinsically challenging to produce in CHO cells. We exploited a cumate-inducible CHO platform allowing reduced expression of various classes of r-proteins during selection of stable pools. Fed-batch productions showed that pools generated without cumate (OFF-pools) were significantly more productive. Cell viability was lower and pool recovery was delayed during selection of ON-pools (mimicking constitutive expression), suggesting that high producers were likely lost or overgrown by faster-growing, low-producing cells. Using an inducible system to minimize r-protein expression during pool selection can contribute to reducing cellular stresses, including ER stress and metabolic burden, leading to improved productivity.

1:00 pm PROTiQ: Machine Learning Tools to Mitigate Risks in Traditional & Hypothetical Protein Development Workflows

Carter Mitchell, CSO, Kemp Proteins

ProtIQ is a machine learning-based suite of tools that aggregates protein target information from a variety of sources and annotates and analyzes proteins, when information is lacking. The tool was developed to address the breadth of global hypothetical proteome. Through structural modeling and protein language transformer models, protein sequences are inputted to derive protein design and process development in silico to improve the likelihood of success in the protein development cycle.

1:15 pm CHOnamite® and GlycoExpress®: Versatile expression platforms to reach biopharma productivity and quality goals

Lars Stöckl, PhD, Managing Director, FyoniBio GmbH

During this presentation, we will duscuss: versatile expression platform for different quality needs of biopharmaceutical: CHOnamite® vs GlycoExpress®; special emphasis on glycosylation from different host cell systems of crucial importance for biosimilar development; and increase of productivity and quality (e.g., glycosylation) by process optimization. Case studies will also be presented.

Session Break1:30 pm

1:40 pm Luncheon Presentation I:Innovative Platforms for Producing Mini Proteins & T-Cell Related Therapeutic Targets

Jiansheng Wu, PhD, Vice President, Protein Sciences, WuXi Biologics

Mini proteins and T-cell-related proteins are gaining traction as new modalities of biologic drug. Our Mini Protein Line innovates beyond traditional E. coli methods, using high-titer CHO expression for enhanced mammalian expression and extra-low endotoxin level. Our T Cell Mate efficiently produces challenging T-cell-related proteins like sTCR, TCR-Ab fusions, SCT, and RF-pMHC, ensuring high throughput and yields, critical for therapeutic protein advancement.

2:10 pm LUNCHEON PRESENTATION:Microbial Production Platform Suitable for Long-Chain Peptides and VHHs

Yumi Nagase, MS, Lead Researcher, Biopharma Solutions Group, Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc.

Ajinomoto Bio-Pharma Services as a fully integrated CDMO offers a broad range of innovative platform technologies and end-to-end solutions for biopharmaceutical development and manufacturing. In this presentation, we will introduce our CDMO capabilities and highlight the Corynex® protein and peptide expression platform technology, including its application towards the high-quality manufacture of GLP-1 related peptides, VHHs and ancillary materials to produce pharmaceuticals.

Close of Difficult-to-Express Proteins Conference2:40 pm

Recommended Dinner Short Course6:30 pm

SC7: The Use and Optimization of Eukaryotic Expression Systems to Support Therapeutic Generation and Structural Biology 

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






Register Now

View By:

Premier Sponsors

FairJourneyBiologics GenScript-CRO Integral-Molecular_NEW  OmniAbUnchainedLabs