Cambridge Healthtech Institute's 16th Annual

Difficult-to-Express Proteins

Overcoming Production Challenges

May 11 - 12, 2021 ALL TIMES EDT

What makes a protein ‘difficult’ to produce? CHI’s 16th Annual Difficult-to-Express Proteins conference examines the challenges encountered by researchers when striving for high-yield production of “difficult-to-express” proteins (DTEPs), and the strategies and technologies that have proven successful in overcoming those challenges. The Difficult-to-Express Proteins conference provides the latest developments in improving yield for DTEPs through case studies and breakthrough data.

Tuesday, May 11

OVERCOMING EXPRESSION CHALLENGES

Anne Skaja Robinson, PhD, Professor & Head, Chemical Engineering, Carnegie Mellon University

The adenosine A2A receptor (A2AR) shows exceptional yields in all expression hosts, unlike the closely related G protein-coupled receptors A1R and A3R. By swapping the cytoplasmic C-terminus of A2AR we were able to create chimeric A3R and A1R proteins with increased yields that retain activity and native-like G-protein coupling. This strategy of utilizing chimeric receptor variants thus provides an exciting opportunity to improve active protein expression for “difficult-to-express” receptors.

9:20 am

A Rational Approach to Designing Cell-Free Synthesis-Based Biomolecule Platform Screening Processes

Beatrice Melinek, PhD, Bioprocess Engineer, University College London

Advances in Cell-Free Protein Synthesis (CFPS) offer the prospect of industrially relevant screening and even production processes, with the advantage of greater speed, control over process environment, integration into high-throughput systems and improved consistency. We present studies of how biochemical engineering approaches can be used to improve the titre and quality of a virus-like particle (VLP) protein product, by manipulation of process environment parameters and plasmid design.

9:40 am

Standard mAb ≠ Easy-to-Express: How the Production of Monoclonal Antibodies Can Turn Into a Difficult-to-Express Challenge

Valerie Schmieder, PostDoc, Cell Line Development, Bioprocess Development, Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG

Although monoclonal antibodies are considered as standard molecules in biotherapeutic industry, some of those drugs are still difficult-to-express molecules. Here, we are highlighting the development of a difficult-to-express monoclonal antibody and its challenges for CHO cell line generation. Therefore, protein engineering and modulation of vector architecture are presented as approaches to improve product yields. Moreover, a new cell line development concept in CHO going beyond random transgene integration is highlighted to further boost the expression of a difficult-to-express molecule.

10:00 am

Improved Two-Stage Protein Production in Engineered E. Coli

Michael D. Lynch, PhD, Assistant Professor, Biomedical Engineering & Chemistry, Duke University

E. coli, the work horse host for protein production still faces many challenges in protein expression.  Therefore, we have engineered strains for the standardized 2-stage expression of heterologous proteins (in stationary phase). 2-stage production enables the utilization of 2-stage dynamic control, wherein the cellular state can be modified  (beyond conditions compatible with growth) to enable optimal protein expression, even with difficult to express proteins. Level of key proteases, chaperones and even the reduction potential of the cytoplasm can be dynamically controlled.  We demonstrate the broad applicability of this approach to express a variety of protein targets of commercial interest.

Renee Tobias, Director, CLD Product Management, Marketing, Berkeley Lights

In the new era of complex modalities and COVID-19 pandemic, speed, capacity, efficiency, and robustness are more vital than ever to a successful biotherapeutics development program. Yet CHO cell line selection still represents a painful, costly bottleneck - requiring weeks waiting for cells to expand and processing of hundreds of well plates. Learn how Opto™ CLD workflow on the Beacon® system integrates cell enrichment, cloning, culture, screening, and selection into a single, automated microscale process that generates the highest titer clones in just a few days. Case studies will highlight how users are accelerating and de-risking their path to IND with capacity for up to 50 campaigns per year, multiple molecules per campaign, and FDA-accepted monoclonality assurance to support regulatory submissions.

10:50 am LIVE PANEL DISCUSSION:

Overcoming Expression Challenges

Panel Moderator:
Anne Skaja Robinson, PhD, Professor & Head, Chemical Engineering, Carnegie Mellon University
Panelists:
Michael D. Lynch, PhD, Assistant Professor, Biomedical Engineering & Chemistry, Duke University
Beatrice Melinek, PhD, Bioprocess Engineer, University College London
Valerie Schmieder, PostDoc, Cell Line Development, Bioprocess Development, Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG
Renee Tobias, Director, CLD Product Management, Marketing, Berkeley Lights
11:10 am Session Break - View Our Virtual Exhibit Hall

PLENARY KEYNOTE ADDRESS

11:25 am

Plenary Keynote Introduction

Jennifer R. Cochran, PhD, Shriram Chair & Professor, Bioengineering & Chemical Engineering, Stanford University
11:30 am

The Coming of Age of de Novo Protein Design

David A. Baker, PhD, Henrietta & Aubrey David Endowed Professor, Biochemistry, University of Washington

Proteins mediate the critical processes of life and beautifully solve the challenges faced during the evolution of modern organisms. Our goal is to design a new generation of proteins that address current day problems not faced during evolution. In contrast to traditional protein engineering efforts, which have focused on modifying naturally occurring proteins, we design new proteins from scratch based on Anfinsen’s principle that proteins fold to their global free energy minimum. We compute amino acid sequences predicted to fold into proteins with new structures and functions, produce synthetic genes encoding these sequences, and characterize them experimentally. I will describe the de novo design of fluorescent proteins, membrane penetrating macrocycles, transmembrane protein channels, allosteric proteins that carry out logic operations, and self-assembling nanomaterials and polyhedra. I will also discuss the application of these methods to COVID-19 challenges.

12:00 pm LIVE:

Q&A with Audience

Panel Moderator:
Jennifer R. Cochran, PhD, Shriram Chair & Professor, Bioengineering & Chemical Engineering, Stanford University
Panelist:
David A. Baker, PhD, Henrietta & Aubrey David Endowed Professor, Biochemistry, University of Washington
12:10 pm Session Break - View Our Virtual Exhibit Hall

Breakout Discussions

12:20 pm Problem Solving Discussions

Join your colleagues and fellow delegates for a focused, informal discussion moderated by a member of our speaking faculty. A small group format allows participants to meet potential collaborators, share examples from their own work and discuss ideas with peers. See website for a full list of topics.

TABLE: Ranking Product Attributes to Select an Appropriate Expression Platform

Taylor H. Schreiber, MD, PhD, CEO, Shattuck Labs
  • Can you predict?
  • When do you first know you got it right?
  • When is it too late to change course?
1:00 pm Session Break - View Our Virtual Exhibit Hall

OVERCOMING PURIFICATION CHALLENGES

1:10 pm

Optimizing the Purification Process of the Membrane Protein Bcl-2 for Structural Studies

Jorgen Aden, PhD, Principal Research Engineer, Chemistry, Umea University

The intrinsic pathway of programmed cell death is tightly regulated by the Bcl-2 protein family located at the mitochondrial outer membrane, controlling the release of apoptotic factors. For a long time, obtaining enough Bcl-2 membrane protein for biophysical studies was difficult, which demanded a more efficient purification protocol. This was finally solved by using a classical Escherichia coli expression system, increasing the yield to produce 10-20 mg of full-length protein.

Russell Coleman, Associate Director, Strain Engineering, Pelican Expression Technology

Pelican Expression Technology (formerly Pfenex) is a validated, cost-effective, and scalable platform for recombinant protein production and is well-suited for large-scale protein production, where traditional systems are not suitable. Learn how this Pseudomonas-based expression platform was developed for recombinant protein production. Case studies presented demonstrate the genetic elements, host strains, and automated strain screening workflows enabled broad exploration of expression strategies for two complex antibody scaffolds: Fab fragment and multimeric nanobodies.

1:50 pm

Sane in the Membrane – Salipro One-Step Reconstitution of Membrane Proteins for Drug Development

Jens Frauenfeld, PhD, Founder & CEO, Salipro Biotech AB

Membrane proteins are important drug targets (GPCRs, ion channels), yet are notoriously difficult to work with. We have developed a novel one-step approach for the incorporation of membrane proteins directly from crude cell membranes into lipid Salipro particles. This direct approach termed Salipro DirectMX presents new opportunities for de novo development and characterisation of biologics and small molecules, including phage display, B-cell sorting and cryoEM.

2:10 pm

Small Affinity Tags for Expression, Purification, and Biophysical Studies of G Protein-Coupled Membrane Receptors

Alexei Yeliseev, PhD, Staff Scientist, Group Leader, LMBB, NIH/NIAAA

Affinity tags have been widely applied to purification of G protein-coupled receptors (GPCR) for structural studies. We developed a novel calcium-dependent EF-based affinity system that allows capture and high recovery of GPCR from dilute solutions l. The binding of the EF1 tag to the resin is very strong (high picomolar to low nanomolar range) that allows efficient purification without any loss of the target protein.

Lars Stöckl, Dr., Division Manager, FyoniBio - Service Branch of Glycotope

During the live cycle of a biopharmaceutical project the production needs to stay up to date with productivity and quality demands from early pre-clinical to market phase. Optimization can be done at different stages and on different levels with selecting the right cell line, selecting the right clone or optimizing the media/feed combination and / or optimizing process parameters. We provide case studies which address the different possibilities of optimization.

3:00 pm LIVE PANEL DISCUSSION:

Overcoming Purification Challenges

Panel Moderator:
Alexei Yeliseev, PhD, Staff Scientist, Group Leader, LMBB, NIH/NIAAA
Panelists:
Jorgen Aden, PhD, Principal Research Engineer, Chemistry, Umea University
Russell Coleman, Associate Director, Strain Engineering, Pelican Expression Technology
Jens Frauenfeld, PhD, Founder & CEO, Salipro Biotech AB
Gerhard Grobner, PhD, Senior Lecturer, Department of Chemistry, Umea University
Lars Stöckl, Dr., Division Manager, FyoniBio - Service Branch of Glycotope
3:20 pm PEGS Connects - View Our Virtual Exhibit Hall
4:00 pm Close of Day One
3:30 pm SC1: CAR T Cell Therapy from A-Z
SC2: Intro to Gene Therapy Products Manufacturing & Analytics

Separate registration required. See short course page for details.

Wednesday, May 12

OVERCOMING PRODUCTION CHALLENGES

9:00 am

Protein Engineering and Process Mode for Successful Production of the Extracellular Domain of CD19

Renate Kunert, PhD, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU)

CD19 on malignant B cells constitutes the target of approved CAR T cell-based cancer immunotherapies, but assessment of CAR T cells is hampered by limited availability of CD19. We expressed a novel fusion construct consisting of the full-length extracellular domain of CD19 and domain two of human serum-albumin (CD19-AD2) in CHO and optimized production by continuous fermentation. Purified monomeric CD19-AD2 showed specificity to CAR T cells and also activation of T cell function.

9:20 am

Protein Production: Dealing with the Unpredictable

Neesha Dedi, PhD, Senior Scientist, Protein Sciences, UCB Pharma

Often the first activity within a new project is the generation of protein reagents for assay development, antibody discovery or structural studies.  Giving meaningful project timelines for such activities is challenging as it is almost impossible to predict how long it will take to express and purify a protein.  Using worked examples, I will highlight the unpredictability of recombinant protein production and share details on the processes implemented at UCB to increase chances of success.

 

Yoshihiko Matsuda, PhD, Principal Researcher, Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc.

We have developed a unique recombinant protein secretion system by using Corynebacterium glutamicum and protein expression service Corynex®. It has some advantages such as the secretion of correctly folded and active form, high purity, and endotoxin-free. Recently, “Antibody mimetics” have been developing as well as conventional major antibodies, so we have examined the production of those proteins. We demonstrate Corynex® is very suitable system to produce various kinds of antibody mimetics.

10:00 am

Multi-Epitope Insert Modulates Solubility-based and Chromatographic Purification of Human Papilloma Virus 16 L1-based Vaccine without Inhibiting Virus-Like Particle Assembly

Mike Zhang, PhD, Professor, Biological Systems Engineering, Virginia Polytechnic Institute & State University

Protein purification has been made much easier by the incorporation of many different tags, but most tags don’t have other functions beyond facilitating the purification. Here we show the modifications of a protein, recombinant human papilloma virus 16 L1 (rHPV 16 L1), that not only improve the purification of the modified protein but also afford the protein with desired immunological properties. 

Alengo Nyamay’antu, PhD, Scientific Communication Specialist, Polyplus-transfection
To accelerate process development to produce biotherapeutic protein and antibodies, optimized transient protein production is critical. Choosing the right transfection reagent is key to overcome productivity challenges when working with a wide array of therapeutic antibodies to difficult to express proteins. We demonstrate how FectoPRO® transfection reagent is the go-to transfection reagent that combines productivity and flexibility in suspension CHO and HEK-293 cell lines.
10:50 am LIVE PANEL DISCUSSION:

Overcoming Production Challenges

Panel Moderator:
Renate Kunert, PhD, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU)
Panelists:
Neesha Dedi, PhD, Senior Scientist, Protein Sciences, UCB Pharma
Mike Zhang, PhD, Professor, Biological Systems Engineering, Virginia Polytechnic Institute & State University
Yoshihiko Matsuda, PhD, Principal Researcher, Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc.
Alengo Nyamay’antu, PhD, Scientific Communication Specialist, Polyplus-transfection
11:10 am Session Break - View Our Virtual Exhibit Hall
Matt Stone, Biologics Workflow Specialist, SCIEX
Mukesh Malik, Sr. Application Scientist, SCIEX

Join SCIEX analytical technology experts Matthew Stone and Mukesh Malik during an interactive Q&A session to discuss analytical development and optimization of capillary electrophoresis and liquid chromatography-mass spectrometry based methods for protein characterization and quantification. Exciting topics covered include mAbs and mAb variants, cell and gene therapy, vaccines and more! 

YOUNG SCIENTIST KEYNOTE

11:30 am

Cryo-Electron Microscopy Structures of Spike Glycoproteins Suggest Pan-Coronavirus Antiviral Strategy

Christine Toelzer, Research Associate, University of Bristol, United Kingdom

We research antivirals to combat present and future coronavirus pandemics. We discovered linoleic acid bound to a hydrophobic pocket in the Cryo-EM structure of the SARS-CoV-2 Spike glycoprotein. Ligand binding locked the protein in a compact closed conformation. Conservation of key AA residues suggested a similar pocket exists in other pathogenic coronaviruses.

12:00 pm LIVE:

Q&A with Young Scientist Keynote

Panel Moderator:
Kent Simmons, Senior Conference Producer, Cambridge Healthtech Institute
Panelist:
Christine Toelzer, Research Associate, University of Bristol, United Kingdom
12:10 pm Session Break - View Our Virtual Exhibit Hall
1:00 pm Close of Difficult-to-Express Proteins Conference
Michael G. Tovey, Ph.D, Chief Scientific Advisor of Svar Life Science France, Svar Life Science
Svar´s Expert session will focus on AAV mediated gene therapy and potential neutralizing antibody response to AAV vectors. We know the importance of precisely quantify both the neutralizing antibody response prior to treatment, and the neutralizing antibody response to the recombinant AAV vector following treatment. Talk to us about how our highly sensitive reporter-gene assays are used for the quantification of NAbs to recombinant AAV vectors with different capsid specificities.





Register Now

View By:

Premier Sponsors

FairJourneyBiologics GenScript-CRO Integral-Molecular_NEW  OmniAbUnchainedLabs