2019 Archived Content
p8

Cambridge Healthtech Institute’s 21st Annual

Recombinant Protein Expression and Production

Achieving Quality and Quantity

January 15-16, 2019

 

Great strides have been made in the expression, production, and purification of biotherapeutics. However, hurdles remain. The efficient expression and production of these valuable biomolecules face challenges in improving their quantity and quality while minimizing time and cost. Thus, higher-throughput expression and purification as well as more flexible expression platforms are in even greater demand. Unfortunately, there is no “universal” production system which can guarantee high yields of recombinant protein, particularly as every biomolecule itself causes its own issues in terms of expression.

Cambridge Healthtech Institute’s 21st Annual Recombinant Protein Expression and Production conference explores the newest data and innovations relating to the best choices in hosts/systems, as well as ways to “rescue” existing systems and make them work more effectively to produce the quality and quantity of the desired biotherapeutic.

Final Agenda

TUESDAY, JANUARY 15

1:00 pm Registration (Sapphire West Foyer)

1:30 Refreshment Break in the Exhibit Hall with Poster Viewing (Sapphire Ballroom)

From Protein Expression to Biotherapeutic Product
Sapphire 400

2:00 Chairperson’s Opening Remarks

Henry C. Chiou, PhD, Director, Cell Biology, Life Science Solutions, Thermo Fisher Scientific


KEYNOTE PRESENTATION

 2:05 Expression Systems for Various Biologics Modalities: Today and Tomorrow

Zhimei Du, PhD, Director, Bioprocess & Clinical Manufacturing, Merck

Developing a robust expression system is the most critical step during biologics development for all modalities, including mAb, non-mAb complex molecules, and CAR-T, etc. A robust expression system can impact the productivity, also product qualities and process controls. In this presentation, we discuss the details of the major factors that need to be considered when developing the new expression system, and how to apply Quality-by-Design strategy at this stage.

2:45 NEW: Discovering de novo Peptide Substrates for Enzymes Using Machine Learning

Kim_WoojooWoojoo Eunice Kim, Research Scientist, Burkart Lab, Department of Chemistry and Biochemistry, University of California, San Diego

Peptide Optimization with Optimal Learning (POOL) method is an iterative machine learning process by which experimental data is deposited into a mathematical algorithm that selects potential peptide substrates to be tested experimentally. This process is repeated until a suitable set of de novo peptide substrates are discovered. We employed this technology to discover orthogonal peptide substrates for 4’-phosphopantetheinyl transferase, an enzyme that covalently modifies proteins.

3:15 ExpiSf, ExpiCHO and Expi293: Latest Developments in High-Titer Transient Protein Expression

Zmuda_Jonathan_PEX_Pipeline7Jonathan Zmuda, PhD, Director, Cell Biology, Thermo Fisher Scientific

The Expi Expression Systems comprise three different cell hosts to provide researchers with unprecedented access to high-titer recombinant proteins. Here, we highlight the latest data and recent additions to the Expi family of products, including the first ever chemically defined insect expression system, ExpiSf, a structural biology module for the Expi293 expression system, GMP-banked Expi293 and ExpiCHO-S cells and ExpiCHO Stable Production Media to support the transition from transient to stable protein expression.

3:45 Refreshment Break in the Exhibit Hall with Poster Viewing (Sapphire Ballroom)

 

Effective Expression and Production of Vectors for Gene Therapy
Sapphire 400

 

4:30 Optimizing Sf9-Based Stable Cell Lines for the Production of Highly Infectious rAAV Vectors

Zolotukhin_Sergei Sergei Zolotukhin, PhD, Professor, Department of Pediatrics, College of Medicine, University of Florida

We describe a new insect cell-based production platform utilizing attenuated Kozak sequence and a leaky ribosome scanning to achieve a serotype-specific modulation of AAV capsid proteins stoichiometry. By way of example, rAAV5 and rAAV9 were produced and comprehensively characterized side by side with HEK293-derived vectors. The data will be presented demonstrating a superior infectivity and higher genetic identity of OneBac-derived rAAV vectors providing a scalable platform for good manufacturing practice (GMP)-grade vector production.

5:00 LVV Production Process: Recent Advances and Opportunities for Innovation

Waghmare_YogeshYogesh Waghmare, PhD, Associate Director, Vector Downstream Process Development, Bluebird Bio

LentiViral Vector (LVV)-based Cell and Gene Therapy products are steadily increasing in number. Industrial production of LVV poses significant challenges compared to AAV due to the large size, complexity, and labile nature of LVV. An overview of industrial LVV production process evolution, recent technological advances, and LVV specific challenges will be presented.

5:30 Close of Day

5:30 - 5:45 Short Course Registration (Sapphire Ballroom)


5:45 - 8:45 Recommended Dinner Short Courses*

SC5: Transient Protein Production in Mammalian Cells - Detailed Agenda

Click here for more details.

*Separate registration required

WEDNESDAY, JANUARY 16

7:45 am Registration and Morning Coffee

Effective Expression and Production of Antibodies
Sapphire 400

 

8:15 Chairperson’s Remarks

Jie Zhu, PhD, Associate Director, Cell Culture & Fermentation Sciences, MedImmune

FEATURED PRESENTATION

8:20 Controlling Protein Quality and Antibody Expression

Robinson_Anne Anne Skaja Robinson, PhD, Head, Department of Chemical Engineering, Carnegie Mellon University

Monoclonal antibodies (mAbs) are a class of commercially valuable biopharmaceuticals that are used for treating diseases that are typically expressed in mammalian cell lines such as Chinese Hamster Ovary (CHO) cells to enable posttranslational modifications. One such posttranslational modification that results in structural and pharmacological changes in the protein is N-linked glycosylation. This talk addresses approaches to maintaining desired product quality of mAbs in the presence of process variations during manufacturing.

8:50 Therapeutic Antibody Fragments: Simplifying the Choice of the Expression Platform and Optimizing Protein L Capture

Billiald_PhilippePhilippe Billiald, PharmD, PhD, Professor, Biochemistry, University of Paris-Sud; Co-Founder, Acticor Biotech

Therapeutic antibody fragments are produced from various hosts, but no downstream process is well established. Here, we report a universal method to confer Protein L binding ability to any antibody fragment. In addition, based on a case study, we assess E. coli, P. pastoris and CHO expression systems in terms of cell line development, culture time, product quality and cost. We report differences to consider before pharmaceutical development and moving forward to the clinic.

9:20 Overcoming the Key Bottlenecks in Cell Line Development: Increasing Titers and Streamlining Cell Line Development

Fay Saunders, Head of Upstream Mammalian Development, Mammalian Cell Culture,  FUJIFILM Diosynth Biotechnologies

 

9:35 New: Scaling Up and Scaling Out: Pushing the Boundaries of Transient Protein Production

Michael Fiebig, PhD, Director, Products and Innovations,  Absolute Antibody Ltd

Whilst transient yields have improved drastically in the last decade, scalable systems are time-consuming and costly to implement. Absolute Antibody has developed systems which scale up and scale out protein expression and purification, enabling the rapid and cost-effective production of milligram-to-gram quantities of large panels of proteins.

9:50 Coffee Break in the Exhibit Hall with Poster Viewing (Sapphire Ballroom)

10:35 Multi-Specificity of a Recombinant Monoclonal Antibody

McLean_Gary Gary McLean, PhD, Reader in Molecular Immunology, Cellular and Molecular Immunology Research Centre, London Metropolitan University; Honorary Senior Research Fellow, National Heart and Lung Institute, Imperial College London

Whilst studying the cellular molecular switch protein m-ras, a recombinant monoclonal antibody was generated to an m-ras-specific peptide sequence. The recombinant antibody was expressed in mammalian cells and interestingly the IgG bound to both recombinant full-length m-ras and h-ras proteins. The cross-reactive binding of the monoclonal Ab to h-ras revealed that the Ab showed preference for peptide sequences containing multiple positively charged amino acid residues. These data reinforce the concept of antibody multi-specificity through multiple interactions of the Ab paratope with diverse polypeptides and emphasize the importance of immunogen and Ab selection processes when generating recombinant monoclonal Abs.

11:05 Mammalian Display Platform for Facile, FACS-Based Engineering of Antibodies and Other Receptors

Maynard_Jennifer Jennifer Maynard, PhD, Associate Professor, Chemical Engineering, University of Texas at Austin

Discovery of new antibodies is most commonly performed using phage or yeast display, but mammalian cells are used for large-scale production because of the complex antibody structure, including multiple disulfide bonds and a key glycosylation required for function. To circumvent problems associated with changing hosts, we developed a screening platform on CHO cells which allows for antibody selection in the same host used for manufacturing. We have used this approach to affinity mature an antibody Fab, a human T cell receptor and modulate binding of human IgG1 Fc to the FcgRIIIa receptor. Finally, modifications of the approach allow us to select for receptors that activate T cells in the presence of ligand as opposed to just binding the ligand.

11:35 Understanding and Engineering Fc Glycans in CHO Cells for the Production of Therapeutic Proteins

Zhu_Jie Jie Zhu, PhD, Associate Director, Cell Culture & Fermentation Sciences, MedImmune

Glycosylation of monoclonal antibody and derivatives plays an important role for complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) functions. Case studies are presented here on the generation of stable CHO cells cell line to produce recombinant proteins with desirable and consistent glycosylation patterns in Fc domain using both vector and host engineering approaches.

12:05 pm Session Break

12:15 Luncheon Presentation (Sponsorship Opportunity Available) or Enjoy Lunch on Your Own

1:15 Session Break


PLENARY KEYNOTE PANEL (Aqua Salon)

Click here for more details.

PepTalk Perspectives: Point-Counterpoint Discussions

2:00 Plenary Keynote Introduction

Norman Packard, PhD, CEO, Daptics


2:10 Plenary Keynote Panel

Moderator:
Howard Levine, PhD, President and CEO, BioProcess Technology Consultants
Panelists:
George BadescuGeorge Badescu, PhD, Vice President, Scientific Affairs, Heidelberg Pharma AG
Cox ManonManon Cox, PhD, Co-Founder & CEO, NextWaveBio
Zhimei Du, PhD, Director, Bioprocess & Clinical Manufacturing, Merck
Paul Jorjorian, Vice President, BioProcess Sciences, Thermo Fisher Scientific
Marina Kirkitadze, PhD, Deputy Director, Head of Biophysics and Conformation Unit, Analytical R&D Biochemistry, Sanofi Pasteur, Canada
Stefan SchmidtStefan R. Schmidt, PhD, MBA, Head, Operations (COO), BioAtrium AG

 

3:05 Refreshment Break in the Exhibit Hall with Poster Viewing (Sapphire Ballroom) 

Effective Expression and Production of Recombinant Proteins
Sapphire 400

4:00 Chairperson’s Remarks

Bjørn Voldborg, MSc, Director, CHO Cell Line Development, The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark

4:05 Implementing Next-Generation Sequencing for DNA-Based Sequence Variant Analysis of Recombinant Proteins

Goepfert_Ulrich Ulrich Goepfert, PhD, Principal Scientist, Large Molecule Research, Roche Pharma Research & Early Development, Roche Innovation Center Munich

Sequence variants are unintended amino acid substitutions in biopharmaceuticals, which can either be due to the manufacturing process or mutations of the transgene. Transgene mutations are permanent properties of affected cell lines and may give rise to critical quality attributes. Therefore, mutated cell lines need to be identified and excluded from development. We share our experience with Next-Generation Sequencing as an efficient and highly sensitive method to detect DNA-based sequence variants.

4:35 The BEST of Both Worlds – Targeted Integration and Multiple Copies: How Can These Go Together for Improved Cell Line Development?

Bauer_Anton Anton Bauer, PhD, MBA, COO, R&D, The Antibody Lab GmbH

Targeted Hot Spot integration and multiplication of independent expression units – can this go together and even speed up cell line development? Targeted integration of the gene-of-interest in an expression hot spot is contributing to high protein expression yields as well as to epigenetic stability of selected clones. This increases the probability of identifying promising clones and lowers the attrition rate during process development. By targeting the Rosa26 Hot Spot in vitro we generated BAC-based expression vectors, which integrated in multiple copies into the CHO host cell chromatin and acted as independent expression units. This allowed us to adapt the selection process and developed long-term stable high-yield production cell lines at an unprecedented speed. New data from case studies and further improved vector versions demonstrate the capability of the technology.

5:05 Optimizing Productivity and Product Quality of Difficult-to-Express Biosimilars with a Novel NS0 Platform

Sampey_DarrylDarryl Sampey, PhD, President & CEO, Research & Development, BioFactura, Inc.

Biosimilar cell lines that produce complex glycoproteins such as monoclonal antibodies must be both highly productive and express a product with critical quality attributes closely matching those of the innovator references. In this presentation, a novel biomanufacturing platform and case studies are described that harness the commercially established NS0 host cell in new ways to create stable, productive cell lines with product characteristics meeting biosimilar technical and regulatory demands.

5:35 Engineering CHO Cell Lines for the Production of Hard-to-Produce Proteins

Voldborg_Bjørn Bjørn Voldborg, MSc, Director, CHO Cell Line Development, The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark

Using our high-throughput cell line engineering platform, we have engineered CHO cells able to produce a therapeutic protein that has previously not been possible to produce in CHO cells. This approach may result in improved therapeutic proteins, with better biological properties, such as increased half-life, improved activity, etc.

6:05 - 7:00 Networking Reception in the Exhibit Hall with Poster Viewing (Sapphire Ballroom)

7:00 Close of Recombinant Protein Expression and Production Conference