Cambridge Healthtech Institute’s 4th Annual

Cell Line Development

Technologies and Best Practices for Engineering Robust Cell Lines

23 - 24 March 2022 ALL TIMES CET

As the biopharma field becomes increasingly more complex, the engineering of cell lines remains a foundation step for developing biologic drugs. This foundation applies emerging research and breakthrough strategies to achieve cell lines that accomplish project goals quickly and inexpensively. In support of these goals, the 4th Annual Cell Line Development conference offers an expert faculty sharing strategies and best practices on codon optimization, vector construction, and engineering of clones and host systems. Presentations for 2022 reflect a focus on emerging modalities, new technologies, quality systems and next generation informatics.

Wednesday, 23 March

PLENARY LOCATION: Vivaldi 1 & 2

PLENARY SESSION: FUTURE OF BIOPROCESSING

11:15

Chairperson's Remarks

Margit Holzer, PhD, Owner, Ulysse Consult
11:20

PLENARY PRESENTATION: Is Current Bioprocessing Fit for the Future?

Alois Jungbauer, PhD, Professor & Head, Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences (BOKU)

The future of global bioprocessing demands flexible, scalable solutions that can accommodate the rapidly changing landscape of the biopharmaceutical industry while also minimizing the impact on the environment in the face of climate change. Currently, two extreme production scenarios exist – the use of fully disposable factories offering flexibility and speed; and large stainless steel plants designed for high capacity. This presentation will discuss how bioprocessing can meet the needs of both the industry and the environment for the benefit of patients, economics and supply, and whether current bioprocessing is fit for the future.

11:50

PLENARY PRESENTATION: Intensification Strategies: The Path to Continuous Processing

Stefan R. Schmidt, MBA, PhD, COO & Head, Operations, BioAtrium AG

Continuous processing is the holy grail for many industries and became popular for bioprocessing in the last decade, too. Intensification is a prerequisite to enable a step wise transformation towards that goal. This presentation gives a comprehensive overview on strategies where and how to implement process intensification and quantifies the benefits like plant occupancy time and optimizing capacity based on successful examples and case studies.

12:20 Session Break

ROOM LOCATION: Rossini 1

Simon Margerison, Director, Customer Application Support, Berkeley Lights, Inc

CHO cell line selection is a painful bottleneck in biotherapeutic development, particularly for complex molecules like bispecifics. The Opto™ CLD workflow on the Beacon® system accelerates early CLD by integrating high throughput cell sorting, cloning, culture, productivity, growth, and product quality assays into a single, 5-day automated process. Hear about capabilities of on-chip detection that pinpoints best clones early on.  

13:00 Refreshment Break in the Exhibit Hall with Poster Viewing (Verdi)

DEVELOPING CHO-BASED CELL LINES

13:45

Chairperson’s Opening Remarks

Valentine Chevallier, PhD, Scientist, Upstream Process Development, UCB, Belgium
13:50

Precision Engineering of CHO Cell Expression Systems Using Synthetic Biology

David C. James, PhD, Professor, Bioprocess Engineering, University of Sheffield

Genetic vector design underpins both optimal synthesis of an encoded recombinant protein and engineering of the host cell to improve functional performance. Engineering context is paramount.  However, biopharmaceutical proteins made by CHO cells are structurally variable and accordingly require a combination of dynamic cellular processes attuned to their idiosyncratic requirements. To achieve product-specific cell engineering we need new tools and strategies that enable us to design and precisely control the relative rate of synthetic processes encoded on genetic vectors that directly impact product manufacturability.

14:20

Oxidative Stress-Alleviating Strategies to Improve Recombinant Protein Production in CHO Cells

Valentine Chevallier, PhD, Scientist, Upstream Process Development, UCB, Belgium

Understanding of oxidative stress is key to support future development of control strategies. In literature, oxidative stress is defined as the imbalance between oxidant molecules generation and antioxidant defense within the cells. Under bioprocess conditions, reactive oxygen species can be generated by multiple sources. By playing on media composition we modulated oxidative stress to improve product quality and process performance.

14:50

Pre-Stage Perfusion and Ultra-High Seeding Cell Density in CHO Fed-Batch Culture: A Case Study for Process Intensification Guided by Systems Biotechnology

Raphael Drerup, Associate Director, Cell Culture, Boehringer Ingelheim Pharma GmbH & Co., Germany

Process intensification is broadly discussed to increase biopharmaceutical production output. This encompasses classic elements of process development combined with largely increased cell densities, the latter being usually enabled by cell retention techniques of various kinds. We show a case study to apply such an approach to an existing cell line and analyze consequential requirements for optimal cell lines to start right from the beginning in a highly intensified process setting.

Fernando Aprile Garcia, PhD, Cell Line Development, Cytena

Cell line development (CLD) is time and resource intensive, with a constant need for faster clone generation while providing assurance of monoclonality. The all-new C.STATION fully automates stable CLD for monoclonal antibody production. This workstation integrates CYTENA’s flagship instrument, the UP.SIGHT, to  enable a probability of clonality >99.99% and fulfill required regulatory expectations. Furthermore, the workstation screens hundreds of clones without user interaction, enabling confluency, cell count and mAb titer measurement.

Oren Beske, PhD, Amalgamator of Business and Biology, Business Development, ATUM

Launched only a few years ago, the Leap-In Transposase platform has rapidly become an industry standard technology for the generation of CHO cells for the manufacturing of antibodies and other biologics.  This presentation will highlight achievements and case studies of the platform including high titer mAb manufacturing, rapid anti-COVID responses, and some novel, next generation, applications.  

15:50 Refreshment Break in the Exhibit Hall with Poster Viewing (Verdi)

OPTIMIZING CELL LINE DEVELOPMENT

16:25

Top-Down and Bottom-Up Strategies for Production and Characterization of Human Multi-Protein Complexes

Arnaud Poterszman, PhD, Research Director, Integrated Structural Biology, IGBMC

Macromolecular complexes are cornerstones of most, if not all, biological processes in cells. We will illustrate how the CrispR/Cas9 editing technology allows to label and isolate native protein assemblies from their natural cellular environment and discuss the potential of the baculovirus expression vector system for reconstitution of multi-subunit complexes. As model systems, we will use transcription regulators such pTefb, nuclear receptors or the 10 subunits transcription factor TFIIH.

16:55

A Blueprint from Nature: Multi-Omic Analysis of CHO and Plasma Cells Unveils Novel Targets for Cell Line Engineering

Nikolas Zeh, PhD, Postdoctoral Ressearcher, Cell Biology, Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Germany

Biotherapeutics are mainly produced by Chinese hamster ovary (CHO) cells. However, in nature terminally differentiated plasma cells are responsible for highly efficient antibody production in mammals. In this study we performed a comprehensive multi-omics analysis of CHO and plasma cells to identify crucial differentially expressed genes with beneficial impact on CHO cell characteristics such as productivity, growth or viability and rationally optimized our industrial workhorse by leveraging nature as blueprint.

17:25

Improvement of the Efficiency and Quality in Developing a New CHO Host Cell Line

Zhimei Du, PhD, Vice President, Process Sciences, Atara Biotherapeutics

To maximize the productivity of biotherapeutics, we developed a novel expression system, which allows for the simultaneous integration of numerous independent selective markers. Combining a newly created multi-auxotrophic mutant of CHO-K1 host lineage and transposon-based vector system, a single-step transfection with 8 transgenes resulted in a high frequency of transfectant clones bearing high copy numbers of incorporated vectors and that are capable of pronounced productivity.

18:00 Close of Day

Thursday, 24 March

08:00 Registration Open and Morning Coffee (Foyer)

ROOM LOCATION: Rossini 1

CELL ENGINEERING

08:25

Chairperson’s Remarks

Bjørn Voldborg, MSc, Head, National Biologics Facility, DTU Bioengineering, Technical University of Denmark
08:30

Denmark’s National Biologics Facility

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

The National Biologics Facility has delivered state-of-the-art cell line engineering work since 2012. The obtained knowhow, expertise, infrastructure and optimised CHO cell lines are now being deployed for the delivery of high quality cell line engineering/development and protein production to the benefit of industry and academia. Examples of engineered CHO cell lines resulting in improved product quality, robust bioprocess, predictable gene integration, elimination of clonal variation and tailored N-glycans will be presented.

09:00

Proteomic Profiling of IgG1 Producing CHO Cells Using LC/LC-SPS-MS 3: The Effects of Bioprocessing Conditions on Productivity and Product Quality

Jonathan Bones, PhD, Principal Investigator, Characterisation and Comparability Laboratory, National Institute for Bioprocessing Research and Training (NIBRT), Ireland

Application of LC-MS based quantitative proteomics is described for investigating bioprocess behaviour. In the first study, we used proteomics to determine process response to altered conditions, combined with in depth characterisation of the expressed monoclonal antibody to explore product process interplay. In the second study, proteomics was applied to study cells pre and post transfection for AAV viral vector generation followed by functional validation targeting identified pathways to increase yield.

09:30 KEYNOTE PRESENTATION:

Strategies for Optimizing Cell Line Development

Zorica Dragic, PhD, Director, Cell LIne Screening and Development, Novartis Pharma AG, Switzerland
Lars Stöckl, PhD, Managing Director, FyoniBio GmbH

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.

 

10:30 Coffee Break in the Exhibit Hall with Poster Viewing (Verdi)
11:15

Reprogramming Cells to Build Better Manufacturing Factories

Alan Dickson, PhD, Professor of Biotechnology; Director, Centre of Excellence in Biopharmaceuticals, Manchester Institute of Biotechnology, The University of Manchester, United Kingdom

Nature has provided us with mammalian cells that can (and are used) as factories for production of the desirable biological therapeutics. However, yield and capacity to make specific products can be limited by the properties of different cell types. Nature also tells us (although we may not fully understand it) that specific cellular properties will enable us to generate better cell factories, properties that can be engineered into existing cells.

11:45

Systems Biology Lessons from Bioproduction of Challenging Proteins in CHO and HEK293

Johan Rockberg, PhD, Professor, Antibody Technology and Directed Evolution, KTH Royal Institute of Technology

CHO has proven capable to produce a vast array of biological products at high yield and quality during the last couple of decades. Here we report on our attempts to use systems biology to learn from the cases where the secretional, translational and metabolic burden challenges the normal capacity of these cells. We further show how challenging proteins could be rescued in HEK293 cells and our hypothesis around why. Further, we exemplify how OMICS can be used to define limiting factors improving the activity of therapeutic enzymes over 100-fold.

12:15

Using Site Specific Integration Expression System to Generate Cell Lines for Multi-Specific Modalities

Barbara Tevelev, PhD, Senior Scientist, Cell Line Development, Biotherapeutics Pharmaceutical Sciences, Pfizer, United States

Site Specific Integration (SSI) expression systems offer robust means of generating highly productive and stable cell lines for standard mAbs.  However, complex multi-specific modalities can prove challenging for SSI expression systems.  Here we illustrate how an SSI system can be implemented for the production of a bi-specific molecule, and present work towards vector engineering for production of multi-chain, multi-specific molecules.

12:45 Session Break
Rachel Richer, Staff Scientist, Cell Line Development Group, FUJIFILM Diosynth Biotechnologies

ApolloX™ is a cell line development platform, incorporating next-generation screening technology, enhanced media selection, and technical expertise. The technology supports aggressive cell line development timelines, whilst ensuring robust and high quality cell lines are obtained. Also described here is a ‘manufacturability assessment’ strategy, which is performed both in-silico using a proprietary bioinformatics tool (Orbit™) together with material generated from transfectant pools, and used to evaluate platform-fit for multiple candidate molecules.

13:25 Session Break

PROBLEMS AND SOLUTIONS

13:45

Chairperson’s Remarks

Emanuel Kreidl, PhD, Senior Research Officer, Upstream Processsing, Ichnos Sciences, Switzerland
13:50

Benchmarking Commercial Media and Impact on Clone Selection

Emanuel Kreidl, PhD, Senior Research Officer, Upstream Processsing, Ichnos Sciences, Switzerland

The impact of cell culture media and feeds on the performance of CHO cells has been well established with many groups going so far as to develop specific in-house media for each project/clone. As this approach is generally incompatible with today’s highly compacted timelines in development, we compared the performance of commercially available media and feeds both before and after selection of production clones.

14:20

Novel Aspects in Engineering the Correct Chain Pairing in Bi- and Multi-Specific Antibodies

Gordana Wozniak-Knopp, PhD, Senior Scientist, Molecular Biology, University of Natural Resources and Life Sciences Vienna

With over 300 candidates in clinical trials and preclinical development, bispecific antibodies are bound to make a difference to human health, and are invaluable drugs due to their unique modes of action. Their intense engineering resulted in a plethora of different formats. Especially in asymmetrical multispecifics, the issue of chain mispairing requires novel solutions; some of them, based on Fab constant domain-exchanged antibody and chemically composed trispecifics, will be presented.

14:50

Cell-Free Glycosylation of mAbs

Elli Makrydaki, PhD, Research Associate, Chemical Engineering, Imperial College London

Cell-free protein synthesis (CFPS) is a dynamic field that facilitates rapid protein expression enabling high-throughput screening prior to large scale cell-based expression. The vast majority of protein-based biotherapeutics such as monoclonal antibodies are predominantly produced in mammalian cell-based expression systems, primarily Chinese hamster ovary (CHO). A key feature of CHO cells is the ability to perform human-like glycosylation. This presentation will focus on evaluating the performance of an in-house CHO CFPS for the production of different monoclonal antibody targets whilst investigating their glycosylation pattern. Finally, a novel modular strategy to modify the glycosylation pattern will also be presented.

15:20 Close of Summit