Cell Line and Systems Engineering banner

Developing and engineering recombinant protein expression platforms are not for the faint of heart. Many variables must be considered during the development process, including verification and sequence analysis of the gene or protein of interest, codon optimisation, vector construction, and clone/host selection. When challenges arise, protein expression scientists must design new cloning schemes by altering the DNA or amino acid sequence, moving a gene from one vector to another, transfecting the vector to an alternative host, re-selecting the clone, re-characterising the expressed protein or any of the above – a laborious, time-consuming and expensive process. The Cell Line and Systems Engineering conference features effective engineering strategies for recombinant protein expression and production that lead to functional protein products. Learn from seasoned, savvy researchers as they share their real-world experiences, applications, and results.

Sunday, 13 November

Registration Open12:00

Recommended Short Courses*14:00

SC3: Use and Troubleshooting of Eukaryotic Expression Systems
*Separate registration required. See short courses page for details.

Monday, 14 November

Registration and Morning Coffee (Garden Room)07:30

ROOM LOCATION: Group Lounge

TOOLS FOR CELL LINE ENGINEERING AND DEVELOPMENT

08:25

Chairperson's Opening Remarks

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

08:30

Design and Engineering of Mammalian Cell Expression Systems Using Synthetic Biology

Adam J. Brown, PhD, Associate Professor, Chemical & Biological Engineering, University of Sheffield

A genetic expression vector encodes not only the product, it also harbours other DNA sequences that direct the rate of cellular synthetic processes acting in concert to massively impact the functional performance of the engineered cell and product manufacturability.  Our genetic parts toolbox is however rather limited; new parts and engineering strategies are required to enable product-specific design of mammalian expression systems.

09:00

Materials Science Solutions to Enable Co-Culture of Organisms with Different Growth Rates and Specialisations

Karen M. Polizzi, PhD, Professor of Biotechnology, Department of Chemical Engineering, Imperial College London

Co-culture of multiple organisms in the same vessel can be used to facilitate bioproduction or to integrate living cells as analytics in bioprocessing. However, growing organisms together that have different growth rates can lead to one type of cell outcompeting the others.  Here we discuss a materials science solution to enable co-cultures by using cell encapsulation to contain the fast-growing population, allowing cells to grow together in harmony. We have successfully deployed this approach to enable bacterial whole-cell biosensors to be grown alongside mammalian producer cells without detrimental effects on recombinant protein production.

09:30

Genome Editing-Based Approaches to Facilitate the Purification and Characterization of Human Macromolecular Assemblies

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

Macromolecular complexes are cornerstones of most, if not all, biological processes in cells and their preparation in quantity and quality is often a bottleneck. After an overview of widely used recombinant expression approaches, we will illustrate the potential of the CrispR/Cas9 editing technology.  By generating knock-in cell lines where the protein of interest is fused to an affinity tag, the CRISPR methodology facilitates the isolation of macromolecular complexes assembled under physiological conditions. Benefits and limitations of this approach for the purification of multi-protein complexes as well as for other applications will be discussed.

10:00 Automated Workflow for Screening of CRISPR-Edited Cell Lines and Analysis of Monoclonality

Carola Mancini, PhD, Application Scientist, BioPharma Division, Molecular Devices

CRISPR/Cas9 technology has revolutionized targeted gene editing within different applications, including disease model development. We describe an automated workflow integrating different devices to streamline cell line editing. We used CRISPR technology to knockdown the p53 protein in HEK293 cell line. Edited cells were imaged for monoclonality assessment. Measurement of the apoptotic marker Annexin V showed that p53-knockdown cells has decreased cellular apoptotic activity on induction of apoptosis.

10:15 Optimizing Cell Line and Cell Culture Processes

Julian Riba, PhD, CEO, CYTENA

There is an ever-growing need to make the CLD process more efficient in order to keep up with the demand for better therapies. I will present the benefits of the UP.SIGHT, CYTENA’s new single-cell cloning and plate imaging instrument that achieves a probability of clonality >99.99% using 3D Full Well Imagining. I will also introduce a new, automated work station for screening hundreds of clones without user interaction. 

Coffee Break in the Exhibit Hall with Poster Viewing (Verdi and Vivaldi 1&2)10:30

11:15

Enhanced Metabolism and Negative Regulation of ER Stress Support Higher Secretion of Glycoproteins in HEK293

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

Recombinant protein production burdens cell metabolism which may affect titer and quality. Stable HEK293 clones producing either secreted erythropoietin or GFP at different rates were subjected to multi-omics characterization. EPO producers displayed both a shift in oxidative phosphorylation and in ribosomal structure compared to host and GFP. A super producer clone of EPO displayed high expression of negative regulation of ER stress genes which was functionally validated to increase titer.

11:45

A Cell-based Receptor Discovery Platform Enables the Identification of Host Factors Specifically Targeted by the SARS CoV-2 Spike

Bushra Husain, PhD, Director, Biologics Engineering, AstraZeneca

We built a comprehensive library of human proteins engineered for controlled cell surface expression. Coupled to tetramer-based screening for increased binding avidity, we developed a high throughput cell-based platform that enables systematic interrogation of receptor-ligand interactomes. Using this technology, we characterized the cell surface proteins targeted by the receptor binding domain (RBD) of the SARS-CoV spike protein. Host factors that specifically bind to SARS CoV-2 but not SARS CoV RBD were identified, including proteins that are expressed in the nervous system or olfactory epithelium.

12:15 The Leap-in Transposase Platform: Past, Present and Future

Oren Beske, PhD, Amalgamator of Business and Biology, 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. 

Session Break12:45

12:55 Accelerating Upstream Process Development with Direct CQA and Media Analysis Feedback

Nick Pittman, Marketing Manager, Global Biopharmaceutical Business, Waters Corporation

Real-time product attribute and spent media information is important to upstream bioprocess optimization, analysis results are often lagging by weeks. Engineers can now take decisions faster by producing their own at-line quality data, accessible workflows coupling small bioreactors like Sartorius Ambr systems to Waters’ BioAccord LC-MS system. Drug quality and yield can be maximized, and downstream impurities minimized. Development is accelerated from weeks to days, saving resources from multiple optimization cycles.

 

13:25 Efficient Therapeutic Development Using the Pelican Expression Technology Platform

Diane Retallack, Senior Vice President, Platform Technology and Innovation, Pelican - A Ligand Technology

The Pelican Expression Technology platform (formerly Pfenex Expression Technology) is a robust, cost-effective, commercially validated, P. fluorescens-based platform for recombinant protein production, with four approved products utilizing the technology. Case studies are presented demonstrating the extensive Pelican toolbox of genetic elements, host strains, and automated strain screening that enabled rapid screening and development of several candidates including enzymes and challenging antibody formats.

Session Break13:55

CHO CELL LINE ENGINEERING AND DEVELOPMENT

14:15

Chairperson's Remarks

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

14:20

Reprogramming of CHO Cells towards Enhanced Protein Secretion

Mauro Torres, PhD, Research Associate, Manchester Institute of Biotechnology, University of Manchester

A robust secretory phenotype is fundamental for the high-level production of biopharmaceuticals, particularly for those with complex molecular architecture. However, the current CHO cell platforms present deficient protein secretion rates (when compared to dedicated secretory cells). Here, we will discuss the use of regulatory transcription factors as tools for reshaping cellular phenotype and show their potential for engineering CHO cells towards an increased protein secretion.

14:50

Generation of Superior Host Cell Lines for Biomanufacturing Using Plasmid Design & Cell Line Engineering Technologies

Anett Ritter, PhD, Investigator III, Cell Line Screening & Development, Novartis Pharma AG

The presentation covers recent advances of Novartis Cell Line Development toolbox of plasmid elements and novel engineered CHO cell lines, which resulted in increased clone productivity and genetic stability as well as an improved product quality for complex therapeutic proteins. Combining vector technologies with a robust CHO cell line, an accelerated cell line development process was developed for antibodies with significantly reduced efforts for the generation of high-producing CHO clones.

15:20

Engineering and Validation of a Dual Luciferase Reporter System for Quantitative and Systematic Assessment of Regulatory Sequences in CHO Cells

Serif Senturk, PhD, Research Group Leader, Functional Cancer Genomics Group, Izmir Biomedicine and Genome Center, Dokuz Eylul University

Selecting potent regulatory sequences with robust transgene expression is critical for CHO cell line engineering. This talk will summarize the development and validation of a dual luciferase reporter system for quantitative interrogation of such sequences in transient and stable transfectants of CHO cells. Functional execution of this toolkit was achieved with several known constitutive promoters. Experimentally, CMV-mIE yielded the highest transcriptional activity in transient transfectants, while CHEF1a was the strongest in stable pools of CHO-DG44 cells. Together, the reporter system is a viable tool for selecting established or identifying novel regulatory sequences in CHO and perhaps other mammalian cell lines.

15:50

StoCellAtor: A Bacterial Cell Modelling Framework Linking Resource-Based Stochastic Translation to the Optimal Design of Synthetic Constructs

Peter Sarvari, MEng, MSci, eTrading Quant, Global Markets, BNP Paribas

We built StoCellAtor, a bacterial whole-cell modelling (WCM) framework that entails a detailed, codon-level translation model combined with the stochastic version of an existing WCM. StoCellAtor can be used to link a synthetic construct’s modular design (promoter, ribosome binding site, and codon composition) to protein yield and cellular burden during continuous culture, with a particular focus on the effects of low-efficiency codons and their impact on ribosomal queues.

Refreshment Break in the Hall with Poster Viewing (Verdi and Vivaldi 1&2)16:20

17:05

Tailored Glycosylation, Improved Quality and Faster Cell Line Development

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

Through targeted and systematic CHO cell line engineering, we have developed CHO cell-based platforms, enabling rapid production of tailored glycoforms on therapeutic proteins, with improved protein quality and predictable cell line development. With the glycoengineered CHO platform (geCHO), the effect of N-glycans on therapeutic proteins can be screened, to determine the optimal glycoform which can then be manufactured using the geCHO cell lines.

17:35

The GlycomiR Toolbox: A Novel System for Glycosylation Engineering by Natural and Artificial miRNAs

Florian Klingler, MSc, Researcher, Kerstin Otte Laboratory, Biberach University of Applied Sciences

N-linked glycosylation is a critical quality attribute of many biopharmaceutical products that needs to be controlled. A screening of a miRNA library identified 82 miRNA sequences capable of altering galactosylation, sialylation, and fucosylation. Subsequent validation provided insight into the intracellular mode of action of miRNAs regulating cellular glycosylation pathways. Moreover, a multiplex modulation approach and rational design of artificial miRNAs demonstrated the potential of miRNAs to fine tune expressed glycosylation patterns.

Welcome Reception in the Exhibit Hall with Poster Viewing (Verdi and Vivaldi 1&2)18:05

Close of Cell Line & Systems Engineering Conference19:05