Cambridge Healthtech Institute’s 6th Annual

Cell Line Development

New Technologies, Big Data Solutions, and Best Practices for Engineering Robust Cell Lines

20 - 21 March 2024 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 6th Annual Cell Line Development conference offers an expert faculty sharing strategies and best practices on cell engineering, engineering for emerging modalities, new technologies, and the integration of modeling and machine learning into this stage of upstream processing.

Wednesday, 20 March

Registration Open10:30

PLENARY KEYNOTE SESSION

BACK TO THE FUTURE OF BIOPROCESSING—ANTIBODIES TO EXTRACELLULAR VESICLES

11:15

Chairperson's Opening Remarks

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

11:20 PLENARY PRESENTATION:

What Have Monoclonal Antibodies Ever Done for Us? Past, Present, and Future Perspectives on Antibodies and How They Have Driven Bioprocessing Progress

Paul Varley, PhD, Senior Vice President, Development, Alchemab Therapeutics

Advances in bioprocessing have been pivotal to the emergence of monoclonal antibodies as one of the most successful classes of drugs in modern medicine. In this talk we will consider this journey and ask what's next for antibodies. We will also explore how advances in antibody bioprocessing continue to enable the next generation of biological medicines through the emergence of new product modalities.

11:50 PLENARY PRESENTATION:

Extracellular Vesicles as Promising Drug Modalities in Spinal Cord Injury and Other (Neuro-)Degenerative Diseases

Eva Rohde, MD, Chair, Transfusion Medicine, Director GMP Unit, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University Salzburg

Extracellular vesicles (EVs) have emerged as promising new biologic drug modalities. EV therapeutics (EV-Tx) from mesenchymal stromal cells (MSC) exert anti-inflammatory, anti-fibrotic and regenerative effects. MSC-EV-Tx could optimise healing after acute traumatic injury. Challenges in reproducible EV-Tx manufacturing prevent comprehensive evaluation of their efficacy. In early research, the paradigm of “the-process-is-the-product” is valid for complex biologicals. A “one-size-fits-all” approach to solve technical and regulatory issues is not available for EV-Tx. The claimed disease-related mechanisms of action (MoA) of candidate EV-Tx will determine regulatory requirements to be met. This presentation will introduce concepts to accelerate EV-Tx testing in various target diseases.

Session Break12:20

Sponsored Presentation (Opportunity Available)12:35

Networking Lunch (Sponsored Opportunity Available)13:05

CELL ENGINEERING

14:15

Chairperson's Remarks

Emanuel Kreidl, PhD, Senior Expert, Science & Technology, Technical Research & Development, Novartis Pharmaceutical Manufacturing GmbH

14:20

FEATURED PRESENTATION: CHO Synthetic Promoters Improve Expression and Product Quality of Biotherapeutic Proteins

Susie Sou, PhD, Associate Principal Scientist, Cell Line Development and Engineering, AstraZeneca

Strong viral promoters commonly used for recombinant biotherapeutics production in mammalian cells enable maximal expression but provide limited scope to alter their transcription dynamics. In contrast, synthetic promoters designed to provide tunable transcriptional activity can aid more precise regulation for product quality, yield, or reduction of product related-contaminants. Here, we highlight the advantages of employing synthetic promoters with different transcriptional activities for improved production of more complex recombinant proteins.

14:50

Mid-Stage Re-Engineering of Cell Line to Eliminate Problematic HCP

Emanuel Kreidl, PhD, Senior Expert, Science & Technology, Technical Research & Development, Novartis Pharmaceutical Manufacturing GmbH

Despite the definition of critical quality attributes early in the development process having long been the standard in the industry, the sudden identification of novel risk factors can lead to a need for major redevelopment at mid- or even late-stage process development. We show how the redevelopment of a production cell line to remove an HCP can result in suddenly having to focus on completely different attributes.

15:20

RNA Modifications to Improve Biologics Production

Niall Barron, PhD, Principal Investigator, National Institute for Bioprocessing Research & Training (NIBRT)

Epigenetic modifications to the nucleotides in RNA species have been generating considerable interest in recent years. The role of methylation in particular, including characterising the proteins that add (writers), remove (erasers), and interpret (readers) this epigenetic mark, will be discussed. This talk will consider the potential of targeted methylation as an enhancer of mRNA translation and how this mechanism might be applied to improving biologics production.

15:50 End-to-End Automation for Generating Stable Cell Lines for the Development of Advanced Therapeutics

Jolanda Scheenhart, Sales Manager Europe, CYTENA GmbH

Revolutionize cell line development (CLD) workflows with CYTENA's C.STATION. This turnkey automated solution offers efficient single cell isolation, documented clonality assurance, high producer/high-quality clone enrichment, increased throughput, process consistency, and improved data traceability and integrity. It is tailored and configured with the best-in-class instruments and software for monoclonal antibody development, viral vector production, and iPSCs for cell therapy. Join us to explore its transformative impact and discover the future of CLD.

16:05 Digitize your Cell Line Development: Combine Product and Process Insight to Accelerate Your Path to Success

Unjulie Bhanot, Product Marketing Manager for Process Development and Manufacturing, Product Marketing, IDBS

Stable cell line development and identification of lead clones is an iterative and high-throughput process, generating huge volumes of data. Reconciling data from multiple processes across diverse systems can impact the speed of the critical insight scientists need, to select the best performing cell line and clone. Join us to learn how a digital data backbone allows you to capture, analyze, report and leverage your data to make strategic decisions faster. 

Refreshment Break in the Exhibit Hall with Poster Viewing16:35

17:00

Cell Line Engineering for the Characterisation of Macromolecular Complexes Expressed in Physiological Conditions

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 can be used for gene tagging in order to introduce affinity tags and facilitate the purification of proteins/macromolecular assemblies expressed in physiological conditions. We will also discuss tagging proteins with fluorescent reporters in view of imaging and functional proteomics applications.

17:30

KEYNOTE PRESENTATION: Synthetic Biology for Tailored Genetic Vectors

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

Synthetic biology offers a new paradigm for genetic vector design, enabling product-specific cell engineering based on combinatorial tuning of primary cellular synthetic processes such as transcription, translation, and translocation. Our engineering design system utilises a unique platform of genome-scale mining and informatic tools to generate libraries of synthetic parts with user-defined functionality and that can boost biologic manufacturability, replacing “one-size-fits-all” vectorology with design of context-specific genetic systems.

INTERACTIVE BREAKOUT DISCUSSIONS

18:00Interactive Breakout Discussions

Interactive Breakout Discussions are informal, moderated discussions, allowing participants to exchange ideas and experiences and develop future collaborations around a focused topic. Each discussion will be led by a facilitator who keeps the discussion on track and the group engaged. To get the most out of this format, please come prepared to share examples from your work, be a part of a collective, problem-solving session, and participate in active idea sharing. Please visit the Interactive Breakout Discussions page on the conference website for a complete listing of topics and descriptions.

IN-PERSON ONLY BREAKOUT:

IN-PERSON ONLY BREAKOUT: The Role and Future of Cell Line Engineering for Product Quality Optimization

Emanuel Kreidl, PhD, Senior Expert, Science & Technology, Technical Research & Development, Novartis Pharmaceutical Manufacturing GmbH

  • Current trends
  • When to implement in the development process? Is it possible to address and/or initiate this as part of the QbD process?
  • Removal of protein from expression cell line vs. introduction of GOI into dedicated knockdown/knockout cell line?
  • IP, especially in the context of long development timelines – potential advantage of late filing?​

Close of Day18:30

Thursday, 21 March

Registration and Morning Coffee08:00

EMERGING TECHNOLOGIES

08:25

Chairperson's Remarks

Zorica Dragic, PhD, Director, Cell Line Screening and Development, Novartis Pharma AG

08:30

Next-Generation Cell Line Selection Methodology Leveraging Data Lakes, Natural Language Generation, and Advanced Data Analytics

Stephen Goldrick, PhD, Lecturer, Digital Bioprocess Engineering, University College London

We present CLD4, a four-step method for autonomous lead clone selection for biopharmaceutical processes. Step 1 digitises and stores data in a structured data lake. Step 2 calculates the cell line manufacturability index (MICL). Step 3 deploys machine learning for process understanding. Step 4 uses NLG to generate automated reports. CLD4 revealed hidden issues in a CHO cell line, showcasing the power of industry 4.0 principles for informed decision-making.

09:00

Baculovirus-Free Production of Proteins and Virus-Like Particles in Insect Cells

Nina Lehmler, Researcher, Biotechnology, TU Braunschweig

Insect cells are a high-yield and low-cost expression system with mammalian-like post-translational modifications. In contrast to the commonly used baculovirus expression vector system (BEVS), the plasmid-based system enables faster production in high yield and quality, especially of secreted proteins and Virus-Like Particles (VLP). We here present the production of different proteins as well as of Hanta, influenza, entero-, and noro-VLPs, and their applications for antibody development.

09:30

High-Throughput Screening for Product Quality Attributes Early during Mammalian Process Development

Chi-Ting Ho, PhD, Process Expert, Development Operation, Boehringer Ingelheim Pharma GmbH & Co. KG

Cell-line development typically starts with pool generation, and is followed by single-cell cloning, starting with thousands or hundreds of clones and several rounds of ranking and selection, until the final clone can be nominated. Selection and ranking is usually based on cell growth, productivity, monoclonality, stability, and product quality attributes. The latter is gaining more and more importance—highlighting the need to look at product quality attributes as early as possible.

10:00 Efficient and Scalable Eukaryotic Cell-Free Protein Production

Maja Lieven, Dr, Head of Protein Solutions, Protein Solutions, LenioBio

Eukaryotic cell-free protein synthesis (CFPS) shows biomanufacturing potential but has historically faced yield and scalability challenges. ALiCE® marks a breakthrough as fully scalable Eukaryotic CFPS that overcomes these hurdles. We present data on scaled ALiCE® manufacture and protein production and show ALiCE®'s ability to streamline production of proteins containing complex post translational modifications. ALiCE® marks a significant leap forward in CFPS technology, promising expedited bioproduction timelines and increased manufacturing flexibility. 

Coffee Break in the Exhibit Hall with Poster Viewing10:30

11:00

Protein Engineering and Production Platform Based on Cell-Free Technologies

Takanori Kigawa, PhD, Senior Scientist, RIKEN Center for Biosystems Dynamics Research

Protein production using recombinant DNA technology is time-consuming and labor-intensive. We have established a protein production platform based on the cell-free technologies that can produce milligram quantities of proteins totally without the use of recombinant DNA technology. By using this platform, time-consuming and labor-intensive protein expression/purification processes are dramatically accelerated, especially when combined with automated equipment. This platform is therefore very useful for upstream processing.

11:30

Industrial Use of Drosophila S2 Cells for Production of Highly Immunogenic Antigens

Max Søgaard, PhD, Senior Vice President, R&D and Technology, ExpreS2ion Biotechnologies

Viral vaccine antigens produced in mammalian cells often hide from the immune system using glycan shields. S2 cell-produced antigens can reduce glycan shielding due to paucimannosidic glycosylation. Conversely, glyco-engineering S2 cells enhances vaccine effectiveness. Suitable for large-scale production, S2-produced antigens have reached Phase III clinical trials. Utilizing image-based confirmation of monoclonality and micro-scale cultivation accelerates monoclonal S2 cell-line generation, improving efficiency and throughput in cell-line development.

12:00

Exploring Gene Regulatory Networks of IgG Glycosylation Using CRISPR/dCas9 Technology

Anika Mijakovac, PhD, Researcher, University of Zagreb

Immunoglobulin G (IgG) glycans regulate inflammation and as such participate in both disease and aging. To unravel the unknown mechanisms responsible for changes in IgG glycome composition, we utilize a dual approach: large-scale genome-wide association studies (GWAS) to identify genes that associate with IgG glycosylation, and CRISPR/dCas9-based in vitro systems for gene validation. We have discovered that the regulatory networks of IgG glycosylation extend far beyond the known glycosylation-related genes.

12:30 Guide Decision in Early Development with an icIEF-UV/MS Workflow for Comprehensive Charge Variant Characterization

Steven Calciano, BioPharma Business Development Manager, SCIEX

In the evolving landscape of biopharmaceuticals, characterization of charge variants is a critical component of analytical control strategies. Charge variants, arising from PTMs, can significantly impact the efficacy, safety and stability of biologics.

This presentation highlights an integrated icIEF-UV/MS technology to provide in-line MS detection of charge variant species. Applications extend beyond characterization, playing a pivotal role in upstream development, and in elucidating the effect of critical bioprocess parameters on biotherapeutics. 

Networking Lunch (Sponsorship Opportunity Available)13:00

13:45

Chairperson's Remarks

Chrysanthi Sitmalidou, Scientist II, Cell & Gene Therapy Technologies, Technical Operations & Global Technical Development, Orchard Therapeutics

13:50

Advancing Complex Modalities from Genes to Therapies—Impact of Optimized Cell Line Development

Zorica Dragic, PhD, Director, Cell Line Screening and Development, Novartis Pharma AG

CLD FOR CELL AND GENE THERAPIES

14:20

Establishing a Cell-Line Development Platform for Improved GMP LVV Manufacturing in Cell and Gene Therapies

Chrysanthi Sitmalidou, Scientist II, Cell & Gene Therapy Technologies, Technical Operations & Global Technical Development, Orchard Therapeutics

Stable cell lines are widely considered to be the stepping stone to simplify manufacturing processes and further increase yields. In this presentation you will be given an overview of the importance of lentiviral vectors for cell and gene therapy and presentation of Orchard’s stable cell-line development platform. Details on steps to an optimised CLD workflow and an overview of GMP requirements when manufacturing LVV with stable cell lines.

14:50

Towards a Scalable Bioprocess for rAAV Production Using a HeLa Stable Cell Line

Jose Escandell, PhD, Senior Scientist, Animal Cell Technology Unit, iBET

In this presentation we will discuss key aspects related to rAAV production systems' scalability plus current challenges and potential directions for product characterisation. Our presentation focus lies in improving the scalable HeLaS3-based production system. Efforts involve optimising the cell line generation process and improving the cell host; relevant genes for rAAV production, identified through CRISPR-Cas9 genetic screens, will be engineered to create a cell host with enhanced production capabilities.

Close of Summit15:20