Optimising Expression Platforms banner

The utilization of recombinant proteins for basic research, target development, clinical diagnostics, and therapy continues to expand. Consequently, the efficient expression and production of these valuable biomolecules face challenges in improving their quantity and quality while minimizing time and cost. To meet these demands, an increasing variety of recombinant production platforms called “cell factories” are being developed. Unfortunately, there is no “universal” production system which can guarantee high yields, particularly as every protein itself causes its own issues in terms of expression and production. Through case studies, Cambridge Healthtech Institute's Optimising Expression Platforms conference offers comparisons, evaluations, and solutions that enable protein expression researchers to efficiently express the therapeutic protein of their choice.

Wednesday, 15 November

Registration Open and Morning Coffee07:30

OVERCOMING EXPRESSION AND PRODUCTION CHALLENGES OF DIFFICULT-TO-EXPRESS PROTEINS

08:25

Chairperson's Opening Remarks

Ana Sofia Coroadinha, PhD, Lab Head, Health & Pharma Division, Animal Cell Technology Unit Cell Line Development and Molecular Biotechnology Lab, IBET

08:30

Development of Specialized Bacterial Strains for High-Level Production of Recombinant Membrane Proteins

Georgios Skretas, PhD, Director, Institute for Bio-innovation, Biomedical Sciences Research Center "Alexander Fleming;" Founder & CEO, ResQ Biotech

We will describe the development of first- and second-generation E. coli SuptoxD and SuptoxR, two specialized strains for high-level recombinant membrane protein (MP) production. These engineered strains can: (1) suppress the toxicity that frequently accompanies MP overexpression, thus enabling enhanced levels of final bacterial biomass; and (2) markedly increase the cellular accumulation of membrane-embedded protein. Combined, these two positive effects result in dramatically enhanced volumetric yields for various prokaryotic and eukaryotic recombinant MPs.

08:50

Membrane Protein Production Using Insect Cells

Alice Rothnie, DPhil, Senior Lecturer, Biochemistry, Aston University

Membrane proteins play important roles in cell signaling, the influx and efflux of nutrients and metabolites, and many are potential drug targets. For their structural and functional analysis, high yields of correctly folded and modified protein are needed. Multidrug resistance protein 4 (MRP4/ABCC4) is a multi-substrate primary transporter that has been expressed using both recombinant baculovirus infection of Sf9 insect cells, and baculovirus-mediated transduction of Freestyle HEK cells.

09:10

Lactococcus lactis, a Promising Cell Factory to Functionally Express Membrane Proteins

Annie Frelet-Barrand, PhD, Researcher, MN2S, Institut FEMTO-ST

Membrane proteins (MPs), important drug targets, display crucial functions in organisms but their study remains difficult (hydrophobicity and low abundance). Their overexpression is mandatory for structural and functional characterizations but this strategy could encounter obstacles. Using the NICE system and the L. lactis strains, in the last twenty years, more than 100 MPs were expressed allowing either their functional and/or structural characterization. Recently, one eukaryotic membrane protein was expressed at a high-expression yield and allowed the formation of intracellular vesicles. In conclusion, L. lactis represents an interesting system for expression of MPs.

09:30 Fyonibio's Versatile Cell Line Expression Platforms for the Development of Complex Molecules

Lena Tholen, Director, Cell line and Bioprocess Development, FyoniBio GmbH

Here FyoniBio presents its versatile highly productive expression platforms, the mammalian host cell systems CHOnamite®, for e.g. bi-specific mAbs incl. our CHOFlow® for afucosylated antibodies and the human GEX® platform for the development and production of complex glyco-biopharmaceuticals in the desired quality. A case study from a customer project demonstrates the suitability of our cell line platform for the production of complex bispecific mAbs in combination with bioprocess optimization capabilities.

09:45 From Modular DNA Assembly to Recombinant Protein Production at Polyplus

Marine Houdou, Genetic Engineering Specialist, Polyplus

With recent acquisitions of e-Zyvec and Xpress Biologics, Polyplus offers now a unique integrated pDNA service with plasmid engineering and manufacturing. Our unique DNA assembly technology and proprietary software allow the tailor-made design and production of any plasmid. Synergistically, the 30+ years of combined expertise at Xpress Biologics reinforces our services at Polyplus to support recombinant protein manufacturing from 100 mg to 50 g at Research, GLP and GMP grade.

Session Break to Transition into Plenary Keynote10:00

PLENARY KEYNOTE SESSION

10:10

Plenary Keynote Introduction

Enkelejda Miho, PhD, Professor, University of Applied Sciences and Arts Northwestern Switzerland, and Managing Director, aiNET

10:15

Benchmarking the Impact of AI Biologics Discovery and Optimisation for Pharma

Rebecca Croasdale-Wood, PhD, Director, Augmented Biologics Discovery & Design, Biologics Engineering, Oncology, AstraZeneca

The biologics landscape is rapidly changing with the number of AI-enabled biologics in pre-clinical and clinical stages estimated to be 50-60 (1). This change is driven by the increase in enterprise software solutions to capture and store data, augmented discovery workflows, improvements in machine learning technology, and advances in computing power. Augmented biologics discovery has the potential to revolutionize biologics discovery, yet information of how in silico technologies perform, versus traditional discovery platforms is scarce. At PEGS Europe, we will present current in silico biologics design and optimisation technologies, with a focus on our internal efforts to benchmark the impact of combining novel in silico technologies with our existing biologics discovery platforms.

10:45

Keynote Chat 

Rebecca Croasdale-Wood, PhD, Director, Augmented Biologics Discovery & Design, Biologics Engineering, Oncology, AstraZeneca

Interviewed By:

Enkelejda Miho, PhD, Professor, University of Applied Sciences and Arts Northwestern Switzerland, and Managing Director, aiNET

Coffee Break in the Exhibit Hall with Poster Viewing11:00

11:45

FEATURED PRESENTATION: Cell Line Development and Engineering Strategies for the Manufacture of Lentiviral Gene Therapy Viral Vectors

Ana Sofia Coroadinha, PhD, Lab Head, Health & Pharma Division, Animal Cell Technology Unit Cell Line Development and Molecular Biotechnology Lab, IBET

Advanced therapy medicinal products transformed medicine. Past unmet medical needs are now being treated with cell and gene therapy approaches targeting the cause of the disease. Gene therapies are based on the transfer of genetic material to the patients’ cells and lentiviral (LV) vectors are one of the preferred delivery systems, particularly when targeting hematopoietic cells (e.g., hematopoietic stem cells, T lymphocytes). LV vector engineering can be used to improve their efficacy and manufacturing. Herein, we describe cell and vector engineering strategies for LV manufacturing, namely, modifications enabling constitutive manufacturing through stable cell lines and effective T cell transduction.

12:15

Baculovirus-Free Expression of Virus-Like-Particles in Insect Cells for Antibody Development

Maren Schubert, PhD, Research Group Leader, Department of Biotechnology, Technical University of Braunschweig

The baculovirus expression vector system leads to high yields of Virus-like-Particles (VLPs). Yet, it´s time-intensive, inflexible in regard of protein ratios, its quality can be hampered by low cell viability, and last but not least, purification is challenging due to simultaneously produced baculoviral particles. The here-presented alternative of baculovirus-free VLP production in insect cells avoids the pitfalls of BEVS and produces VLPs in high quality and quantity for antibody development.

12:45 Harnessing Simplicity with the TheraPRO® CHO Media System

Josi Buerger, Associate Principal Scientist, R&D, Lonza Biologics

From generating high-producing clones to bulk drug substance with high protein titre and product quality.

The TheraPRO® CHO Media System builds on Lonza’s longstanding expertise in protein production and adds in one critical element: simplicity. In this talk we will introduce the TheraPRO® CHO platform and discuss the scientific challenges of developing a media system that maintains excellent product quality from clone construction through to industrial scale up.

Session Break13:15

13:20 LUNCHEON PRESENTATION:Best in Class Antiviral Antibodies from Cognate Recombinant Antibody Repertoires of Human Donors

Matthias Hillenbrand, PhD, Head, Infectious Disease Research & Biosafety Officer, Memo Therapeutics AG

Memo Therapeutics AG developed Dropzylla®, an antibody discovery platform based on a high-throughput conversion of B cell repertoires into immortalized mammalian-display biobanks. Deploying the platform, we identified potent nAbs from convalescent donors against BK virus and SARS-CoV-2, AntiBKV being in a pivotal phase II/III clinical trial with FDA fast-track designation. Currently, we work on the identification of a set of nAbs against HCMV, a severe threat to immunosuppressed individuals.

13:50 LUNCHEON PRESENTATION II:Strategies for Unveiling Optimal Bispecific Antibody Pairings

Julia Su, PhD, Associate Director of BD, Protein Sciences, WuXi Biologics

Bispecific antibody production presents challenges like heterogeneity, production complexity, and stability issues, which can impact the quality of the final product. This presentation will disclose the innovative strategies to address these challenges in drug development. It includes the initial small-scale high-throughput production of a vast number of bispecific antibodies in identifying optimal pairings, as well as later stage large-scale production. Real-world case studies will showcase the successful application of these strategies.

Session Break14:20

OVERCOMING EXPRESSION AND PRODUCTION CHALLENGES FOR UNIQUE PROTEINS

14:30

Chairperson's Remarks

Mercedes Márquez Martínez, PhD, Technical Coordinator & Acting Scientific Director, Protein Production Platform (PPP) – Nanbiosis, Autonomous, University of Barcelona (UAB)

14:35

KEYNOTE PRESENTATION: An Automated DNA Assembly Framework Enables Rapid and Scalable Plasmid Generation for Drug Discovery Applications

Robert G. Roth, PhD, Director, Protein Expression & Molecular Biology, Discovery Biology, R&D Biopharmaceuticals, AstraZeneca

Rapid and flexible construct generation at-scale is one of the most limiting first steps in the majority of drug discovery projects. The speed, quality, and cost of this process can be dramatically reduced by modular DNA design principles and automated fragmentation. To this end we have designed a robust, multi-module golden gate-based cloning platform for construct generation with a wide range of applications. In addition, to minimize timelines and cost for complex constructs, we developed a pipeline that performs fragmentation and codon optimization of long coding sequences in an automated manner.  

15:05

Host Comparative Production of Recombinant Proteins for Assembling as Nano- and Micro-Scale Materials for Drug Delivery

Mercedes Márquez Martínez, PhD, Technical Coordinator & Acting Scientific Director, Protein Production Platform (PPP) – Nanbiosis, Autonomous, University of Barcelona (UAB)

Recombinant proteins can be artificially self-assembled in the form of functional structures with promising applications in the field of drug delivery via nanostructured protein-only drugs. The antigenic RBD domain of the SARS-CoV-2 spike was produced in several expression systems such as bacteria, insect, and mammalian cells, to be used as a model to investigate the influence of the protein source in the production of nanoparticles and secretory microparticles. Both structures were generated in all cases but with different biophysical properties indicating thus that the selected expression system is a relevant factor for protein assembly into supramolecular structured and functional materials.

15:35 CLD Platform Optimization for Generation of High Titer CHO Cell Lines at Bioneer

Alexandra Baer, PhD, R&D Manager for Upstream Development of Mammalian Cells, Recombinant Proteins, Bioneer A/S

To strengthen our recombinant protein production capabilities Bioneer has established a cell line development platform in CHO DG44 cells. We have developed new expression vectors that show superior performance compared to commercial vector systems. Different product types like monoclonal antibodies, bispecific antibodies and enzymes have been expressed. Media and feed optimization boosted expression twofold resulting in yields of up to 6 g/L.

15:50 Solving a Key Challenge in Immunotherapeutic Production with the GS System®

Peter O'Callaghan, PhD, Head of Expression System Sciences (Biologics and Licensing), Lonza

Scientists and regulators across the globe have trusted the GS Gene Expression System® to expedite molecules to the clinic for over thirty years. With continuous market-driven innovation, the GS System® continues to keep pace with the evolving demands of biologics expression and production. In this talk, we present how Lonza’s state-of-the-art capabilities in CHO cell line engineering have paved the way for a new host cell line that achieves specific product quality attributes relevant to major therapy spaces, especially the oncology and autoimmune areas.

Refreshment Break in the Exhibit Hall with Poster Viewing16:05

17:00

Venom on Demand: Optimizing Snake Toxin Yield, Folding, and Purity in E. coli and P. pastoris with Biotinylation, Solubility, and Purification Tags

Esperanza Rivera de Torre, PhD, Assistant Professor, Center for Antibody Technologies, Department of Bioengineering, Technical University of Denmark

Animal venoms contain a plethora of biologically active toxins that have the potential to revolutionize antivenom development. However, producing functional toxins with the correct disulfide pattern is challenging. Our approach to engineering co-chaperon systems in Escherichia coli and leveraging Pichia pastoris' secretory capacity to produce natural and designed toxins. Our optimized strategies allow efficient toxin folding and purification, enabling downstream biotechnological applications, such as directed biotinylation for phage display.

17:30

Development of a Broadly Neutralizing Intranasal Anti-SARS-CoV2 Trimeric Sherpabody

Anna R. Makela, PhD, Senior Scientist, Department of Virology, University of Helsinki

There is a need for prophylactic SARS-CoV-2 blocking agents that are invulnerable to mutational viral variation and economical to produce. TriSb92 is a highly manufacturable and stable trimeric antibody-mimetic sherpabody targeted against a conserved region of the viral spike glycoprotein. TriSb92 potently neutralizes SARS-CoV-2, including the latest Omicron subvariants. In mice, intranasal administration of TriSb92 as early as 8 hours before but also 4 hours after SARS-CoV-2 challenge can protect from infection. Triggering of a conformational shift in the spike trimer was revealed as the inhibitory mechanism. TriSb92 could be useful as a nasal spray for protecting from SARS-CoV-2 infection.

18:00

Recombinant Protein L: Production, Purification and Characterization of a Universal Binding Ligand

Stefan Kittler, PhD, Postdoc Researcher, Institute of Chemical Environmental & Biological, TU Wien

Protein L (PpL) is a universal binding ligand that can be used for the detection and purification of antibodies and antibody fragments. Due to the unique interaction with immunoglobulin light chains, it differs from other affinity ligands, like protein A or G. However, due to its current higher market price, PpL is still scarce in applications. We investigated the recombinant production and purification of PpL and characterized the product in detail. In my talk I present a comprehensive roadmap for the production of the versatile protein PpL in E. coli.

18:30 PANEL DISCUSSION:

Protein Production Lab Challenges: Methodologies, Strategies, and the Art of Expressing Recombinant Proteins

PANEL MODERATOR:

Richard Altman, MS, Field Application Scientist, Life Science Solutions, Thermo Fisher Scientific

Protein expression laboratories provide crucial support to drug discovery efforts.  This panel discussion will focus on the concepts, technologies, and strategies necessary to meet the ever-increasing need for recombinant proteins.       ​

  • Know your protein!         
  • Strategies on how to manage multiple “top priority” projects        
  • Total workflow efficiency         
  • The importance of tech development to long term success       
  • Troubleshooting strategies or how much time should be spent before moving to the next option?
PANELISTS:

Nicola Burgess-Brown, PhD, Director of Enzymology and Protein Engineering, Exact Sciences Innovation

Peter Schmidt, Director Protein Biochemistry, CSL Research, Melbourne, Australia

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

Close of Optimising Expression Platforms Conference19:00

Recommended Short Course*
Monday, 13 November, 14:00 – 17:00
SC4: The Use and Optimization of Eukaryotic Expression Systems to Support Therapeutic Generation and Structural Biology
*Separate registration required. See short courses page for details. All short courses take place in-person only.