Abstract

Fed-batch cultures of Chinese Hamster Ovary cells have been used to produce high quantities of biotherapeutics, particularly monoclonal antibodies. However, a growing number of next-generation biotherapeutics, such as bi-specific antibodies and fusion proteins, are difficult to express using standard fed-batch processes. Decoupling cell growth and biotherapeutic production is becoming an increasingly desired strategy for the biomanufacturing industry, especially for difficult-to-express products. Cells are grown to a high cell density in the absence of recombinant protein production (the growth phase), then expression of the recombinant protein is induced and cell proliferation halted (the production phase), usually by combining an inducible gene expression system with a proliferation control strategy. Separating the growth and production phases allows cell resources to be more efficiently directed toward either growth or production, improving growth characteristics and enhancing the production of difficult to express proteins. However, current mammalian cell proliferation control methods rely on temperature shifts and chemical agents, which interact with many non-proliferation pathways, leading to variable impacts on product quality and culture viability. Synthetic biology offers an alternative approach by strategically targeting proliferation pathways to arrest cell growth but have largely remained unused in industrial bioproduction. Due to recent developments in microbial decoupling systems and advances in available mammalian cell engineering tools, we propose that the synthetic biology approach to decoupling growth and production needs revisiting.

Highlights

  • Industrial-scale production of many biotherapeutics relies on the culture of Chinese Hamster Ovary (CHO) cells

  • With the increasing demands on biomanufacturing facilities to produce a wider range of protein biotherapeutics at higher titers and in a shorter period of time, decoupling growth and production provides a viable alternative to traditional constitutive expression strategies

  • Moving the product expression window away from the growth phase mitigates the negative impact that cytotoxic proteins have on cell growth, viability, and stability

Read more

Summary

INTRODUCTION

Industrial-scale production of many biotherapeutics relies on the culture of Chinese Hamster Ovary (CHO) cells. An increasingly attractive method of production is to decouple the growth and production phases of the culture process, usually by combining an inducible gene expression system with a proliferation control strategy (Misaghi et al, 2014; Lam et al, 2017; Poulain et al, 2017). It has been over 20 years since the overexpression of cyclindependent kinase inhibitors (p21Cip, p27K ip, and p53) was first used to decouple growth and production in mammalian cells (Fussenegger et al, 1997) Implementation of this strategy was limited by a lack of appropriate inducible gene expression systems and its weak impact on mammalian cell proliferation (Weber and Fussenegger, 2007). Activation of a caffeine-inducible mammalian protein kinase R switches off non-product protein translation, while a viral IRES sequence protects recombinant protein

Method
DISCUSSION
DATA AVAILABILITY STATEMENT
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call