Pharmaceutical protein production by yeast: towards production of human blood proteins by microbial fermentation

https://doi.org/10.1016/j.copbio.2012.03.011Get rights and content

Since the approval of recombinant insulin from Escherichia coli for its clinical use in the early 1980s, the amount of recombinant pharmaceutical proteins obtained by microbial fermentations has significantly increased. The recent advances in genomics together with high throughput analysis techniques (the so-called  omics approaches) and integrative approaches (systems biology) allow the development of novel microbial cell factories as valuable platforms for large scale production of therapeutic proteins. This review summarizes the main achievements and the current situation in the field of recombinant therapeutics using yeast Saccharomyces cerevisiae as a model platform, and discusses the future potential of this platform for production of blood proteins and substitutes.

Highlights

► Recombinant therapeutic protein production is a multibillion dollar market. ► E. coli comprises 30% of recombinant protein production but not suitable for human therapeutics. ► Eukaryotic systems other than yeast are costly or not so efficient regarding protein yields. ► S. cerevisiae shows a high potential to be a suitable platform for therapeutic proteins. ► Human blood proteins are the next candidates to be challenged by S. cerevisiae system.

Introduction

Microorganisms have been extensively used since ancient times for the production of fermented food and beverages, thousands of years before the actual nature of the fermentative processes was known. In the early 20th century the production of citric acid based on microbial fermentation was initiated as the first large scale fermentation product and this was followed by industrial production of penicillin as the first antibiotic. Introduction of the genetic engineering in the 1970s paved the way for the establishment and development of the current biotech industry, allowing the commercial production of industrial enzymes and biopharmaceutical proteins. In 1980, the FDA approved for clinical use the recombinant insulin obtained from Escherichia coli, becoming the first recombinant pharmaceutical protein to enter the market [1••]. Since then, the biotechnology industry has grown substantially, and currently about 25% of commercial pharmaceuticals are biopharmaceuticals [2] with 2010 sales exceeding USD100 billions [3]. About half of the world-wide sales are in the USA with monoclonal antibodies representing the majority (>USD18 billions) followed by hormones (USD11 billions) and growth factors (>USD10 billions) [4]. Together with the production of industrial enzymes, the recombinant protein production market is expected to rise to 169 billion dollars in 2014 [3] (Figure 1).

Section snippets

Platforms for production of pharmaceutical proteins

Industrial biotechnology has traditionally used numerous bacterial and eukaryal cells as production platforms, with the main criterion for host selection being the ability to produce the desired compound. However, with the advent of genetic engineering it became possible to introduce heterologous genes and create new traits in non-natural producers, allowing the development of cell factories for the production of chemicals through metabolic engineering. E. coli was the earliest platform to be

How to make S. cerevisiae a better producer of pharmaceutical proteins?

The technology for industrial production of recombinant pharmaceutical proteins in S. cerevisiae is well established and currently applied for production of human insulin, hepatitis virus vaccines and human papilloma virus vaccines, and its potential to be used for large scale production of many other proteins in the forthcoming years is therefore high. Furthermore, the advent of systems biology allowing global metabolism analysis and the application of so-called ‘omics’ approaches such as

Production of recombinant human blood proteins

Among the 58 biopharmaceuticals approved in the United States and/or Europe from 2006 to 2010 four are blood related proteins, including a rh coagulation factor VIII produced in CHO cells, a rh antithrombin from milk of transgenic goats, a plasma kallikrein inhibitor produced in Pichia pastoris, and a rh thrombin produced in CHO cells [27]. All have therapeutic use for treatment of hemophilia. To date, most of the recombinant blood related biopharmaceuticals approved for clinical treatment are

S. cerevisiae as a cell factory for human hemoglobin production

All the proteins described above are blood plasma components which contribute to different roles of blood such as coagulation, clotting, transport of iron, maintain blood osmotic pressure, and blood volume. The additional crucial role of blood is the transport of oxygen and the only component in blood that possesses oxygen carrier function is hemoglobin (found in erythrocytes), and this is therefore a key component for development of human blood substitutes for treatment of patients with

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We would like to thank Zihe Liu and Dr. Jin Hou for suggestions and comments on the manuscript. This work has been funded by the Chalmers Foundation, European Research Council project INSYSBIO (Grant no. 247013) and the Novo Nordisk Foundation.

References (67)

  • M.D. Kim et al.

    Production of antithrombotic hirudin in GAL1-disrupted Saccharomyces cerevisiae

    Appl Microbiol Biotechnol

    (2004)
  • D. Ning et al.

    Expression, purification, and characterization of humanized anti-HBs Fab fragment

    J Biochem

    (2003)
  • R. Fleer et al.

    High-level secretion of correctly processed recombinant human interleukin-1 beta in Kluyveromyces lactis

    Gene

    (1991)
  • K. Kobayashi et al.

    High-level expression of recombinant human serum albumin from the methylotrophic yeast Pichia pastoris with minimal protease production and activation

    J Biosci Bioeng

    (2000)
  • C. Gurramkonda et al.

    Application of simple fed-batch technique to high-level secretory production of insulin precursor using Pichia pastoris with subsequent purification and conversion to human insulin

    Microb Cell Fact

    (2010)
  • N. Ferrer-Miralles et al.

    Microbial factories forrecombinant pharmaceuticals

    Microb Cell Fact

    (2009)
  • E.-R. Redwan

    Cumulative updating of approved biopharmaceuticals

    Hum Antibodies

    (2007)
  • M. Goodman

    Market watch: sales of biologics to show robust growth through to 2013

    Nat Rev Drug Discov

    (2009)
  • S. Aggarwal

    What's fueling the biotech engine  2010 to 2011

    Nat Biotechnol

    (2011)
  • K. Terpe

    Overview of bacterial expression systems for heterologous protein production: from molecular and biochemical fundamentals to commercial systems

    Appl Microbiol Biotechnol

    (2006)
  • A.K. Panda

    Bioprocessing of therapeutic proteins from the inclusion bodies of Escherichia coli

    Adv Biochem Eng Biotechnol

    (2003)
  • D. Porro et al.

    Production of recombinant proteins and metabolites in yeasts: when are these systems better than bacterial production systems?

    Appl Microbiol Biotechnol

    (2011)
  • N.K. Tripathi et al.

    PVL: high yield production of heterologous proteins with Escherichia coli

    Defence Sci J

    (2009)
  • G. Walsh et al.

    Post-translational modifications in the context of therapeutic proteins

    Nat Biotechnol

    (2006)
  • K. De Pourcq et al.

    Engineering of glycosylation in yeast and other fungi: current state and perspectives

    Appl Microbiol Biotechnol

    (2010)
  • R.P. Nolan et al.

    Dynamic model for CHO cell engineering

    J Biotechnol

    (2012)
  • K.J. Morrow

    Improving protein production processes

    Gen Eng News

    (2007)
  • M.M. Cox

    Recombinant protein vaccines produced in insect cells

    Vaccine

    (2012)
  • A.L. Demain et al.

    Production of recombinant proteins by microbes and higher organisms

    Biotechnol Adv

    (2009)
  • Y. Chigira et al.

    Engineering of a mammalian O-glycosylation pathway in the yeast Saccharomyces cerevisiae: production of O-fucosylated epidermal growth factor domains

    Glycobiology

    (2008)
  • A. Graf et al.

    Yeast systems biotechnology for the production of heterologous proteins

    FEMS Yeast Res

    (2009)
  • I.K. Kim et al.

    A systems-level approach for metabolic engineering of yeast cell factories

    FEMS Yeast Res

    (2012)
  • A. Krivoruchko et al.

    Opportunities for yeast metabolic engineering: lessons from synthetic biology

    Biotechnol J

    (2011)
  • Cited by (67)

    • Microbiology of biofuels: Cultivating the future

      2022, Relationship between Microbes and the Environment for Sustainable Ecosystem Services: Microbial Tools for Sustainable Ecosystem Services: Volume 3
    • Biotechnology applications in infectious disease

      2022, Biotechnology in Healthcare, Volume 2: Applications and Initiatives
    • Trichoderma-functional metabolomics to genetic engineering

      2020, New and Future Developments in Microbial Biotechnology and Bioengineering: Recent Developments in Trichoderma Research
    View all citing articles on Scopus
    3

    These authors contributed equally to this article.

    View full text