Engineering Aggregation Resistance in IgG by Two Independent Mechanisms: Lessons from Comparison of Pichia pastoris and Mammalian Cell Expression

https://doi.org/10.1016/j.jmb.2012.01.027Get rights and content

Abstract

Aggregation is an important concern for therapeutic antibodies, since it can lead to reduced bioactivity and increase the risk of immunogenicity. In our analysis of immunoglobulin G (IgG) molecules of identical amino acid sequence but produced either in mammalian cells (HEK293) or in the yeast Pichia pastoris (PP), dramatic differences in their aggregation susceptibilities were encountered. The antibodies produced in Pichia were much more resistant to aggregation under many conditions, a phenomenon found to be mainly caused by two factors. First, the mannose-rich glycan of the IgG from Pichia, while slightly thermally destabilizing the IgG, strongly inhibited its aggregation susceptibility, compared to the complex mammalian glycan. Second, on the Pichia-produced IgGs, amino acids belonging to the α-factor pre-pro sequence were left at the N-termini of both chains. These additional residues proved to considerably increase the temperature of the onset of aggregation and reduced the aggregate formation after extended incubation at elevated temperatures. The attachment of these residues to IgGs produced in cell culture confirmed their beneficial effect on the aggregation resistance. Secretion of IgGs with native N-termini in the yeast system became possible after systematic engineering of the precursor proteins and the processing site. Taken together, the present results will be useful for the successful production of full-length IgGs in Pichia, give indications on how to engineer aggregation-resistant IgGs and shed new light on potential biophysical effects of tag sequences in general.

Graphical Abstract

Research Highlights

Pichia-produced IgGs are more aggregation resistant than IgGs made in human embryonic kidney cells. ► Two causes are as follows: different glycans and remains from incomplete processing of pro-region. ► Four N-terminal residues added to human embryonic kidney-produced IgG increase aggregation resistance. ► Results have defined IgG constructs that generate authentic N-termini in Pichia. ► Results help rationalize antibody engineering toward aggregation resistance.

Introduction

Antibodies and their derivatives have found a broad range of applications, from basic research to medical therapy. With the use of many different techniques for their generation,1 several therapeutic immunoglobulins, usually in the whole immunoglobulin G (IgG) format, have been approved by the Food and Drug Administration and applied to the treatment of a wide range of diseases.2, 3, 4 One major challenge encountered in the development of antibody-based therapeutics, however, is their aggregation susceptibility under both formulation conditions5, 6 and in vivo, where aggregation is involved in a significant degradation pathway for monoclonal antibodies.7 Irreversible aggregation thereby leads to reduced bioactivity8 and presents an increased risk of immunogenicity.9, 10, 11, 12, 13, 14 Aggregation of even only a few percent of the administered therapeutic antibody can cause severe problems, as already low amounts of aggregates have long been known to cause anaphylactoid reactions.15, 16 Therefore, engineering IgG to reduce or eliminate its self-association is of great importance and might further extend the potential of therapeutic antibodies.

Since nearly all recombinant therapeutic antibodies are expressed in the IgG format, choices must be made in the molecular composition and expression system. Currently, all approved therapeutic antibodies are produced in mammalian cells,17 since only this expression system can introduce the complex glycosylation that is necessary for the interaction of the antibodies with some of the activating Fcγ receptors (FcγR).18 IgGs normally possess a single N-linked glycosylation at Asn297 in the CH2 domain, maintaining these domains at a critical distance to bind to the FcγR.

In some cases, however, this FcγR interaction is neither needed nor wanted, thus opening the range of possible production hosts also to Pichia pastoris (PP). In the past, this methylotrophic yeast has been widely used for the expression of both secreted and intracellular eukaryotic proteins.19 While it is able to carry out efficient disulfide bond formation and isomerization using the eukaryotic secretion quality control machinery,19, 20, 21 Pichia expression generates mannose-rich glycosylation of IgG.22 The attached glycan moieties differ markedly from the product of mammalian cells, resulting in antibodies that lack FcγR binding capacity. However, recently, Pichia has also been engineered to introduce complex, human-like glycosylation.23, 24, 25, 26

Due to its respiratory growth, Pichia can grow to very high cell densities27 and therefore express high levels of recombinant protein through efficient and tightly regulated promoters, accounting for up to 30% of the total cell protein.28 Moreover, comparatively few endogenous proteins are secreted by this yeast system, which offers further advantages for subsequent protein purification and downstream processing.29 Since Ridder et al. reported the first successful production of functional antibody fragments in Pichia,30 more than 50 reports describing antibody expression in this system have been published.31 While most of these molecules were single-chain variable fragments and fusions thereof, several Fab fragments have been produced in Pichia as well.32, 33, 34 Interestingly, until today, Pichia has only been used as an expression host for a handful of full-length IgGs.25, 26, 35

Almost all heterologous proteins produced in Pichia have been expressed under the control of the methanol-inducible alcohol oxidase 1 (AOX1) promoter.20, 36 However, throughout this study, the constitutive glyceraldehyde-3-phosphate dehydrogenase (GAP) promoter37 was used, since the ability to compare mutant proteins for their expression and aggregation tendencies was crucial. The GAP promoter is usually considered to result in lower protein yields compared to AOX1,38 even though some reports describe the GAP promoter to be superior to AOX1 for certain proteins,39, 40 including Fab fragments.41 A potential reason could be the lower rate of expression that might better match the corresponding folding rate and, thus, reduces the stress level for the cells.

Previously, our laboratory analyzed sequence modifications in the variable domain in various antibody formats for aggregation propensity and conformational stability.42, 43, 44 Eukaryotic expression systems, where functional expression yields can be compared and mutant proteins can be isolated and biophysically characterized, are required to carry out such studies for whole IgGs. Therefore, it is vital that isogenic strains can be produced, only differing by the sequence of the protein to be tested. As a first step, we investigated homologous recombination in HEK293 cells45 and Pichia. As Pichia does not maintain episomal vectors, the expression cassettes are generally designed for integration into the yeast genome, thus leading to stable cell lines expressing the protein of interest (POI). In initial experiments, we were very surprised to find large differences between the aggregation behaviors of purified IgG protein from both systems, even though their amino acid sequence was identical: the same recombinant IgG expressed in HEK293 cells was much more aggregation prone than its counterpart from Pichia.

Here, we describe our investigations of this phenomenon and pinpoint it to differences in the processing of the precursor proteins, as well as to differences between the unequal glycosylations. As an initial model system, an antibody that had originally been selected from the HuCAL library46 by panning against myoglobin from horse skeletal muscle and subsequently been stability engineered was used.47 For this study, it was converted to a full-length IgG and investigated for its processing and expression in mammalian HEK293 cells and in the yeast Pichia pastoris.

Section snippets

Comparison of various IgG constructs

In this study, we compared IgGs of identical amino acid sequence but produced in different eukaryotic expression systems. Unexpectedly, these molecules revealed dramatic differences in some of their biophysical properties. To monitor changes in their structure as a function of temperature, we analyzed purified IgGs by circular dichroism (CD). Since the signals stemming from the IgG's β-sheets and random coil essentially canceled out to zero at 208 nm, thermal denaturation of the IgG molecules

Discussion

When IgG molecules of identical amino acid sequence were produced in different eukaryotic systems, dramatic differences in their aggregation susceptibilities were encountered. This observation turned out to be due to two major factors: (i) differences in the glycan structures and (ii) residues remaining from the Pichia secretion system used, the α-factor pre-pro sequence (αMFpp).

Conclusions

Taken together, our results indicate that the production of full-length IgG in the yeast Pichia—being attractive for large-scale preparations due to its convenient handling—is possible and results in molecules with the desired antigen binding and thermal stability. However, obtaining correctly processed IgGs requires the direct fusion of a signal sequence to the heavy chain. When using the commonly utilized α-factor pre-pro secretion system, the secreted antibodies will contain the EAEA

Materials and cultivation of mammalian and Pichia cells

All media and supplements for mammalian expression were purchased either from Sigma-Aldrich (Missouri, USA), Invitrogen (California, USA) or Amimed (BioConcept, Switzerland). The antibiotics that were used, Zeocin™ and Hygromycin B, were bought from Invitrogen and PAA (Austria), respectively. All solutions used were sterilized by filtration through 0. 22-μm filters (Millipore, Massachusetts, USA). Stably transformed HEK293 cells were maintained in Dulbecco's modified Eagle's medium (DMEM;

Acknowledgements

We want to thank Dr. I. Jelezarov, Dr. P. Gimeson, Dr. B. Dreier for their help with the DSC and MALS analyses. We are grateful to the personnel of the Functional Genomics Center at University of Zurich for their help with MS analyses and protein sequencing. Our thanks are also due to Dr. S. Duhr from NanoTemper, Germany, for his help and the chance to determine affinities by MST. The polyclonal rabbit αMFpp antiserum was a kind gift of Dr. R. Schekman (University of California, Berkeley). We

References (93)

  • WaterhamH.R. et al.

    Isolation of the Pichia pastoris glyceraldehyde-3-phosphate dehydrogenase gene and regulation and use of its promoter

    Gene

    (1997)
  • KnappikA. et al.

    Fully synthetic human combinatorial antibody libraries (HuCAL) based on modular consensus frameworks and CDRs randomized with trinucleotides

    J. Mol. Biol.

    (2000)
  • HarrisR.J.

    Processing of C-terminal lysine and arginine residues of proteins isolated from mammalian cell culture

    J. Chromatogr., A

    (1995)
  • KaplanA.P. et al.

    Amino terminal sequences of human immunoglobulin heavy chains

    Immunochemistry

    (1971)
  • KurjanJ. et al.

    Structure of a yeast pheromone gene (MFα): a putative α-factor precursor contains four tandem copies of mature α-factor

    Cell

    (1982)
  • EmbersonL.M. et al.

    Expression of an anti-CD33 single-chain antibody by Pichia pastoris

    J. Immunol. Methods

    (2005)
  • GarberE. et al.

    A broad range of Fab stabilities within a host of therapeutic IgGs

    Biochem. Biophys. Res. Commun.

    (2007)
  • RothJ. et al.

    Protein N-glycosylation, protein folding, and protein quality control

    Mol. Cell

    (2010)
  • FeigeM.J. et al.

    Folding mechanism of the CH2 antibody domain

    J. Mol. Biol.

    (2004)
  • FeigeM.J. et al.

    How antibodies fold

    Trends Biochem. Sci.

    (2010)
  • KrappS. et al.

    Structural analysis of human IgG-Fc glycoforms reveals a correlation between glycosylation and structural integrity

    J. Mol. Biol.

    (2003)
  • HulettM.D. et al.

    Identification of the IgG binding site of the human low affinity receptor for IgG FcγRII. Enhancement and ablation of binding by site-directed mutagenesis

    J. Biol. Chem.

    (1994)
  • FerraraC. et al.

    The carbohydrate at FcγRIIIa Asn-162. An element required for high affinity binding to non-fucosylated IgG glycoforms

    J. Biol. Chem.

    (2006)
  • CereghinoJ.L. et al.

    Heterologous protein expression in the methylotrophic yeast Pichia pastoris

    FEMS Microbiol. Rev.

    (2000)
  • MonsalveR.I. et al.

    Expressions of recombinant venom allergen, antigen 5 of yellowjacket (Vespula vulgaris) and paper wasp (Polistes annularis), in bacteria or yeast

    Protein Expression Purif.

    (1999)
  • ZseboK.M. et al.

    Protein secretion from Saccharomyces cerevisiae directed by the prepro-α-factor leader region

    J. Biol. Chem.

    (1986)
  • KjeldsenT. et al.

    A removable spacer peptide in an α-factor-leader/insulin precursor fusion protein improves processing and concomitant yield of the insulin precursor in Saccharomyces cerevisiae

    Gene

    (1996)
  • LiuY.D. et al.

    N-terminal glutamate to pyroglutamate conversion in vivo for human IgG2 antibodies

    J. Biol. Chem.

    (2011)
  • VinckeC. et al.

    General strategy to humanize a camelid single-domain antibody and identification of a universal humanized nanobody scaffold

    J. Biol. Chem.

    (2009)
  • WernerW.E. et al.

    The removal of pyroglutamic acid from monoclonal antibodies without denaturation of the protein chains

    Anal. Biochem.

    (2005)
  • PlückthunA. et al.

    Modern antibody technology: the impact on drug development

  • CarterP.J.

    Potent antibody therapeutics by design

    Nat. Rev., Immunol.

    (2006)
  • FontouraP.

    Monoclonal antibody therapy in multiple sclerosis: paradigm shifts and emerging challenges

    mAbs

    (2010)
  • DemarestS.J. et al.

    Antibody therapeutics, antibody engineering, and the merits of protein stability

    Curr. Opin. Drug Discov. Dev

    (2008)
  • Di PaoloC. et al.

    A recombinant immunotoxin derived from a humanized epithelial cell adhesion molecule-specific single-chain antibody fragment has potent and selective antitumor activity

    Clin. Cancer Res.

    (2003)
  • BraunA. et al.

    Protein aggregates seem to play a key role among the parameters influencing the antigenicity of interferon alpha (IFN-α) in normal and transgenic mice

    Pharm. Res.

    (1997)
  • HermelingS. et al.

    Structure–immunogenicity relationships of therapeutic proteins

    Pharm. Res.

    (2004)
  • SchellekensH.

    Factors influencing the immunogenicity of therapeutic proteins

    Nephrol., Dial., Transplant.

    (2005)
  • RosenbergA.S.

    Effects of protein aggregates: an immunologic perspective

    AAPS J.

    (2006)
  • KesslerM. et al.

    Immunogenicity of biopharmaceuticals

    Nephrol., Dial., Transplant.

    (2006)
  • RingJ. et al.

    Anaphylactoid reactions due to non-immune complex serum protein aggregates

    Monogr. Allergy

    (1977)
  • RingJ. et al.

    Anaphylactoid reactions to infusions of plasma protein and human serum albumin. Role of aggregated proteins and of stabilizers added during production

    Clin. Allergy

    (1979)
  • FaridS.S.

    Established bioprocesses for producing antibodies as a basis for future planning

    Adv. Biochem. Eng./Biotechnol.

    (2006)
  • ArnoldJ.N. et al.

    The impact of glycosylation on the biological function and structure of human immunoglobulins

    Annu. Rev. Immunol.

    (2007)
  • BretthauerR.K. et al.

    Glycosylation of Pichia pastoris-derived proteins

    Biotechnol. Appl. Biochem.

    (1999)
  • JacobsP.P. et al.

    Engineering complex-type N-glycosylation in Pichia pastoris using GlycoSwitch technology

    Nat. Protoc.

    (2009)
  • Cited by (0)

    View full text