Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
J Biotechnol ; 208: 13-21, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26015261

ABSTRACT

Monoclonal antibody (mAb) therapy has been successfully used for the treatment of B-cell lymphomas and is currently extended for the treatment of multiple myeloma (MM). New developments in MM therapeutics have achieved significant survival gains in patients but the disease still remains incurable. Elotuzumab (HuLuc63), an anti-CS1 monoclonal IgG1 antibody, is believed to induce anti-tumor activity and MM cytotoxicity through antibody dependent cellular cytotoxicity (ADCC) and inhibition of MM cell adhesion to bone marrow stromal cells (BMSCs). Modulations of the Fc glycan composition at the N297 site by selective mutations or afucosylation have been explored as strategies to develop bio-better therapeutics with enhanced ADCC activity. Afucosylated therapeutic antibodies with enhanced ADCC activity have been reported to possess greater efficacy in tumor growth inhibition at lower doses when compared to fucosylated therapeutic antibodies. The N-linked glycosylation pathway in Pichia pastoris has been engineered to produce human-like N-linked glycosylation with uniform afucosylated complex type glycans. The purpose of this study was to compare afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris with fucosylated anti-CS1 mAb expressed in mammalian HEK293 cells through in vitro ADCC and in vivo tumor inhibition models. Our results indicate that Fc glycosylation is critical for in vivo efficacy and afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris shows a better in vivo efficacy in tumor regression when compared to fucosylated anti-CS1 mAb expressed in HEK293 cells. Glycoengineered Pichia pastoris could provide an alternative platform for generating homogeneous afucosylated recombinant antibodies where Fc mediated immune effector function is important for efficacy.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neoplasm , Cell Engineering , Multiple Myeloma/drug therapy , Neoplasms, Experimental/drug therapy , Pichia , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/genetics , Cell Line, Tumor , Glycosylation , HEK293 Cells , Humans , Mice, SCID , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Xenograft Model Antitumor Assays
2.
Appl Environ Microbiol ; 81(1): 260-71, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25344235

ABSTRACT

With a completely reengineered and humanized glycosylation pathway, glycoengineered Pichia pastoris has emerged as a promising production host for the manufacture of therapeutic glycoproteins. However, the extensive genetic modifications have also negatively affected the overall fitness levels of the glycoengineered host cells. To make glycoengineered Pichia strains more compatible with a scalable industrial fermentation process, we sought to identify genetic solutions to broadly improve cell robustness during fermentation. In this study, we report that mutations within the Pichia pastoris ATT1 (PpATT1) gene (a homolog of the Saccharomyces cerevisiae GAL4 [ScGAL4] transcriptional activator) dramatically increased the cellular fitness levels of glycoengineered Pichia strains. We demonstrate that deletion of the PpATT1 gene enabled glycoengineered Pichia strains to improve their thermal tolerance levels, reduce their cell lysis defects, and greatly improve fermentation robustness. The extension of the duration of fermentation enabled the PpATT1-modified glycoengineered Pichia strains to increase their product yields significantly without any sacrifice in product quality. Because the ATT1 gene could be deleted from any Pichia strains, including empty hosts and protein-expressing production strains alike, we suggest that the findings described in this study are broadly applicable to any Pichia strains used for the production of therapeutic proteins, including monoclonal antibodies, Fc fusions, peptides, hormones, and growth factors.


Subject(s)
Gene Deletion , Gene Expression Regulation, Fungal , Metabolic Engineering , Pichia/genetics , Pichia/metabolism , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , Glycosylation , Microbial Viability , Pichia/growth & development , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology , Transcription Factors/genetics , Transcription, Genetic , Virulence
3.
Biotechnol Prog ; 30(6): 1488-96, 2014.
Article in English | MEDLINE | ID: mdl-25196297

ABSTRACT

Human serum albumin (HSA) is a cysteine rich molecule that is most abundant in human blood plasma. To remain viable in the market due to lower marketing costs for HSA, it is important to produce a large quantity in an economical manner by recombinant technology. The objective of this study was to maximize recombinant HSA (rHSA) production using a Mut(s) Pichia pastoris strain by fermentation process optimization. We evaluated the impact of process parameters on the production of rHSA, including induction cell density (wet cell weight, g/L) and the control of specific growth rate at induction. In this study, we demonstrated that induction cell density is a critical factor for high level production of rHSA under controlled specific growth rate. We observed higher specific productivities at higher induction cell densities (285 g/L) and at lower specific growth rates (0.0022-0.0024/h) during methanol induction phase, and achieved the broth titer of rHSA up to 10 g/L. The temperature shift from 24 to 28(o) C was effective to control the specific growth rate at low level (≤0.0024/h) during methanol induction phase while maintaining high specific productivity [0.0908 mgrHSA /(gwcw h)].


Subject(s)
Biotechnology/methods , Pichia/genetics , Recombinant Proteins/metabolism , Serum Albumin/metabolism , Gene Knockout Techniques , Humans , Phenotype , Pichia/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Serum Albumin/chemistry , Serum Albumin/genetics
4.
Curr Opin Biotechnol ; 30: 120-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25014890

ABSTRACT

The selection of an expression platform for recombinant biopharmaceuticals is often centered upon suitable product titers and critical quality attributes, including post-translational modifications. Although notable differences between microbial, yeast, plant, and mammalian host systems exist, recent advances have greatly mitigated any inherent liabilities of yeasts. Yeast expression platforms are important to both the supply of marketed biopharmaceuticals and the pipelines of novel therapeutics. In this review, recent advances in yeast-based expression of biopharmaceuticals will be discussed. The advantages of using glycoengineered yeast as a production host and in the discovery space will be illustrated. These advancements, in turn, are transforming yeast platforms from simple production systems to key technological assets in the discovery and selection of biopharmaceutical lead candidates.


Subject(s)
Biological Products/metabolism , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Technology, Pharmaceutical/methods , Yeasts/metabolism , Animals , Glycosylation , Plants/metabolism , Protein Processing, Post-Translational , Recombinant Proteins/genetics , Saccharomyces cerevisiae/genetics , Yeasts/genetics
5.
Appl Microbiol Biotechnol ; 98(6): 2573-83, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24526360

ABSTRACT

Yeast are important production platforms for the generation of recombinant proteins. Nonetheless, their use has been restricted in the production of therapeutic proteins due to differences in their glycosylation profile with that of higher eukaryotes. The yeast strain Pichia pastoris is an industrially important organism. Recent advances in the glycoengineering of this strain offer the potential to produce therapeutic glycoproteins with sialylated human-like N- and O-linked glycans. However, like higher eukaryotes, yeast also express numerous proteases, many of which are either localized to the secretory pathway or pass through it en route to their final destination. As a consequence, nondesirable proteolysis of some recombinant proteins may occur, with the specific cleavage being dependent on the class of protease involved. Dipeptidyl aminopeptidases (DPP) are a class of proteolytic enzymes which remove a two-amino acid peptide from the N-terminus of a protein. In P. pastoris, two such enzymes have been identified, Ste13p and Dap2p. In the current report, we demonstrate that while the knockout of STE13 alone may protect certain proteins from N-terminal clipping, other proteins may require the double knockout of both STE13 and DAP2. As such, this understanding of DPP activity enhances the utility of the P. pastoris expression system, thus facilitating the production of recombinant therapeutic proteins with their intact native sequences.


Subject(s)
Biological Products/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Gene Knockout Techniques , Peptides/metabolism , Pichia/enzymology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Peptides/genetics , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
J Mass Spectrom ; 48(12): 1308-17, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24338886

ABSTRACT

Glycosylation plays a critical role in the in vivo efficacy of both endogenous and recombinant erythropoietin (EPO). Using mass spectrometry, we characterized the N-/O-linked glycosylation of recombinant human EPO (rhEPO) produced in glycoengineered Pichia pastoris and compared with the glycosylation of Chinese hamster ovary (CHO) cell-derived rhEPO. While the three predicted N-linked glycosylation sites (Asn24, Asn38 and Asn83) showed complete site occupancy, Pichia- and CHO-derived rhEPO showed distinct differences in the glycan structures with the former containing sialylated bi-antennary glycoforms and the latter containing a mixture of sialylated bi-, tri- and tetra-antennary structures. Additionally, the N-linked glycans from Pichia-produced rhEPO were similar across all three sites. A low level of O-linked mannosylation was detected on Pichia-produced rhEPO at position Ser126, which is also the O-linked glycosylation site for endogenous human EPO and CHO-derived rhEPO. In summary, the mass spectrometric analyses revealed that rhEPO derived from glycoengineered Pichia has a highly uniform bi-antennary N-linked glycan composition and preserves the orthogonal O-linked glycosylation site present on endogenous human EPO and CHO-derived rhEPO.


Subject(s)
Erythropoietin/chemistry , Pichia/metabolism , Recombinant Proteins/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Erythropoietin/analysis , Erythropoietin/metabolism , Humans , Metabolic Engineering , Pichia/genetics , Recombinant Proteins/analysis , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
Glycobiology ; 23(10): 1192-203, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23893788

ABSTRACT

The methylotrophic yeast, Pichia pastoris, is an important organism used for the production of therapeutic proteins. Previously, we have reported the glycoengineering of this organism to produce human-like N-linked glycans but up to now no one has addressed engineering the O-linked glycosylation pathway. Typically, O-linked glycans produced by wild-type P. pastoris are linear chains of four to five α-linked mannose residues, which may be capped with ß- or phospho-mannose. Previous genetic engineering of the N-linked glycosylation pathway of P. pastoris has eliminated both of these two latter modifications, resulting in O-linked glycans which are linear α-linked mannose structures. Here, we describe a method for the co-expression of an α-1,2-mannosidase, which reduces these glycans to primarily a single O-linked mannose residue. In doing so, we have reduced the potential of these glycans to interact with carbohydrate-binding proteins, such as dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin. Furthermore, the introduction of the enzyme protein-O-linked-mannose ß-1,2-N-acetylglucosaminyltransferase 1, resulted in the capping of the single O-linked mannose residues with N-acetylglucosamine. Subsequently, this glycoform was extended into human-like sialylated glycans, similar in structure to α-dystroglycan-type glycoforms. As such, this represents the first example of sialylated O-linked glycans being produced in yeast and extends the utility of the P. pastoris production platform beyond N-linked glycosylated biotherapeutics to include molecules possessing O-linked glycans.


Subject(s)
Mannose/biosynthesis , Metabolic Engineering/methods , Pichia/metabolism , alpha-Mannosidase/metabolism , Pichia/growth & development , Protein Engineering , alpha-Mannosidase/genetics
8.
PLoS One ; 8(7): e70190, 2013.
Article in English | MEDLINE | ID: mdl-23875020

ABSTRACT

State-of-the-art monoclonal antibody (mAb) discovery methods that utilize surface display techniques in prokaryotic and eukaryotic cells require multiple steps of reformatting and switching of hosts to transition from display to expression. This results in a separation between antibody affinity maturation and full-length mAb production platforms. Here, we report for the first time, a method in Glyco-engineered Pichiapastoris that enables simultaneous surface display and secretion of full-length mAb molecules with human-like N-glycans using the same yeast cell. This paradigm takes advantage of homo-dimerization of the Fc portion of an IgG molecule to a surface-anchored "bait" Fc, which results in targeting functional "half" IgGs to the cell wall of Pichiapastoris without interfering with the secretion of full length mAb. We show the utility of this method in isolating high affinity, well-expressed anti-PCSK9 leads from a designed library that was created by mating yeasts containing either light chain or heavy chain IgG libraries. Coupled with Glyco-engineered Pichiapastoris, this method provides a powerful tool for the discovery and production of therapeutic human mAbs in the same host thus improving drug developability and potentially shortening the discovery time cycle.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibody Formation , Pichia , Protein Engineering/methods , Antibodies, Monoclonal/genetics , Antibody Affinity/genetics , Antibody Affinity/immunology , Antibody Formation/genetics , Cell Separation/methods , Glycosylation , Humans , Organisms, Genetically Modified , Peptide Library , Pichia/genetics , Pichia/metabolism , Protein Multimerization , Protein Processing, Post-Translational
9.
J Biotechnol ; 166(4): 174-81, 2013 Jul 20.
Article in English | MEDLINE | ID: mdl-23735484

ABSTRACT

Production of recombinant proteins is affected by process conditions, where transcriptional regulation of Pichia pastoris alcohol oxidase 1 (PpAOX1) promoter has been a key factor to influence expression levels of proteins of interest. Here, we demonstrate that the AOX1 promoter and peroxisome biogenesis are regulated based on different process conditions. Two types of GFP-fusion proteins, Ub-R-GFP (short-lived GFP in the cytosol) and GFP-SKL (peroxisomal targeting GFP), were successfully used to characterize the time-course of the AOX1 promoter and peroxisome biogenesis, respectively. The activity of the AOX1 promoter and peroxisome biogenesis was highly subjected to different fermentation process conditions - methanol-limited condition at normoxy (ML), switched feeding of carbon sources (e.g., glucose and methanol) under carbon-limited condition at normoxy (SML), and oxygen-limited (OL) condition. The AOX1 promoter was most active under the ML, but less active under the OL. Peroxisome biogenesis showed a high dependency on methanol consumption. In addition, the proliferation of peroxisomes was inhibited in a medium containing glucose and stimulated in the methanol phase under a carbon-limited fed-batch culture condition. The specific productivity of a monoclonal antibody (qp) under the AOX1 promoter was higher at 86h of induction in the ML than in the OL (0.026 vs 0.020mgg(-1)h(-1)). However, the oxygen-limited condition was a robust process suitable for longer induction (180h) due to high cell fitness. Our study suggests that the maximal production of a recombinant protein is highly dependent on methanol consumption rate that is affected by the availability of methanol and oxygen molecules.


Subject(s)
Aldehyde Oxidase/genetics , Fermentation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Peroxisomes/metabolism , Pichia/genetics , Recombinant Fusion Proteins/biosynthesis , Batch Cell Culture Techniques/methods , Bioreactors , Cells, Cultured , Glucose/metabolism , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Methanol/metabolism , Oxygen/metabolism , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics
10.
PLoS One ; 8(5): e62229, 2013.
Article in English | MEDLINE | ID: mdl-23667461

ABSTRACT

Protein O-mannosyltransferases (PMTs) catalyze the initial reaction of protein O-mannosylation by transferring the first mannose unit onto serine and threonine residues of a nascent polypeptide being synthesized in the endoplasmic reticulum (ER). The PMTs are well conserved in eukaryotic organisms, and in vivo defects of these enzymes result in cell death in yeast and congenital diseases in humans. A group of rhodanine-3-acetic acid derivatives (PMTi) specifically inhibits PMT activity both in vitro and in vivo. As such, these chemical compounds have been effectively used to minimize the extent of O-mannosylation on heterologously produced proteins from different yeast expression hosts. However, very little is known about how these PMT-inhibitors interact with the PMT enzyme, or what structural features of the PMTs are required for inhibitor-protein interactions. To better understand the inhibitor-enzyme interactions, and to gain potential insights for developing more effective PMT-inhibitors, we isolated PMTi-resistant mutants in Pichia pastoris. In this study, we report the identification and characterization of a point mutation within the PpPMT2 gene. We demonstrate that this F664S point mutation resulted in a near complete loss of PMTi sensitivity, both in terms of growth-inhibition and reduction in O-mannosylglycan site occupancy. Our results provide genetic evidence demonstrating that the F664 residue plays a critical role in mediating the inhibitory effects of these PMTi compounds. Our data also indicate that the main target of these PMT-inhibitors in P. pastoris is Pmt2p, and that the F664 residue most likely interacts directly with the PMTi-compounds.


Subject(s)
Enzyme Inhibitors/pharmacology , Mannosyltransferases/antagonists & inhibitors , Mannosyltransferases/genetics , Pichia/enzymology , Acetates/pharmacology , Amino Acid Substitution , Endoplasmic Reticulum/metabolism , Mutagenesis , Mutation, Missense/genetics , Pichia/genetics , Plasmids/genetics , Point Mutation/genetics , Rhodanine/pharmacology
11.
Pharm Res ; 30(3): 803-12, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23135825

ABSTRACT

PURPOSE: P. pastoris has previously been genetically engineered to generate strains that are capable of producing mammalian-like glycoforms. Our objective was to investigate the correlation between sialic acid content and pharmacokinetic properties of recombinant TNFR2:Fc fusion proteins generated in glycoengineered P. pastoris strains. METHODS: TNFR2:Fc fusion proteins were generated with varying degrees of sialic acid content. The pharmacokinetic properties of these proteins were assessed by intravenous and subcutaneous routes of administration in rats. The binding of these variants to FcRn were also evaluated for possible correlations between in vitro binding and in vivo PK. RESULTS: The pharmacokinetic profiles of recombinant TNFR2:Fc produced in P. pastoris demonstrated a direct positive correlation between the extent of glycoprotein sialylation and in vivo pharmacokinetic properties. Furthermore, recombinant TNFR2:Fc produced in glycoengineered Pichia, with a similar sialic acid content to CHO-produced etanercept, demonstrated similar in vivo pharmacokinetic properties to the commercial material. In vitro surface plasmon resonance FcRn binding at pH6.0 showed an inverse relationship between sialic acid content and receptor binding affinity, with the higher affinity binders having poorer in vivo PK profiles. CONCLUSIONS: Sialic acid content is a critical attribute for modulating the pharmacokinetics of recombinant TNFR2:Fc produced in glycoengineered P. pastoris.


Subject(s)
Immunoglobulin G/blood , Immunoglobulin G/genetics , Immunosuppressive Agents/blood , Pichia/genetics , Receptors, Tumor Necrosis Factor/blood , Receptors, Tumor Necrosis Factor/genetics , Animals , Cloning, Molecular , Etanercept , Genetic Engineering , Glycosylation , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/metabolism , Male , N-Acetylneuraminic Acid/analysis , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Fc/metabolism , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/metabolism
12.
J Immunol Methods ; 386(1-2): 34-42, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-22982058

ABSTRACT

Previous studies have shown that glycoproteins expressed in wild-type Pichia pastoris bind to Dendritic cell-SIGN (DC-Specific Intercellular adhesion molecule-3 Grabbing Nonintegrin), a mannose-binding receptor found on dendritic cells in peripheral tissues which is involved in antigen presentation and the initiation of an immune response. However, the binding of DC-SIGN to glycoproteins purified from P. pastoris strains engineered to express humanized N- and O-linked glycans has not been tested to date. In this study, the binding of glycoproteins with specific high-mannose or human N- and O-linked glycan structures to DC-SIGN was tested. Proteins with humanized N-glycans including Man5 structures and O-glycans (up to as many as 24) with single mannose chain length showed DC-SIGN binding that was comparable to that measured for a CHO-produced IgG1 which lacks O-linked mannose. Glycoproteins with wild-type N-glycans and mannotriose and higher O-glycans bound to DC-SIGN in a manner that was strongly inhibited by either the use of enzymatic N-deglycosylation or sodium meta-periodate oxidation. Mannan purified from humanized P. pastoris also showed lower ability to inhibit DC-SIGN binding to glycoproteins with wild type fungal glycosylation than mannan purified from wild type strains. This study shows that humanized P. pastoris can produce glycoproteins that do not bind to DC-SIGN.


Subject(s)
Cell Adhesion Molecules/metabolism , Glycoproteins/metabolism , Immunoglobulin G/metabolism , Lectins, C-Type/metabolism , Pichia/genetics , Polysaccharides/metabolism , Receptors, Cell Surface/metabolism , Animals , CHO Cells , Cricetinae , Glycoproteins/genetics , Glycosylation , Humans , Immunoglobulin G/genetics , Mannose/metabolism , Protein Binding/genetics , Protein Engineering
13.
Microb Cell Fact ; 11: 91, 2012 Jul 02.
Article in English | MEDLINE | ID: mdl-22748191

ABSTRACT

BACKGROUND: Yeast mating provides an efficient means for strain and library construction. However, biotechnological applications of mating in the methylotrophic yeast Pichia pastoris have been hampered because of concerns about strain stability of P. pastoris diploids. The aim of the study reported here is to investigate heterologous protein expression in diploid P. pastoris strains and to evaluate diploid strain stability using high cell density fermentation processes. RESULTS: By using a monoclonal antibody as a target protein, we demonstrate that recombinant protein production in both wild-type and glycoengineered P. pastoris diploids is stable and efficient during a nutrient rich shake flask cultivation. When diploid strains were cultivated under bioreactor conditions, sporulation was observed. Nevertheless, both wild-type and glycoengineered P. pastoris diploids showed robust productivity and secreted recombinant antibody of high quality. Specifically, the yeast culture maintained a diploid state for 240 h post-induction phase while protein titer and N-linked glycosylation profiles were comparable to that of a haploid strain expressing the same antibody. As an application of mating, we also constructed an antibody display library and used mating to generate novel full-length antibody sequences. CONCLUSIONS: To the best of our knowledge, this study reports for the first time a comprehensive characterization of recombinant protein expression and fermentation using diploid P. pastoris strains. Data presented here support the use of mating for various applications including strain consolidation, variable-region glycosylation antibody display library, and process optimization.


Subject(s)
Pichia/metabolism , Recombinant Proteins/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Biomass , Diploidy , Fermentation , Haploidy , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Pichia/growth & development , Recombinant Proteins/genetics , Recombinant Proteins/immunology
14.
PLoS One ; 7(6): e37915, 2012.
Article in English | MEDLINE | ID: mdl-22685548

ABSTRACT

Biopharmaceuticals represent the fastest growing sector of the global pharmaceutical industry. Cost-efficient production of these biologic drugs requires a robust host organism for generating high titers of protein during fermentation. Understanding key cellular processes that limit protein production and secretion is, therefore, essential for rational strain engineering. Here, with single-cell resolution, we systematically analysed the productivity of a series of Pichia pastoris strains that produce different proteins both constitutively and inducibly. We characterized each strain by qPCR, RT-qPCR, microengraving, and imaging cytometry. We then developed a simple mathematical model describing the flux of folded protein through the ER. This combination of single-cell measurements and computational modelling shows that protein trafficking through the secretory machinery is often the rate-limiting step in single-cell production, and strategies to enhance the overall capacity of protein secretion within hosts for the production of heterologous proteins may improve productivity.


Subject(s)
Pichia/metabolism , Recombinant Fusion Proteins/metabolism , Secretory Pathway , Single-Cell Analysis/methods , Algorithms , Endoplasmic Reticulum/metabolism , Flow Cytometry , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Microscopy, Fluorescence , Models, Biological , Pichia/genetics , Plasmids/genetics , Recombinant Fusion Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transformation, Genetic
15.
Appl Microbiol Biotechnol ; 95(3): 671-82, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22569635

ABSTRACT

Yeast is capable of performing posttranslational modifications, such as N- or O-glycosylation. It has been demonstrated that N-glycans play critical biological roles in therapeutic glycoproteins by modulating pharmacokinetics and pharmacodynamics. However, N-glycan sites on recombinant glycoproteins produced in yeast can be underglycosylated, and hence, not completely occupied. Genomic homology analysis indicates that the Pichia pastoris oligosaccharyltransferase (OST) complex consists of multiple subunits, including OST1, OST2, OST3, OST4, OST5, OST6, STT3, SWP1, and WBP1. Monoclonal antibodies produced in P. pastoris show that N-glycan site occupancy ranges from 75-85 % and is affected mainly by the OST function, and in part, by process conditions. In this study, we demonstrate that N-glycan site occupancy of antibodies can be improved to greater than 99 %, comparable to that of antibodies produced in mammalian cells (CHO), by overexpressing Leishmania major STT3D (LmSTT3D) under the control of an inducible alcohol oxidase 1 (AOX1) promoter. N-glycan site occupancy of non-antibody glycoproteins such as recombinant human granulocyte macrophage colony-stimulating factor (rhGM-CSF) was also significantly improved, suggesting that LmSTT3D has broad substrate specificity. These results suggest that the glycosylation status of recombinant proteins can be improved by heterologous STT3 expression, which will allow for the customization of therapeutic protein profiles.


Subject(s)
Glycoproteins/metabolism , Glycosylation , Pichia/metabolism , Protein Processing, Post-Translational , Animals , Antibodies, Monoclonal/metabolism , CHO Cells , Cricetinae , Gene Expression , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Leishmania major/enzymology , Leishmania major/genetics , Metabolic Engineering , Recombinant Proteins/metabolism
16.
J Biotechnol ; 157(1): 198-206, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22100268

ABSTRACT

Pichia pastoris is a methylotropic yeast that has gained great importance as an organism for protein expression in recent years. Here, we report the expression of recombinant human erythropoietin (rhEPO) in glycoengineered P. pastoris. We show that glycosylation fidelity is maintained in fermentation volumes spanning six orders of magnitude and that the protein can be purified to high homogeneity. In order to increase the half-life of rhEPO, the purified protein was coupled to polyethylene glycol (PEG) and then compared to the currently marketed erythropoiesis stimulating agent, Aranesp(®) (darbepoetin). In in vitro cell proliferation assays the PEGylated protein was slightly, and the non-PEGylated protein was significantly more active than comparator. Pharmacodynamics as well as pharmacokinetic activity of PEGylated rhEPO in animals was comparable to that of Aranesp(®). Taken together, our results show that glycoengineered P. pastoris is a suitable production host for rhEPO, yielding an active biologic that is comparable to those produced in current mammalian host systems.


Subject(s)
Erythropoietin/biosynthesis , Pichia/metabolism , Protein Engineering/methods , Animals , Cell Proliferation/drug effects , Darbepoetin alfa , Erythropoietin/analogs & derivatives , Erythropoietin/blood , Erythropoietin/genetics , Erythropoietin/pharmacokinetics , Erythropoietin/pharmacology , Female , Glycosylation , Humans , Male , Mice , Pichia/genetics , Polyethylene Glycols , Polysaccharides/chemistry , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
17.
J Immunol Methods ; 375(1-2): 159-65, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22019510

ABSTRACT

A fragment of antigen binding (Fab) surface display system was developed using a glycoengineered Pichia pastoris host strain genetically modified to secrete glycoproteins with mammalian mannose-type Man(5)GlcNAc(2) N-linked glycans. The surface display method described here takes advantage of a pair of coiled-coil peptides as the linker while using the Saccharomyces cerevisiae Sed1p GPI-anchored cell surface protein as an anchoring domain. Several Fabs were successfully displayed on the cell surface using this system and the expression level of the displayed Fabs was correlated to that of secreted Fabs from the same glycoengineered host in the absence of the cell wall anchor. Strains displaying different model Fabs were mixed and, through cell sorting, the strain displaying more expressed Fab molecule or the strain displaying the Fab with higher affinity for an antigen was effectively enriched by FACS. This novel yeast surface display system provides a general platform for the display of Fab libraries for affinity and/or expression maturation using glycoengineered Pichia.


Subject(s)
Glycoproteins/genetics , Glycoproteins/metabolism , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Pichia/genetics , Pichia/metabolism , Antigens, Surface/genetics , Antigens, Surface/metabolism , Cell Wall/genetics , Cell Wall/metabolism , Genetic Vectors/genetics , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Peptides/genetics , Peptides/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
18.
Glycobiology ; 21(12): 1616-26, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21840970

ABSTRACT

The methylotrophic yeast, Pichia pastoris, is an important organism used for the production of therapeutic proteins. However, the presence of fungal-like glycans, such as those containing ß-mannose (Man) linkages, can elicit an immune response or bind to Man receptors, thus reducing their efficacy. Recent studies have confirmed that P. pastoris has four genes from the ß-mannosyl transferase (BMT) family and that Bmt2p is responsible for the majority of ß-Man linkages on glycans. While expressing recombinant human erythropoietin (rhEPO) in a developmental glycoengineered strain devoid of BMT2 gene expression, cross-reactivity was observed with an antibody raised against host cell antigens. Treatment of the rhEPO with protein N-glycosidase F eliminated cross-reactivity, indicating that the antigen was associated with the glycan. Thorough analysis of the glycan profile of rhEPO demonstrated the presence of low amounts of α-1,2-mannosidase resistant high-Man glycoforms. In an attempt to eliminate the α-mannosidase resistant glycoforms, we used a systemic approach to genetically knock-out the remaining members of the BMT family culminating in a quadruple bmt2,4,1,3 knock-out strain. Data presented here conclude that the additive elimination of Bmt2p, Bmt3p and Bmt1p activities are required for total abolition of ß-Man-associated glycans and their related antigenicity. Taken together, the elimination of ß-Man containing glycoforms represents an important step forward for the Pichia production platform as a suitable system for the production of therapeutic glycoproteins.


Subject(s)
Mannose/chemistry , Pichia/chemistry , Polysaccharides/chemistry , Polysaccharides/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Carbohydrate Conformation , Cross Reactions , Erythropoietin/chemistry , Erythropoietin/genetics , Erythropoietin/isolation & purification , Erythropoietin/metabolism , Humans , Mannose/metabolism , Mannosyltransferases/genetics , Mannosyltransferases/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Pichia/enzymology , Pichia/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
19.
Glycobiology ; 21(12): 1606-15, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21798867

ABSTRACT

The N-glycosylation pathway in Pichia pastoris has been humanized by the deletion of genes responsible for fungal-type glycosylation (high mannose) as well as the introduction of heterologous genes capable of forming human-like N-glycosylation. This results in a yeast host that is capable of expressing therapeutic glycoproteins. A thorough investigation was performed to examine whether glycoproteins expressed in glycoengineered P. pastoris strains may contain residual fungal-type high-mannose structures. In a pool of N-linked glycans enzymatically released by protein N-glycosidase from a reporter glycoprotein expressed in a developmental glycoengineered P. pastoris strain, an oligosaccharide with a mass consistent with a Hexose(9)GlcNAc(2) oligosaccharide was identified. When this structure was analyzed by a normal-phase high-performance liquid chromatography (HPLC), its retention time was identical to a Man(9)GlcNAc(2) standard. However, this Hexose(9)GlcNAc(2) oligosaccharide was found to be resistant to α-1,2-mannosidase as well as endomannosidase, which preferentially catabolizes endoplasmic reticulum oligosaccharides containing terminal α-linked glucose. To further characterize this oligosaccharide, we purified the Hexose(9)GlcNAc(2) oligosaccharide by HPLC and analyzed the structure by high-field one-dimensional (1D) and two-dimensional (2D) (1)H NMR (nuclear magnetic resonance) spectroscopy followed by structural elucidation by homonuclear and heteronuclear 1D and 2D (1)H and (13)C NMR spectroscopy. The results of these experiments lead to the identification of an oligosaccharide α-Man-(1 → 2)-ß-Man-(1 → 2)-ß-Man-(1 → 2)-α-Man-(1 → 2) moiety as part of a tri-antennary structure. The difference in enzymatic reactivity can be attributed to multiple ß-linkages on the α-1,3 arm of the Man(9)GlcNAc(2) oligosaccharide.


Subject(s)
Mannosidases/metabolism , Membrane Proteins/metabolism , Oligosaccharides/biosynthesis , Oligosaccharides/chemistry , Pichia/metabolism , Humans , Mannosidases/genetics , Membrane Proteins/genetics , Pichia/genetics , Protein Conformation , Structure-Activity Relationship
20.
MAbs ; 3(3): 289-98, 2011.
Article in English | MEDLINE | ID: mdl-21487242

ABSTRACT

Mammalian cell culture systems are used predominantly for the production of therapeutic monoclonal antibody (mAb) products. A number of alternative platforms, such as Pichia engineered with a humanized N-linked glycosylation pathway, have recently been developed for the production of mAbs. The glycosylation profiles of mAbs produced in glycoengineered Pichia are similar to those of mAbs produced in mammalian systems. This report presents for the first time the comprehensive characterization of an anti-human epidermal growth factor receptor 2 (HER2) mAb produced in a glycoengineered Pichia, and a study comparing the anti-HER2 from Pichia, which had an amino acid sequence identical to trastuzumab, with trastuzumab. The comparative study covered a full spectrum of preclinical evaluation, including bioanalytical characterization, in vitro biological functions, in vivo anti-tumor efficacy and pharmacokinetics in both mice and non-human primates. Cell signaling and proliferation assays showed that anti-HER2 from Pichia had antagonist activities comparable to trastuzumab. However, Pichia-produced material showed a 5-fold increase in binding affinity to FcγIIIA and significantly enhanced antibody dependant cell-mediated cytotoxicity (ADCC) activity, presumably due to the lack of fucose on N-glycans. In a breast cancer xenograft mouse model, anti-HER2 was comparable to trastuzumab in tumor growth inhibition. Furthermore, comparable pharmacokinetic profiles were observed for anti-HER2 and trastuzumab in both mice and cynomolgus monkeys. We conclude that glycoengineered Pichia provides an alternative production platform for therapeutic mAbs and may be of particular interest for production of antibodies for which ADCC is part of the clinical mechanism of action.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal/immunology , Pichia/genetics , Receptor, ErbB-2/immunology , Recombinant Proteins/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibody Affinity/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Area Under Curve , Binding, Competitive/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Fucose/metabolism , Genetic Engineering , Humans , Macaca fascicularis , Mice , Mice, Inbred C57BL , Pichia/metabolism , Polysaccharides/metabolism , Protein Binding/immunology , Receptors, IgG/immunology , Receptors, IgG/metabolism , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Trastuzumab , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...