Available online at www.sciencedirect.com
ScienceDirect Biopharmaceutical discovery and production in yeast Michael A Meehl and Terrance A Stadheim 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 yeastbased 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. Addresses GlycoFi, Biologics Research, Merck & Co., Inc., 16 Cavendish Court, Lebanon, NH 03766, USA Corresponding author: Stadheim, Terrance A (
[email protected]) Current Opinion in Biotechnology 2014, 30:120–127 This review comes from a themed issue on Pharmaceutical biotechnology Edited by Beth Junker and Jamey D Young
http://dx.doi.org/10.1016/j.copbio.2014.06.007 0958-1669/# 2014 Elsevier Ltd. All rights reserved.
Introduction The dawn of recombinant DNA techniques ushered in the modernization of protein-based pharmaceuticals, known as biopharmaceuticals or biologics. In the early 1980s, recombinant human insulin was marketed and provided Type I diabetics with a more homogenous and reliable medicine that no longer required sourcing from animal pancreas. The development of recombinant insulin helped pave the way for the marketing approval of Escherichia coli and Saccharomyces cerevisiae derived products. Three decades later, the current quality of recombinant glycoprotein expression in yeast rivals that of traditional mammalian cell culture systems. Such engineering and process development efforts have occurred in different systems with varying degrees of success. Recent advancements in yeast engineering have provided yeast-based platforms with more than just the ability to Current Opinion in Biotechnology 2014, 30:120–127
manufacture aglycosylated peptides or proteins with low complexity. Considerable effort has focused on strain improvements to develop glycoengineered yeast systems for the production of recombinant glycoproteins with desired physiochemical properties and efficacy. Innovations in yeast genetics and protein secretion will also be discussed that enable yeast to be a state-of-the-art tool for biopharmaceutical research and development.
Yeast production systems and derived products Early biopharmaceuticals originated from animal and human sources. However, the majority of newly marketed products are derived from recombinant expression systems. Yeast, along with bacterial (i.e. E. coli) and mammalian hosts (i.e. Chinese Hamster Ovary) represent the most frequently used expression systems for biopharmaceuticals. In the early 1980s, ZymoGenetics industrialized S. cerevisiae as a host for recombinant human insulin leading to the marketing of Novolin1 with Novo Nordisk in 1987. Today, the S. cerevisiae platform currently delivers half of the world supply of insulin. With the recent interest in biosimilar products, Pichia pastoris has been used to produce recombinant human insulin with titers of 3 g/L [1,2]. In fact, Biocon is the world’s fourth largest supplier of insulin products and utilizes P. pastoris [URL: http://www.biocon.com]. The example of recombinant insulin demonstrates the capabilities of yeast-based biopharmaceutical expression and production. In recent years, many new biopharmaceutical candidates were produced in S. cerevisiae, P. pastoris, and Hansenula polymorpha platforms [3–5,6,7]. The most recent FDA approval of a yeast-expressed biopharmaceutical was in 2012 for ocriplasmin, sold as Jetrea1 by ThromboGenics [8]. Ocriplasmin is an aglycosylated protease expressed in P. pastoris approved for the treatment of symptomatic vitreomacular adhesion [9]. Other notable yeast-based biopharmaceuticals with marketing approval include: a Hepatitis B subunit vaccine using Hepatitis B surface antigen (S. cerevisiae, P. pastoris, and H. polymorpha); recombinant human granulocyte macrophage-colony stimulating factor, Leukine1(sargramostim, S. cerevisiae, 1991); recombinant human platelet derived growth factor, Regranex1 (becaplermin, S. cerevisiae, 1997); human papillomavirus subunit vaccine, Gardasil1 (S. cerevisiae, 2006); and a kallikrein inhibitor, Kalbitor1 (ecallantide, P. pastoris, 2009). Numerous yeast-based biopharmaceuticals are in clinical development. Ablynx reports at least two clinical programs focused on nanobody-based therapeutics www.sciencedirect.com
Biopharmaceutical discovery and production in yeast Meehl and Stadheim 121
expressed in P. pastoris [URL: http://www.ablynx.com]. Alder Biopharmaceuticals reports two clinical antibody leads targeting interleukin-6 (IL-6), named ALD518 (BMS-945429), and calcitonin gene-related peptide (CGRP), named ALD403, in their pipeline using the P. pastoris expression system [URL: http://alderbio.com]. In the case of ALD518 (BMS-945429), the humanized antibody was engineered to lack Fc glycosylation. However, serum half-life is preserved and is comparable to other marketed glycosylated antibodies [10,11]. This latter example of full-length antibody expression in P. pastoris signifies a shift towards the production of more complex biopharmaceuticals. Indeed, there are reports of the production in P. pastoris of biopharmaceuticals with human-like glycosylation patterns, including antibodies and therapeutic proteins [12–14,15,16–18,19,20–22].
suitable for clinical use. Proteins with yeast-type glycosylation, when delivered in vivo, interact with human C-type lectins of the innate immune system resulting in altered pharmacokinetic properties, activation of complement, enhanced anti-drug–antibody formation, or sequestration by circulating anti-glycan antibodies [20,25–27,28]. Furthermore, mannose glycans have been shown to alter the efficacy of recombinant vaccines [29,30]. Many therapeutic glycoprotein candidates require a particular human-like glycoform for optimal efficacy, such as sialic acid for half-life extension [18,31], lack of fucose on antibodies for improved cytotoxic properties [32], and paucimannose for selective targeting to macrophages [33]. While in vitro glycan modifications using processing enzymes are possible, the most robust and efficient methods to produce particular glycosylation profiles are those that can be synthesized in vivo, such as using glycoengineered yeast.
Strain engineering and process improvements N-glycan modifications
Many biopharmaceuticals require proper post-translational modifications, including glycosylation, for optimal pharmacokinetic (PK) and pharmacodynamic (PD) properties [23,24]. Recombinant biopharmaceuticals with yeast-derived high mannose N-glycans are often not
N-glycan humanization centers on the conversion of an extant fungal high mannose oligosaccharide to a complex oligosaccharide that can be further optimized to contain specific terminal sugars (i.e. N-acetylglucosamine (GlcNAc), galactose and sialic acid) (see Figure 1). Over the past several years, this feat of genetic engineering
Figure 1
N-Acetylglucosamine
Mannose
Galactose
Sialic acid
SiaT* n Yeast N-glycan (high mannose)
och1Δ
MnsI
GalT
GnTI
MnsII
SiaT*
GalT
SiaT*
GnTII
GalT
SiaT*
GalT
SiaT* Current Opinion in Biotechnology
Yeast N-glycan engineering. The N-glycosylation pathway of glycoengineered Pichia pastoris was previously reviewed [34]. Glycoproteins harboring predetermined glycoforms [78] are obtained depending on the glycoengineered yeast host used, each of which contains a unique set of gene deletions and glycosylation enzymes, as indicated by arrows. The main glycosylation pathway to obtain mammalian biantennary glycans is shown in the upper left rectangle. As indicated by (*), sialic acid linkages may be exclusively a-2,6 or a-2,3 depending on the chosen sialyltransferase. Other yeast modifications (e.g. beta-linked mannose, mannosylphosphate) are not depicted in the figure. www.sciencedirect.com
Current Opinion in Biotechnology 2014, 30:120–127
122 Pharmaceutical biotechnology
has been achieved most notably in P. pastoris (for reviews, see [17,34]). Through the use of genetic perturbations limiting yeast mannosylation and the introduction of heterologous glycosyltransferases and related pathway machinery, the P. pastoris N-glycosylation pathway was engineered to enable secretion of proteins with complex glycans [16,34–39]. Using glycoengineered P. pastoris, antibodies and therapeutic proteins have been produced with complex glycoforms exhibiting suitable pharmacokinetic and pharmacodynamics properties required for in vivo efficacy [18,19,20,40,41]. N-glycan engineering efforts have also been recently reported in S. cerevisiae [42,43], H. polymorpha [44,45,46], Kluyveromyces lactis [47], and Yarrowia lipolytica [48,49]. These yeast glycoengineering reports are centered on two main stages, namely removal of yeast hypermannosylation and the conversion to complex glycoforms containing terminal GlcNAc, galactose, or sialic acid. In the first stage, varying approaches have been taken to remove or limit hypermannosylation. In one approach, yeast glycosyltransferase genes (e.g. OCH1) were disrupted to eliminate hypermannosylation and mannosidase genes expressed to obtain a human-like Man5GlcNAc2 glycoform [36,42,45,49,50]. In other approaches, the deletion of the ALG3 gene in combination with varying modifications leads to the generation of a Man3GlcNAc2 glycoform [35,39,43,46–48]. In the second stage of humanization, the terminal mannose substrate can be further modified by a specific N-acetylglucosamine transferase, GnT-I, to result in a hybrid-type glycoform containing terminal GlcNAc. In subsequent steps, a second GlcNAc sugar is added to the terminal a-1,6-mannose residue through the activity of GnT-II. The resulting glycan, GlcNAc2Man3GlcNAc2, can be further modified to contain terminal galactose and sialic acid (Figure 1). As evidenced from the profiling of glycoproteins expressed in yeast [51] and the analysis of Leukine1 expressed in S. cerevisiae [52], the occupancy of N-glycan sites is often not complete. Incomplete glycan occupancy may create challenges for certain biotherapeutics by impacting stability and/or efficacy. To eliminate this macroheterogeneity, N-linked glycan site(s) can simply be removed from the molecule if no impact on efficacy or immunogenicity is anticipated. As mentioned earlier, Alder Biopharmaceuticals has used this approach to produce their clinical antibody ALD-518, whereby the Asn residue at position 297 is substituted for an Ala residue [11]. Rather than eliminating N-glycans in yeast-based recombinant proteins, an alternative approach is to maximize N-glycan site occupancy. A genetic solution was provided by heterologous expression of a STT3 enzyme to increase site occupancy of complex glycans to near completion without negatively affecting protein secretion [52]. These recent reports on yeast N-glycan humanization are signaling a shift from the proof of concept phase to implementation by improving characteristics of recombinant proteins suitable for clinical use. Current Opinion in Biotechnology 2014, 30:120–127
O-Glycan modifications Removing wild-type yeast O-glycosylation
In yeast O-glycosylation, the hydroxyl functional groups of serine and threonine residues are modified with a mannose monosaccharide in a non-template driven manner in the endoplasmic reticulum by the PMT (protein O-mannosyltransferase) family of enzymes [53,54]. Mannose residues are then added to the O-glycan in the Golgi apparatus by additional enzymes creating a polydisperse mannose polymer of up to five mannose residues. Proteins made in yeast that are decorated with O-glycans are capable of mediating interactions with human mannose binding lectins, conferring inferior PK properties of biopharmaceuticals [19,55]. Conversely, vaccine antigenicity can be improved by yeast O-glycosylation via targeting to antigen presenting cells [56]. To reduce the presence, or occupancy, of O-glycans, PMT gene disruptions have been employed. Since yeast typically possess multiple protein O-mannosyltransferases, often with overlapping target specificities, a single PMT gene deletion is not sufficient to remove all O-glycosylation [53,57]. Furthermore, chemical PMT inhibitors also demonstrated the ability to significantly reduce O-mannose occupancy in yeast, and interestingly also correlated with improved antibody production [57–59]. Furthermore, expression of mannosidases capable of hydrolyzing a-1,2-mannose linkages was successfully shown to limit O-mannose chain length, and as a result, led to a reduction in lectin binding by the glycoprotein [55]. Overall, data suggests wild-type, or uncontrolled, yeast O-glycosylation may be detrimental for PK properties of biopharmaceuticals, and genetic or process solutions that control O-glycan occupancy and chain length have resulted in proteins with properties undifferentiated from mammalian proteins lacking O-mannosylation [41]. Glycoengineering of yeast O-glycosylation
Rather than limiting the occupancy or extension of O-mannose linkages, an alternative strategy has been described to humanize O-glycans. In humans, two main types of O-glycans are found, the mucin-type characterized by Ser/Thr-O-GalNAc-linkages and the a-dystroglycan-type of Ser/Thr-O-Man-GlcNAc. Efforts to convert yeast O-mannose to O-GalNAc are particularly challenging due to the overlapping functions of the PMT family. Nevertheless, the engineering of mucin-type O-glycosylation in S. cerevisiae has been reported [60]. In an earlier study by the same group, an artificial O-glycosylation pathway was engineered in S. cerevisiae to produce an O-fucosylated epidermal growth factor (EGF) domain, demonstrating the ability to produce a heterologous O-glycoform [61]. The introduction of the enzyme protein-O-linked-mannose b-1,2-N-acetylglucosaminyltransferase 1 (PomGnT1) to P. pastoris led to the production of highly sialylated a-dystroglycan-type www.sciencedirect.com
Biopharmaceutical discovery and production in yeast Meehl and Stadheim 123
O-glycans [15]. In summary, significant remodeling of yeast N-glycosylation and O-glycosylation pathways offers the ability to optimize biopharmaceuticals with desired in vivo properties [19].
Biopharmaceutical R&D using yeast systems The varying efforts of yeast glycoengineering have focused on the development of biopharmaceutical production systems capable of manufacturing proteins with
Figure 2
* *
*
*
IgG with complex N-glycans
(Ib) Rational Libraries
105
1 2 3 4 5 6 7 8 9 10 11 12 A B C D E F G H
104 103 102 0 0 102
(Ia) Cell Surface Display mAbs or Fc-fusion Libraries
103
104
105
(II) FACS Clone Selection and Developability
(III) Clone Evaluation
Co-secretion
(V) Process Development and Manufacturing
(IV) Preclinical Assays Current Opinion in Biotechnology
Discovery of yeast-based biopharmaceuticals. The advancements in recombinant protein expression in yeast enable end-to-end biopharmaceutical production, from discovery to manufacturing. Novel leads are generated from either randomized approaches, such as cell surface display (Ia) or rational designs of glycan and/or amino acid variants (Ib). Clones expressing variants may be selected using FACS (II) and/or in medium-throughput assays (III). In the case of Fc display [73], yeast expressing cell-surface variants are also capable of co-secretion, shortening the time needed for candidate selection. Following the assessment of preclinical assays (in vitro and in vivo) to support candidate selection (IV), final yeast clones are selected for manufacturing (V). www.sciencedirect.com
Current Opinion in Biotechnology 2014, 30:120–127
124 Pharmaceutical biotechnology
superior pharmacokinetic and pharmacodynamic properties. However, yeast platforms have great potential upstream of manufacturing in the discovery of biopharmaceutical candidate molecules. Yeast offer numerous advantages in the discovery space including ease of genetic manipulation with good transformation efficiency, the ability to increase diversity through mating haploid strains, eukaryotic quality control of the secretory pathway, and the rapid generation of test materials using a clonal population. Whereas some candidate molecules are fairly well defined at the outset of a program (e.g. enzymes, growth factors, cytokines, receptors, etc.), other molecules such as antibodies can require large screening effort to find the optimal lead. Transformation efficiencies for S. cerevisiae and P. pastoris are reported to be in the range of 105–106 CFU/mg DNA [62–64]. These efficiencies are readily suitable for the selection of high expression clones with varying gene copies [65]. Randomized library-based screening efforts may require sequences in the 106–1011 range. For further increases in diversity, yeast has the distinct advantage of mating MATa and MATa haploid clones for generating highly diverse antibody libraries [66,67]. Diploid strains obtained though mating are also capable of robust production of a full-length antibody, as shown in a recent study using wild-type and glycoengineered diploid P. pastoris strains under fermentation conditions [68]. Antibodies and antibody fragments are some of the most efficacious and medically important therapies on the market today, as proven by the robust success of the anti-TNF agents [69]. Popular methods to identify the Fab sequence against an antigen include both in vivo (e.g. through immunization of either naı¨ve or transgenic rodents) and in vitro (e.g. phage, yeast, ribosome, mammalian displays) methods. While murine hybridomas and phage display technologies initially dominated the antibody discovery space, recent advances in yeast display have provided powerful advantages for antibody discovery and engineering when used in combination with fluorescence-activated cell sorting (FACS) (for recent reviews, see [70,71]). A number of approaches have been developed to enable yeast surface display, including a report that described thirteen new GPImodified cell wall proteins in P. pastoris that may be sufficient to display heterologous proteins on the cell surface [72]. In a recent and novel approach, a surfaceanchored ‘bait’ Fc-Sed1p fusion was co-expressed in a glycoengineered P. pastoris strain capable of both surface display and full-length antibody secretion [73]. This approach provides a single system for linking genotype with phenotype while evaluating function/ developability of a Fab in the context of a full-length antibody. Current Opinion in Biotechnology 2014, 30:120–127
In other recent studies, yeast display was utilized to find unique antibody sequences that demonstrated cross-reactive binding to a specific epitope on a target protein using competitive panning [74] and to membrane proteins using detergent-solubilized cell lysates containing antigens with near-native conformations [75,76]. A key determinant to the developability of an antibody is the lack of off-target binding. As such, broad panel tissue distribution experiments are often performed following clone identification, but a recent study detailed a FACSbased high-throughput selection tool using soluble membrane proteins coupled with yeast display to counter select against polyspecific antibodies early in the lead identification process [77]. The importance of yeast surface display in discovery continues to grow, as yeast systems are now capable of covering a diverse human library, display on the cell surface for phenotype–genotype linkages, isolate optimized clones while counter selecting undesirable qualities, and secrete fully human antibodies for lead identification efforts (for an illustration, see Figure 2).
Conclusions Recent innovations in yeast have unlocked new applications for recombinant protein production. Yeast systems are established end-to-end platforms for the discovery, development, and manufacture of biopharmaceuticals. Notable improvements include glycoengineering platforms, display technologies and libraries, and robust scalable manufacturing to support the use of yeast in all aspects of research and development. And as such, the next decade of biopharmaceutical development proves to be fruitful for the yeast platform in the supply of lifealtering medicines around the world.
References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as: of special interest of outstanding interest 1.
Gurramkonda C, Polez S, Skoko N, Adnan A, Gabel T, Chugh D, Swaminathan S, Khanna N, Tisminetzky S, Rinas U: 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, 9:31.
2.
Xie T, Liu Q, Xie F, Liu H, Zhang Y: Secretory expression of insulin precursor in Pichia pastoris and simple procedure for producing recombinant human insulin. Prep Biochem Biotechnol 2008, 38:308-317.
3.
Berlec A, Strukelj B: Current state and recent advances in biopharmaceutical production in Escherichia coli, yeasts and mammalian cells. J Ind Microbiol Biotechnol 2013, 40:257-274.
4.
Celik E, Calik P: Production of recombinant proteins by yeast cells. Biotechnol Adv 2012, 30:1108-1118.
5.
Frenzel A, Hust M, Schirrmann T: Expression of recombinant antibodies. Front Immunol 2013, 4:217.
6.
Mattanovich D, Branduardi P, Dato L, Gasser B, Sauer M, Porro D: Recombinant protein production in yeasts. Methods Mol Biol 2012, 824:329-358. www.sciencedirect.com
Biopharmaceutical discovery and production in yeast Meehl and Stadheim 125
A thorough review of the technical tools, both genetic and process, of yeast expression systems leading to higher levels of productivity. 7.
8. 9.
Spadiut O, Capone S, Krainer F, Glieder A, Herwig C: Microbials for the production of monoclonal antibodies and antibody fragments. Trends Biotechnol 2014, 32:54-60. Mullard A: 2012 FDA drug approvals. Nat Rev Drug Discov 2013, 12:87-90. Syed YY, Dhillon S: Ocriplasmin: a review of its use in patients with symptomatic vitreomacular adhesion. Drugs 2013, 73:1617-1625.
10. Mease P, Strand V, Shalamberidze L, Dimic A, Raskina T, Xu LA, Liu Y, Smith J: A phase II, double-blind, randomised, placebocontrolled study of BMS945429 (ALD518) in patients with rheumatoid arthritis with an inadequate response to methotrexate. Ann Rheum Dis 2012, 71:1183-1189. 11. Jung ST, Kang TH, Kelton W, Georgiou G: Bypassing glycosylation: engineering aglycosylated full-length IgG antibodies for human therapy. Curr Opin Biotechnol 2011, 22:858-867.
22. Li H, Sethuraman N, Stadheim TA, Zha D, Prinz B, Ballew N, Bobrowicz P, Choi BK, Cook WJ, Cukan M et al.: Optimization of humanized IgGs in glycoengineered Pichia pastoris. Nat Biotechnol 2006, 24:210-215. 23. Li H, d’Anjou M: Pharmacological significance of glycosylation in therapeutic proteins. Curr Opin Biotechnol 2009, 20:678-684. 24. Sethuraman N, Stadheim TA: Challenges in therapeutic glycoprotein production. Curr Opin Biotechnol 2006, 17:341-346. 25. Alessandri L, Ouellette D, Acquah A, Rieser M, Leblond D, Saltarelli M, Radziejewski C, Fujimori T, Correia I: Increased serum clearance of oligomannose species present on a human IgG1 molecule. MAbs 2012, 4:509-520. 26. Goetze AM, Liu YD, Zhang Z, Shah B, Lee E, Bondarenko PV, Flynn GC: High-mannose glycans on the Fc region of therapeutic IgG antibodies increase serum clearance in humans. Glycobiology 2011, 21:949-959. 27. Yu M, Brown D, Reed C, Chung S, Lutman J, Stefanich E, Wong A, Stephan JP, Bayer R: Production, characterization, and pharmacokinetic properties of antibodies with N-linked mannose-5 glycans. MAbs 2012, 4:475-487.
12. Barnard GC, Kull AR, Sharkey NS, Shaikh SS, Rittenhour AM, Burnina I, Jiang Y, Li F, Lynaugh H, Mitchell T et al.: Highthroughput screening and selection of yeast cell lines expressing monoclonal antibodies. J Ind Microbiol Biotechnol 2010, 37:961-971.
28. Rabinovich GA, van Kooyk Y, Cobb BA: Glycobiology of immune responses. Ann N Y Acad Sci 2012, 1253:1-15. The authors detail the comprehensive information relating the impact of protein glycosylation to human physiology and disease.
13. Potgieter TI, Cukan M, Drummond JE, Houston-Cummings NR, Jiang Y, Li F, Lynaugh H, Mallem M, McKelvey TW, Mitchell T et al.: Production of monoclonal antibodies by glycoengineered Pichia pastoris. J Biotechnol 2009, 139:318-325.
29. Banerjee K, Michael E, Eggink D, van Montfort T, Lasnik AB, Palmer KE, Sanders RW, Moore JP, Klasse PJ: Occluding the mannose moieties on human immunodeficiency virus type 1 gp120 with griffithsin improves the antibody responses to both proteins in mice. AIDS Res Hum Retroviruses 2012, 28:206-214.
14. Zha D: Glycoengineered Pichia-based expression of monoclonal antibodies. Methods Mol Biol 2013, 988:31-43. 15. Hamilton SR, Cook WJ, Gomathinayagam S, Burnina I, Bukowski J, Hopkins D, Schwartz S, Du M, Sharkey NJ, Bobrowicz P et al.: Production of sialylated O-linked glycans in Pichia pastoris. Glycobiology 2013, 23:1192-1203. First example of the production of sialylated O-glycans in yeast on a clinically relevant reporter protein TNFR2:Fc. The authors utilized the PomGnT1 enzyme to generate a-dystroglycan-type O-glycans with a high sialic acid molar content. 16. Hamilton SR, Davidson RC, Sethuraman N, Nett JH, Jiang Y, Rios S, Bobrowicz P, Stadheim TA, Li H, Choi BK et al.: Humanization of yeast to produce complex terminally sialylated glycoproteins. Science 2006, 313:1441-1443. 17. Hamilton SR, Gerngross TU: Glycosylation engineering in yeast: the advent of fully humanized yeast. Curr Opin Biotechnol 2007, 18:387-392. 18. Liu L, Li H, Hamilton SR, Gomathinayagam S, Rayfield WJ, van Maanen M, Yin KC, Hong L, Prueksaritanont T: The impact of sialic acids on the pharmacokinetics of a PEGylated erythropoietin. J Pharm Sci 2012, 101:4414-4418. 19. Liu L, Gomathinayagam S, Hamuro L, Prueksaritanont T, Wang W, Stadheim TA, Hamilton SR: The impact of glycosylation on the pharmacokinetics of a TNFR2:Fc fusion protein expressed in glycoengineered Pichia pastoris. Pharm Res 2013, 30:803-812. Building off the results reported in Ref. [15], the authors detail the pharmacokinetic profile of a highly glycosylated TNFR2:Fc fusion protein in comparison to commercial etanercept. Glycoengineered Pichia strains and processes produced the fusion protein with similar levels of total sialic acid which translated to similar in vivo PK profiles. 20. Liu L, Stadheim A, Hamuro L, Pittman T, Wang W, Zha D, Hochman J, Prueksaritanont T: Pharmacokinetics of IgG1 monoclonal antibodies produced in humanized Pichia pastoris with specific glycoforms: a comparative study with CHO produced materials. Biologicals 2011, 39: 205-210. 21. Choi BK, Actor JK, Rios S, d’Anjou M, Stadheim TA, Warburton S, Giaccone E, Cukan M, Li H, Kull A et al.: Recombinant human lactoferrin expressed in glycoengineered Pichia pastoris: effect of terminal N-acetylneuraminic acid on in vitro secondary humoral immune response. Glycoconj J 2008, 25:581-593. www.sciencedirect.com
30. de Vries RP, Smit CH, de Bruin E, Rigter A, de Vries E, Cornelissen LA, Eggink D, Chung NP, Moore JP, Sanders RW et al.: Glycan-dependent immunogenicity of recombinant soluble trimeric hemagglutinin. J Virol 2012, 86:11735-11744. 31. Yuen CT, Storring PL, Tiplady RJ, Izquierdo M, Wait R, Gee CK, Gerson P, Lloyd P, Cremata JA: Relationships between the N-glycan structures and biological activities of recombinant human erythropoietins produced using different culture conditions and purification procedures. Br J Haematol 2003, 121:511-526. 32. Shields RL, Lai J, Keck R, O’Connell LY, Hong K, Meng YG, Weikert SH, Presta LG: Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human Fcgamma RIII and antibody-dependent cellular toxicity. J Biol Chem 2002, 277:26733-26740. 33. Van Patten SM, Hughes H, Huff MR, Piepenhagen PA, Waire J, Qiu H, Ganesa C, Reczek D, Ward PV, Kutzko JP et al.: Effect of mannose chain length on targeting of glucocerebrosidase for enzyme replacement therapy of Gaucher disease. Glycobiology 2007, 17:467-478. 34. Beck A, Cochet O, Wurch T: GlycoFi’s technology to control the glycosylation of recombinant therapeutic proteins. Expert Opin Drug Discov 2010, 5:95-111. 35. Bobrowicz P, Davidson RC, Li H, Potgieter TI, Nett JH, Hamilton SR, Stadheim TA, Miele RG, Bobrowicz B, Mitchell T et al.: Engineering of an artificial glycosylation pathway blocked in core oligosaccharide assembly in the yeast Pichia pastoris: production of complex humanized glycoproteins with terminal galactose. Glycobiology 2004, 14:757-766. 36. Choi BK, Bobrowicz P, Davidson RC, Hamilton SR, Kung DH, Li H, Miele RG, Nett JH, Wildt S, Gerngross TU: Use of combinatorial genetic libraries to humanize N-linked glycosylation in the yeast Pichia pastoris. Proc Natl Acad Sci U S A 2003, 100: 5022-5027. 37. Jacobs PP, Geysens S, Vervecken W, Contreras R, Callewaert N: Engineering complex-type N-glycosylation in Pichia pastoris using GlycoSwitch technology. Nat Protoc 2009, 4:58-70. 38. Nett JH, Stadheim TA, Li H, Bobrowicz P, Hamilton SR, Davidson RC, Choi BK, Mitchell T, Bobrowicz B, Rittenhour A et al.: A combinatorial genetic library approach to target heterologous glycosylation enzymes to the endoplasmic Current Opinion in Biotechnology 2014, 30:120–127
126 Pharmaceutical biotechnology
reticulum or the Golgi apparatus of Pichia pastoris. Yeast 2011, 28:237-252. 39. Davidson RC, Nett JH, Renfer E, Li H, Stadheim TA, Miller BJ, Miele RG, Hamilton SR, Choi BK, Mitchell TI et al.: Functional analysis of the ALG3 gene encoding the Dol-P-Man: Man5GlcNAc2-PP-Dol mannosyltransferase enzyme of P. pastoris. Glycobiology 2004, 14:399-407. 40. Nett JH, Gomathinayagam S, Hamilton SR, Gong B, Davidson RC, Du M, Hopkins D, Mitchell T, Mallem MR, Nylen A et al.: Optimization of erythropoietin production with controlled glycosylationPEGylated erythropoietin produced in glycoengineered Pichia pastoris. J Biotechnol 2012, 157:198-206. 41. Zhang N, Liu L, Dumitru CD, Cummings NR, Cukan M, Jiang Y, Li Y, Li F, Mitchell T, Mallem MR et al.: Glycoengineered Pichia produced anti-HER2 is comparable to trastuzumab in preclinical study. MAbs 2011, 3:289-298. 42. Arico C, Bonnet C, Javaud C: N-glycosylation humanization for production of therapeutic recombinant glycoproteins in Saccharomyces cerevisiae. Methods Mol Biol 2013, 988:45-57. 43. Parsaie Nasab F, Aebi M, Bernhard G, Frey AD: A combined system for engineering glycosylation efficiency and glycan structure in Saccharomyces cerevisiae. Appl Environ Microbiol 2013, 79:997-1007. 44. Wang H, Song HL, Wang Q, Qiu BS: Expression of glycoproteins bearing complex human-like glycans with galactose terminal in Hansenula polymorpha. World J Microbiol Biotechnol 2013, 29:447-458. The authors describe for the first time the production of glycoproteins in H. polymorpha with N-glycans containing terminal galactose. Through the use of ALG3 and ALG11 mutations, with heterologous mammalian glycosylation genes, strains were fermented at a 3 L research scale with predominant complex glycans. 45. Cheon SA, Kim H, Oh DB, Kwon O, Kang HA: Remodeling of the glycosylation pathway in the methylotrophic yeast Hansenula polymorpha to produce human hybrid-type N-glycans. J Microbiol 2012, 50:341-348. 46. Oh DB, Park JS, Kim MW, Cheon SA, Kim EJ, Moon HY, Kwon O, Rhee SK, Kang HA: Glycoengineering of the methylotrophic yeast Hansenula polymorpha for the production of glycoproteins with trimannosyl core N-glycan by blocking core oligosaccharide assembly. Biotechnol J 2008, 3:659-668. 47. Liu B, Gong X, Chang S, Yang Y, Song M, Duan D, Wang L, Ma Q, Wu J: Disruption of the OCH1 and MNN1 genes decrease N-glycosylation on glycoprotein expressed in Kluyveromyces lactis. J Biotechnol 2009, 143:95-102. 48. De Pourcq K, Tiels P, Van Hecke A, Geysens S, Vervecken W, Callewaert N: Engineering Yarrowia lipolytica to produce glycoproteins homogeneously modified with the universal Man3GlcNAc2 N-glycan core. PLoS ONE 2012, 7:e39976. 49. De Pourcq K, Vervecken W, Dewerte I, Valevska A, Van Hecke A, Callewaert N: Engineering the yeast Yarrowia lipolytica for the production of therapeutic proteins homogeneously glycosylated with Man(8)GlcNAc(2) and Man(5)GlcNAc(2). Microb Cell Fact 2012, 11:53. 50. Vervecken W, Kaigorodov V, Callewaert N, Geysens S, De Vusser K, Contreras R: In vivo synthesis of mammalian-like, hybrid-type N-glycans in Pichia pastoris. Appl Environ Microbiol 2004, 70:2639-2646. 51. Jacobs PP, Inan M, Festjens N, Haustraete J, Van Hecke A, Contreras R, Meagher MM, Callewaert N: Fed-batch fermentation of GM-CSF-producing glycoengineered Pichia pastoris under controlled specific growth rate. Microb Cell Fact 2010, 9:93. 52. Choi BK, Warburton S, Lin H, Patel R, Boldogh I, Meehl M, d’Anjou M, Pon L, Stadheim TA, Sethuraman N: Improvement of N-glycan site occupancy of therapeutic glycoproteins produced in Pichia pastoris. Appl Microbiol Biotechnol 2012, 95:671-682. As expression hosts often have varying levels of N-glycan occupancy, the authors describe the use of LmStt3d in glycoengineered Pichia to significantly improve the occupancy of recombinant antibodies to greater than 99%. Current Opinion in Biotechnology 2014, 30:120–127
53. Nett JH, Cook WJ, Chen MT, Davidson RC, Bobrowicz P, Kett W, Brevnova E, Potgieter TI, Mellon MT, Prinz B et al.: Characterization of the Pichia pastoris protein-Omannosyltransferase gene family. PLoS ONE 2013, 8:e68325. 54. Loibl M, Strahl S: Protein O-mannosylation: what we have learned from baker’s yeast. Biochim Biophys Acta 2013, 1833:2438-2446. 55. Cukan MC, Hopkins D, Burnina I, Button M, Giaccone E, Houston Cummings NR, Jiang Y, Li F, Mallem M, Mitchell T et al.: Binding of DC-SIGN to glycoproteins expressed in glycoengineered Pichia pastoris. J Immunol Methods 2012, 386:34-42. This paper evaluates the binding of N-glycoforms and O-glycoforms on recombinant proteins to DC-SIGN, an important human lectin involved in antigen presentation and immune stimulation. The authors demonstrate mannotriose and wild-type yeast N-glycans, as well as O-glycans with a chain length of at least two mannose sugars, are able to significantly interact with DC-SIGN whereas other glycans do not. 56. Ahlen G, Strindelius L, Johansson T, Nilsson A, Chatzissavidou N, Sjoblom M, Rova U, Holgersson J: Mannosylated mucin-type immunoglobulin fusion proteins enhance antigen-specific antibody and T lymphocyte responses. PLoS ONE 2012, 7:e46959. Through the use of a highly O-glycosylated PSGL-1/mIgG2b fusion protein expressed in P. pastoris, the authors demonstrate robust APCtargeting to enhance both humoral and cellular immune responses of vaccines. 57. Argyros R, Nelson S, Kull A, Chen MT, Stadheim TA, Jiang B: A phenylalanine to serine substitution within an O-protein mannosyltransferase led to strong resistance to PMTinhibitors in Pichia pastoris. PLoS ONE 2013, 8:e62229. 58. Orchard MG, Neuss JC, Galley CM, Carr A, Porter DW, Smith P, Scopes DI, Haydon D, Vousden K, Stubberfield CR et al.: Rhodanine-3-acetic acid derivatives as inhibitors of fungal protein mannosyl transferase 1 (PMT1). Bioorg Med Chem Lett 2004, 14:3975-3978. 59. Kuroda K, Kobayashi K, Kitagawa Y, Nakagawa T, Tsumura H, Komeda T, Shinmi D, Mori E, Motoki K, Fuju K et al.: Efficient antibody production upon suppression of O mannosylation in the yeast Ogataea minuta. Appl Environ Microbiol 2008, 74: 446-453. 60. Amano K, Chiba Y, Kasahara Y, Kato Y, Kaneko MK, Kuno A, Ito H, Kobayashi K, Hirabayashi J, Jigami Y et al.: Engineering of mucin-type human glycoproteins in yeast cells. Proc Natl Acad Sci U S A 2008, 105:3232-3237. 61. Chigira Y, Oka T, Okajima T, Jigami Y: Engineering of a mammalian O-glycosylation pathway in the yeast Saccharomyces cerevisiae: production of O-fucosylated epidermal growth factor domains. Glycobiology 2008, 18: 303-314. 62. Suga M, Hatakeyama T: High-efficiency electroporation by freezing intact yeast cells with addition of calcium. Curr Genet 2003, 43:206-211. 63. Cregg JM: DNA-mediated transformation. Methods Mol Biol 2007, 389:27-42. 64. Wu S, Letchworth GJ: High efficiency transformation by electroporation of Pichia pastoris pretreated with lithium acetate and dithiothreitol. Biotechniques 2004, 36:152-154. 65. Aw R, Polizzi KM: Can too many copies spoil the broth? Microb Cell Fact 2013, 12:128. 66. Baek DS, Kim YS: Construction of a large synthetic human Fab antibody library on yeast cell surface by optimized yeast mating. J Microbiol Biotechnol 2014. (Epub ahead of print). 67. Blaise L, Wehnert A, Steukers MP, van den Beucken T, Hoogenboom HR, Hufton SE: Construction and diversification of yeast cell surface displayed libraries by yeast mating: application to the affinity maturation of Fab antibody fragments. Gene 2004, 342:211-218. 68. Chen MT, Lin S, Shandil I, Andrews D, Stadheim TA, Choi BK: Generation of diploid Pichia pastoris strains by mating and their application for recombinant protein production. Microb Cell Fact 2012, 11:91. www.sciencedirect.com
Biopharmaceutical discovery and production in yeast Meehl and Stadheim 127
69. Ioannidis JP, Karassa FB, Druyts E, Thorlund K, Mills EJ: Biologic agents in rheumatology: unmet issues after 200 trials and $200 billion sales. Nat Rev Rheumatol 2013, 9:665-673. 70. Doerner A, Rhiel L, Zielonka S, Kolmar H: Therapeutic antibody engineering by high efficiency cell screening. FEBS Lett 2014, 588:278-287. In this recent review, the authors focus on antibody engineering using surface display and related high efficiency cell screening methods. 71. Gera N, Hussain M, Rao BM: Protein selection using yeast surface display. Methods 2013, 60:15-26. 72. Zhang L, Liang S, Zhou X, Jin Z, Jiang F, Han S, Zheng S, Lin Y: Screening for glycosylphosphatidylinositol-modified cell wall proteins in Pichia pastoris and their recombinant expression on the cell surface. Appl Environ Microbiol 2013, 79:5519-5526. 73. Shaheen HH, Prinz B, Chen MT, Pavoor T, Lin S, Houston Cummings NR, Moore R, Stadheim TA, Zha D: A dual-mode surface display system for the maturation and production of monoclonal antibodies in glyco-engineered Pichia pastoris. PLoS ONE 2013, 8:e70190. In an innovative strategy for yeast cell surface display, the authors describe a method using a surface-anchored ‘bait’ Fc that enables simultaneous surface display and secretion of full-length antibodies in the same glycoengineered yeast cell. This method provides a tool for the discovery of optimized mAb sequences while shortening the cycle time needed to produce full-length mAb material for in vitro and in vivo studies. 74. Puri V, Streaker E, Prabakaran P, Zhu Z, Dimitrov DS: Highly efficient selection of epitope specific antibody through
www.sciencedirect.com
competitive yeast display library sorting. MAbs 2013, 5: 533-539. 75. Tillotson BJ, Cho YK, Shusta EV: Cells and cell lysates: a direct approach for engineering antibodies against membrane proteins using yeast surface display. Methods 2013, 60:27-37. The authors describe a direct approach to identify and optimize antibody leads against membrane proteins through the use of yeast cell surface display with solubilized whole-cell lysates of mammalian cells containing membrane-bound antigen. 76. Tillotson BJ, de Larrinoa IF, Skinner CA, Klavas DM, Shusta EV: Antibody affinity maturation using yeast display with detergent-solubilized membrane proteins as antigen sources. Protein Eng Des Sel 2013, 26:101-112. 77. Xu Y, Roach W, Sun T, Jain T, Prinz B, Yu TY, Torrey J, Thomas J, Bobrowicz P, Vasquez M et al.: Addressing polyspecificity of antibodies selected from an in vitro yeast presentation system: a FACS-based, high-throughput selection and analytical tool. Protein Eng Des Sel 2013, 26:663-670. To address polyspecificity of antibodies derived from yeast-based libraries, the authors report a flow cytometric assay to counter select antibody sequences that interact with non-specific membrane preparations. In this manner, the authors present a protocol that is added onto the existing Adimab workflow to interrogate antibody developability using FACS-based screening. 78. Ceroni A, Maass K, Geyer H, Geyer R, Dell A, Haslam SM: GlycoWorkbench: a tool for the computer-assisted annotation of mass spectra of glycans. J Proteome Res 2008, 7:1650-1659.
Current Opinion in Biotechnology 2014, 30:120–127