1,133
Views
18
CrossRef citations to date
0
Altmetric
Review

Lepidopteran cells, an alternative for the production of recombinant antibodies?

&
Pages 294-309 | Published online: 26 Apr 2012

Abstract

Monoclonal antibodies are used with great success in many different therapeutic domains. In order to satisfy the growing demand and to lower the production cost of these molecules, many alternative systems have been explored. Among them, the baculovirus/insect cells system is a good candidate. This system is very safe, given that the baculoviruses have a highly restricted host range and they are not pathogenic to vertebrates or plants. But the major asset is the speed with which it is possible to obtain very stable recombinant viruses capable of producing fully active proteins whose glycosylation pattern can be modulated to make it similar to the human one. These features could ultimately make the difference by enabling the production of antibodies with very low costs. However, efforts are still needed, in particular to increase production rates and thus make this system commercially viable for the production of these therapeutic agents.

Introduction

Antibody-based drugs are now estimated to comprise ~30% of the global biologic drug market because of their success in different therapeutic applications such as cancer, infectious diseases, inflammation and allergy. Mammalian cells, such as Chinese hamster ovary (CHO) cells are commonly used to produce therapeutic antibody molecules at the commercial level. The establishment of stable mammalian immunoglobulin expression cell lines is time-consuming and the production-purification process expensive. To satisfy the growing demand and to lower the cost of production of these molecules, many different systems are being explored. Among these, the baculovirus/insect cell system may offer a good alternative to mammalian expression systems.

The baculovirus insect cell system is widely used to produce recombinant proteins for research, diagnostic and, more recently, for use as therapeutics and vaccines. In 1983, Smith et al.Citation1 were the first to use this virus to produce recombinant proteins by constructing a baculovirus that expresses human β- interferon. In 1990, two groups, Hasemann and CapraCitation2 and zu Putlitz et al.Citation3 showed that this system can be used to produce fully functional immunoglobulins with polypeptide chains that are correctly processed, glycosylated and assembled.

Production of recombinant antibodies using the Baculovirus/insect cell system

Baculoviruses are arthropod specific viruses. Their genome consists of a large circular double stranded DNA molecule that encodes about 150 proteins.Citation4 During the very late stage of its replication, two non-essential proteins, polyhedrin (PH) and protein 10 (P10) are expressed at a very high level (). Although the polyhedrin protein is not required in vitro, it forms occlusion-bodies in which viral particles are embedded to form polyhedra that are essential in protecting the virus from UV exposure and dehydration in the environment. Baculoviruses isolated from Autographa californica (AcMNPV) or from the silkworm Bombyx mori (BmMNPV) are commonly used for the production of recombinant proteins. The ability of baculovirus to produce two non-essential proteins at very high levels, led to their development as expression vectors.Citation5 The general principle is to replace PH or P10 genes with a DNA sequence encoding a foreign protein of interest. This replacement is easily promoted by homologous recombination between DNA purified from the wild type virus and a plasmid called “transfer vector.” This vector is a bacterial plasmid containing a fragment of the viral genome encompassing the non-essential gene (i.e., Fragment EcoRI I for the PH gene) flanked with large viral DNA sequences. These flanking sequences allow for a specific homologous recombination and integration of the foreign gene into the genome of the baculovirus. The foreign gene is cloned in place of the non-essential gene, conserving all viral 3′ and 5′ non-coding sequences involved in transcription and translation control. Lepidopteran cells are co-transfected with a mixture of genomic viral DNA and recombinant transfer vector. Homologous recombination takes place between the target gene flanking sequences in the transfer vector and the viral genome. The first attempts performed inserted foreign sequences at the site of the polyhedrin gene. The recombinant viruses were then selected according to their phenotype, i.e., the inability to produce polyhedra, using plaque assays. More recently, new approaches using defective non-infectious viral DNA i.e., linearized genomes or Bacmid-derived systems have been used.Citation6,Citation7 The latter involves the deletion of an essential gene and the insertion of an origin of replication from bacteria (mini-F) into the viral genome allowing the DNA (Bacmid) to replicate in E. coli. Furthermore, the identification of many other non-essential genes has allowed the insertion and simultaneous expression of several genes into a single virus.Citation8 Finally, by using different viral or cellular promoters that present both specific transcriptional patterns and expression rates, it is possible to modulate the production of each gene.Citation9 This highly efficient and very flexible system has therefore been employed to produce full-length immunoglobulins and antibody-derived molecules. The two types of hosts that are used routinely are either established lepidopteran cells (i.e., Sf9, Sf21, Trichoplusia ni) or animals, mostly the silkworm B. mori.

Figure 1. Baculovirus replication. During the very late stage of replication, two non-essential genes the polyhedrin (PH) and P10 are transcribed at a very high level. Polyhedrin is responsible for the formation of the polyhedra structure in which the virions are embedded. The involvement of P10 remains unclear, but it is generally assumed that it plays a role in cell lysis. These characteristics are the basis for the development of baculovirus as an expression vector.

Figure 1. Baculovirus replication. During the very late stage of replication, two non-essential genes the polyhedrin (PH) and P10 are transcribed at a very high level. Polyhedrin is responsible for the formation of the polyhedra structure in which the virions are embedded. The involvement of P10 remains unclear, but it is generally assumed that it plays a role in cell lysis. These characteristics are the basis for the development of baculovirus as an expression vector.

In order to simplify the construction of a recombinant viral genome capable of expressing an antibody, “universal” baculovirus expression cassettes have been designed. An example of these cassettes is presented in . A generic cassette consists of the following: (1) a viral promoter (P10 or PH promoter) (2) a sequence encoding an Ig signal peptide (3) two unique restriction sites allowing the insertion of the heavy or light chain variable regions in frame with the upstream signal peptide sequence (4) and a downstream sequence encoding a murine or human heavy or light chain constant region.Citation10 These cassettes are flanked with viral sequences, which direct the integration of the foreign genes into a specific locus. In that case, the two transfer vectors are designed to recombine in two different loci (PH and P10). Some Ig expression cassettes based on backbone vectors containing two back-to-back promoters allow the cloning of the H and L genes within a single transfer vector.Citation11 Although a single vector facilitates the process, the use of separate H and L transfer vectors is more convenient for example when one wishes to test the effect of combining many different H and L chain sequences or to analyze the impact of several mutations on one of the two chains. Indeed in these cases the different plasmid pairs carrying either a light or heavy chain sequence can be rapidly combined with each other in all combinations to be tested. Some vectors have also been constructed for the direct cloning of cDNA encoding scFv or Fab isolated from phage-display libraries.Citation11

Figure 2. Schematic representation of the different steps involved in the construction of a baculovirus expressing a recombinant antibody. (A) General organization of specific transfer vectors used to express heavy and light chains. In this example, H and L genes are integrated at two different loci (PH and P10). (B) The cDNAs coding for the variable regions (VH and VL) are inserted in frame with the immunoglobulin signal peptide sequence and the human (or murine) heavy or light constant region. Sf9 cells were transfected with a defective non-infectious viral DNA (Bacmid or linearized viral DNA) and two transfer vectors. Infectious double-recombinant viruses generated after homologous recombination (HR) are cloned by plaque assay. (C) Polyacrylamide gel electrophoresis of the antibody purified from the cell culture supernatant of Sf9 cells which were infected with the recombinant virus (Silver staining, lane 1). Control human IgG1 (lane 2).

Figure 2. Schematic representation of the different steps involved in the construction of a baculovirus expressing a recombinant antibody. (A) General organization of specific transfer vectors used to express heavy and light chains. In this example, H and L genes are integrated at two different loci (PH and P10). (B) The cDNAs coding for the variable regions (VH and VL) are inserted in frame with the immunoglobulin signal peptide sequence and the human (or murine) heavy or light constant region. Sf9 cells were transfected with a defective non-infectious viral DNA (Bacmid or linearized viral DNA) and two transfer vectors. Infectious double-recombinant viruses generated after homologous recombination (HR) are cloned by plaque assay. (C) Polyacrylamide gel electrophoresis of the antibody purified from the cell culture supernatant of Sf9 cells which were infected with the recombinant virus (Silver staining, lane 1). Control human IgG1 (lane 2).

The most critical step for the secretion of soluble and functional recombinant antibodies is to simultaneously produce equal amounts of heavy and light chains. Different approaches have been tested to identify the best strategy for this purpose: (1) double infection with two separate heavy and light chain-expressing viruses, each gene being expressed under the control of the same viral promoter (PH or P10 promoter) and (2) the construction of a double-recombinant virus with both chains expressed independently under the same promoter.Citation2,Citation3 Both approaches can present problems. Indeed one of the fundamental characteristics of these viruses, which has led to their use as expression vectors, is that they can induce a very efficient homologous recombination during their replication, thus facilitating the generation of recombinant viruses. This activity depends on the expression of several viral genes.Citation12-Citation14 However, this characteristic can be detrimental when repeated sequences are introduced into the viral genome. While the coexpression of several foreign genes in a single baculovirus ensures that every cell expresses the genes at the same ratio and allows consistent yields of recombinant protein complexes, the duplication of a promoter may induce instability and lead to the rearrangement of the viral genome and loss of genetic information.Citation8 A number of studies have reported that expressing both H and L chains by different promoters with the same transcriptional pattern is a way of overcoming this problem. However, data indicate (Cérutti M, unpublished data) that the preparation of purified recombinant antibodies produced with H chain under the control of the P10 promoter, and L chain under the PH promoter, was highly prone to aggregation. This is because, as shown by Roelvink et al.Citation15 and Chaabihi et al.Citation16 and contrary to general assumption, the transcriptional patterns of the P10 and PH genes are similar but not identical.Citation16 P10 gene is expressed a few hours before polyhedrin, thus leading to the overexpression of the H chains before the light chains and triggering of secretion of H chains dimers (H2).Citation15 Since the correct folding of the heavy chain requires the presence of light chains, the unfolded H2 molecules may be at the origin of the formation of aggregates observed under these conditions.Citation17 Aggregation may in turn also explain the low expression rate observed in these conditions.Citation18

In conclusion, to obtain adequate and correctly folded Igs in insect cells, it is important to use promoters that direct the synthesis of very similar amounts of heavy an light chains. When 2 different promoters are used, the time course expression of each promoter has to be considered in order to prevent the premature overproduction of heavy chain when the production of the light chain is still too low.

Production in cell culture

Sf9 and High FiveTM (Invitrogen) are commonly used as host cell lines. Although there are exceptions, High Five cells (derived from the lepidopteran T ni) are generally more efficient at expressing and secreting recombinant proteins than Sf9.Citation18,Citation19 However, it should be noted that the N-glycosylation performed by High Five cells is not identical to that achieved in human cells and may even be immunogenic (see below, “Glycosylation in insect cells”).Citation20 Furthermore, the infectious titers of cell culture supernatant are very low, even at a late time after infection.Citation19 Therefore Hi-Five cells require infection using a higher titer viral supernatant generated by infecting Sf9 cells.

Several immunoglobulin isotypes from many different species, including murine IgG,Citation2,Citation3,Citation21-Citation25 chimeric IgG,Citation26-Citation30 human IgG,Citation31-Citation39 IgA,Citation40-Citation42 IgMCitation41 and IgE,Citation43-Citation45 as well as pig IgsCitation30,Citation45 have been successfully expressed in insect cells. These molecules are all correctly processed (signal peptide cleaved), glycosylated and assembled (H2L2) with a production rate varying from 0.75 to 100 mg/l, depending on the antibody (). Carayannopoulos et al.,Citation40 have reported the expression of functional monomeric human IgA as well as dimeric forms after co-infection of insect cells with 3 different recombinant baculoviruses expressing H, L and J chain. After binding to the poly-immunoglobulin receptor (pIgR) dimeric IgA is transported across the epithelial barrier to be secreted on the luminal side of the mucosa. Upon transport, the pIgR is cleaved and the extracellular portion of the molecule, the secretory component (SC), remains bound to the dimeric IgA and released. Binding of the SC to the dimeric IgA forms a secretory IgA (sIgA) and confers to this complex resistance to proteolytic enzymes. Secreted glycosylated human SC has been produced in insect cells, and the SC was able to bind very specifically to dimeric IgA.Citation46

Table 1. Some examples of antibodies or antibody-derived molecules produced in lepidopteran cells

Beside these full-length immunoglobulins, many different antibody-derived molecules have been produced e.g., mono-specific single-chain fragments, scFv,Citation47-Citation50 bi-specific scFv,Citation51 Fab.Citation18,Citation52-Citation54 The expression of scFv is more reliable compared with that of the E. coli system and does not require any re-folding step. Fusion proteins such as chimeric-hormone-antibody molecules (Choriogonadotropin fused with mouse IgG Fc domain), scFv anti-TAG72 fused with IL-2, anti-HLADR heavy chain fused with IL-2 and human Fas receptor extracellular domain fused with human IgG1 Fc domain have also been produced.Citation55-Citation58

Production in larvae

Protein production in whole animals has mostly been developed using the silkworm and the BmMNPV baculovirus due to the large size of the B. mori larvae compared with host animals susceptible to AcMNPV. Specific dual expression vectors have been constructed for the production of recombinant proteins in this host since the BmMNPV is highly restricted to B. mori.Citation59,Citation60 Fifth instar silkworms are injected with a recombinant virus and hemolymph is collected 4 to 7 d after infection. The expression rate is very high compared with the secretion observed in a cell culture.Citation61-Citation63 Indeed Reis et al.Citation60 were the first to report a production of 800 mg/l protein in hemolymph compared with 10 mg/l in the larval body. More recently, this technology has been applied to AcMNPV-derived recombinant viruses by using T. ni larvae. O’Connell et al.Citation64 have designed the “automated insect rearing system” PERLXpress, an original “scalable technology” for whole insect baculovirus expression. In this case, the larvae are infected orally with highly infectious preoccluded virus. Just 4 d after infection, the expression rate is usually in the range of g of purified Fab/kg of larvae.Citation63

Enhancing the production and secretion of recombinant antibodies

Many attempts have been made to optimize the production and secretion of glycoproteins in insect cells. These have included: (1) using alternatives promoters, such as earlier viralCitation9 or cellular promoters,Citation65,Citation66 (2) modifying or exchanging the signal peptide sequenceCitation67 (3) co-expressing key proteins implicated in the secretion machinery (e.g., chaperone proteins)Citation9,Citation24,Citation25 and (4) generating stably-transformed insect cells.Citation65 Comparable experiments were conducted in order to increase the secretion of recombinant antibodies. Usually, the authentic signal peptide sequences of secreted proteins are correctly cleaved, generating sequences identical to the N-terminal end of the parental protein. When H and L chains are expressed with a specific signal peptide, both present the expected N-terminal end. Although the exchange of the signal peptide sequence can significantly increase the production of some proteins, no significant enhancement was observed in immunoglobulin production after exchanging the signal peptide with the honeybee melittin, AcMNPV gp64 or Drosophila BiP sequences.Citation18

From the very first reports of Ig production in insect cells, the examination of whole cell lysates revealed the presence not only of fully functional H2L2 molecules, but also of unprocessed Ig polypeptides (e.g., precursor of light chains) or aggregates.Citation2 This indicated that the folding and assembly machineries may be unable to process all Ig chains synthesized during infection. Furthermore, the levels of secreted Ig have been estimated to be 10 to 100 times lower than that of intracellular molecules.Citation25 Assuming that aggregation of overexpressed proteins results from the absence of sufficient levels of ER and/or cytosolic molecular chaperones in infected cells, the co-expression of recombinant chaperones such as BiP, HSP70, PDI, calnexin, calreticulin and ERp57 has been evaluated in both cell culture and infected larvae.Citation68,Citation69

Molecular chaperone-assisted production

As early as 1994, the protein biosynthesis pathway of the insect cells has been engineered to improve solubility, assembly and secretion of recombinant Ig.Citation24 In B-cells, the chaperone BiP, a member of Hsp70 family, was first identified in association with the H chain polypeptide in the ER. This protein may have an essential role at preventing the aggregation of the H chain and by maintaining a specific conformation, until final folding in presence of light chain takes place.Citation17,Citation70 When Sf9 cells were co-infected with 3 viruses each expressing H, L and murine BiP, Hsu et al.Citation24 showed that the presence of the chaperone improved the level of soluble and functional antibodies within the cell lysates. The secretion, however, was not significantly enhanced. While the solubility of H and L chains was slightly enhanced when Hsp70 protein, a cytosolic chaperone, was co-expressed with H and L genes, the secretion increased by about 50%.Citation25 The same approach was taken to produce Ig in silkworm. A 5-fold increase in production of Ig was observed when human chaperones, calnexin, calreticulin, and BiP were coexpressed with H and L genes of a human Ig.Citation63

Despite chaperone assistance, late during viral infection when the synthesis of the recombinant protein is increasing, the insect physiology is profoundly affected compromising cellular functions such as polypeptide processing, glycosylation and secretion. Therefore as an alternative, stably transformed insect cells have been used to produce recombinant proteins.

Stably transformed insect cells

In 1997, McCaroll and KingCitation65 were the first to describe the transfection of Sf9 cells with two plasmids, one containing a selection cassette expressing an antibiotic resistance gene (i.e., neomycin resistance gene) and the other one designed for the expression of the foreign gene. Cellular promoters, such as cytoplasmic actin promoter and/or promoters from viral immediate-early genes (e.g., ie1 and ie2 from AcMNPV) that are transcribed by the cellular RNA polymerase II were used.Citation65,Citation66 Although these promoters drive very high levels of transcription in their natural environment, they allow only very low expression rates of most foreign genes (). However this system presents a significant advantage, that secreted proteins are processed and secreted more efficiently.Citation71 In 2000, Guttieri et al.Citation72,Citation73 have used this new technology to produce antibodies. Sf9 cells were transfected with 3 independent constructs, each containing one expression cassette for H, L and the neomycin resistance gene. Despite a high copy number of H and L genes integrated into the cellular genome (approx 250 copies for both chains) the production rate of IgG was very low at about 0.06 mg/l of cell culture supernatant, which is around 500-fold lower than the production obtained in the baculovirus-insect cell system expressing the same antibody.Citation74

More recently, new vectors have been developed for the continuous high-level expression of secreted proteins.Citation66 These vectors use the property of B. mori actin promoter to be stimulated with BmMNPV IE1 transactivator via the BmMNPV HR3 enhancer. This combination resulted in a 1000-fold stimulation of foreign gene expression which, at maximum cell density in a shake-culture, can reach a yield of up to 300 mg/l in High Five cells.Citation54 Thus methods to obtain relatively high yields of Igs in stable cultures are now available.

Glycosylation in insect cells

Alterating the glycosylation pattern of proteins, including antibodies, may affect their production, secretion and function. While glycans linked to the Fab domains may be directly involved in the interaction with antigenic epitopes, the role of the glycosylation of Asn297 in the CH2 constant domain is essential for many Fc-dependent effector activities. Recently, the fucosylation levels of the N-glycans in human IgG1 was reported to significantly modulate antibody dependent cellular cytotoxicity (ADCC) in vitro and in vivo.Citation75 Lepidopteran cells have been shown to perform most of the post-translational modifications including N- and O-glycosylations. The N-glycan processing is similar to that of mammals during the early steps of the glycosylation pathway in synthesizing GlcNacMan5(Fuc)GlcNac2.Citation76 However, very low levels of GNT-I activity,Citation76,Citation77 lack of GNT-II,Citation77 β1,4 galactosyltransferase,Citation78 sialyltransferasesCitation78 and the presence of a Golgi-associated N-acetylglucosaminidaseCitation79 lead to the formation of a paucimannose structure, Man3(Fuc)GlcNac2, which has not been described in mammalian cells. Therefore oligomannose-type and partially fucosylated paucimannosic glycans represent the most frequent structures identified in glycoproteins synthesized in infected or non-infected lepidopteran cells. While only α1,6 fucose was found in glycoproteins expressed in B. mori and Sf9 cells,Citation78,Citation80 α1,3 and α1,6 fucose are found in Masmestra brassicaeCitation78,Citation80 and T. ni (High five TM) cell lines.Citation20 The presence of α1,3-linked fucose, a potential allergenic epitope in these cell lines, may constitute a limitation to their use for expressing human glycoproteins.Citation81

Only little work has been done to characterize the glycosylation pattern of recombinant antibodies produced in insect cells.Citation3 Recently, two studiesCitation62,Citation82 have reported the presence of paucimannosidic and oligomannosidic glycans including α1,6 fucose without terminal sialic acids. Interestingly, when antibodies are expressed in insects, the higher Ig production rate observed in infected pupa is associated with a better processing of glycans, with 5-fold GlcNacMan3GlcNac2 structures found on N-glycans, suggesting that glycosylation may promote the expression of a new epitope involved in the secretion process.Citation63

Two strategies have been used to “humanize” glycan structures in insect cells; integrating the missing glycosyltransferases into either the cellular genomeCitation83 or the viral genome.Citation84 Using the latter approach, we have constructed a new baculovirus expressing GNT-I, GNT-II and β1–4 galactosyltransferase (Cérutti et al. unpublished data). In order to obtain a stable genetic construct without any duplicated sequences, we chose to direct the gene expression under the control of RNA polymerase II heterologous promoters. Three new specific transfer vectors that allow homologous recombination into three dispensable genes were constructed. Structural analysis of these recombinant antibodies shows that the expression of glycosyltransferase activity allows the synthesis of mono- and di-galactosylated antibodies. The impact of these glycosylations on production/secretion rates and on the Fc dependent functions is currently being examined in our laboratory.

Biological activities of recombinant antibodies produced in insect cells

The baculovirus expression system has been used for basic research to analyze the molecular basis of specificity,Citation21,Citation22,Citation41,Citation52 autoimmunityCitation32,Citation33,Citation36,Citation39,Citation41,Citation42 or the function of intact antibodies or of isolated immunoglobulin domains.Citation18,Citation40,Citation43-Citation46 It is very easy and fast to generate recombinant viruses expressing H or L bearing specific mutations and to determine the impact of these mutations on the specificity or activity of a given antibody.Citation21,Citation22,Citation41 This strategy has been successfully used to analyze the catalytic activity of the 6D9 monoclonal antibody and the specificity of the PAC1 antibody that binds to the integrin αIIbβ3 on activated platelets.Citation18,Citation52

Universal expression cassettes have also been designed either to directly clone the VH and VL domains of antibodies expressed by murine cells or by human B-cells isolated from patientsCitation26,Citation32,Citation33,Citation36,Citation39 or to convert scFv or Fab fragments selected from phage-display libraries into an intact fully human or chimeric IgG.Citation29,Citation34,Citation35,Citation37,Citation38,Citation42 The aim of many of these experiments was to isolate, characterize and validate antibodies for further therapeutic applications in the treatment of human diseases such as hemolytic disease of newborn,Citation31 cancerCitation23,Citation27,Citation56-Citation58 and infectious diseases, with the selection of neutralizing antibodies against hemorrhagic fever viruses (e.g., hantaan, Puumala viruses),Citation34,Citation35,Citation72 human cytomegalovirus,Citation37 influenza virusCitation38 or prion protein.Citation29

Besides having a high specificity and neutralizing activity, which generally depends upon their epitope specificity, some therapeutic antibodies have to express full effector activities such as complement dependent cytotoxicity (CDC), antibody dependent cellular cytotoxicity (ADCC) and phagocytosis in order to function in vivo.

Antibody dependent cellular cytotoxicity

ADCC is thought to contribute significantly to the in vivo activity of unconjugated IgG1 antibodies, as shown by the improved response to rituximab of lymphoma patients bearing the high affinity polymorphic form of FcγRIIIA (FcγRIIIA158F), the major receptor expressed by human NK cells.Citation85 This observation has led to the active search for Fc modifications, which would improve binding of IgG1 to both polymorphic forms of this receptor. Several point mutations within the lower hinge and CH2 domains of hIgG1 have been constructed, which show higher affinity for FcγRIIIA and mediate ADCC at lower doses than the corresponding wild type antibodies.Citation75 However, the most effective and widely applied modification of IgG1 for improved ADCC is defucosylation, i.e., removal of the core α1,6 fucose from the N-linked glycan moiety, which results in an approximately 50-fold increase in affinity of IgG1 for FcγRIII.Citation86 Lack of fucose in IgG1 antibodies leads to more efficient ADCC, which is most evident at lower antibody concentrations, or when antigens are expressed at low levels. Furthermore, the inhibition of ADCC observed with fucosylated IgG1 antibodies in the presence of excess free IgG (as would be present in plasma) or C3b deposition following complement activation, in contrast, is not observed when afucosylated antibodies are used. Therefore, ADCC mediated by defucosylated IgG1 takes place even in presence of excess IgGs or complement, as found in serum. In contrast, defucosylation does not affect binding to FcRn and pharmacokinetics of antibodies, making this format very appealing. Thus, several afucosylated antibodies directed against a variety of targets, such as CD20 (obinutuzumab, GA101), HER2, CCR4 and CD19, have been shown to have improved therapeutic efficacy in tumor models in vivo.Citation87-Citation90 Indeed several such antibodies are in preclinical or clinical development. With regard to other sugar moieties, the presence or absence of galactose or bisecting GlcNAc in IgG1 glycans has limited effects on ADCC, whereas total removal of N-linked glycans from hIgG1 completely abolishes both FcγRIIIA binding and ADCC.Citation91,Citation92

As mentioned above, antibodies produced in insect cells do not generally have the same glycosylation pattern as those produced in CHO; in particular they mostly lack galactose, bisecting GlcNAc and sialic acid, but bear core fucose residues. Such antibodies have been shown to mediate efficient ADCC in vitro, as expected since Gal, GlcNAc and sialic acid do not play a major role in binding to FcγRIII.Citation23,Citation27,Citation28,Citation31 Future work will no doubt see the engineering of insect cells for the production of afucosylated antibodies. Interestingly, different glycosylation patterns of recombinant antibodies have been reported in different insect tissues, such as lack of fucose reported for antibodies produced by B. mori silk gland.Citation93

Complement dependent cytotoxicity

Complement has been suggested to contribute significantly to the activity of therapeutic antibodies directed against tumor antigens expressed at the cell surface, although some controversy still remains as to its relative importance for in vivo efficacy. Antibodies bearing human IgG1, IgG3 or IgM Fc domains can theoretically bind to hexameric C1q molecule, via contact by its CH2-CH3 domain, and activate the classical pathway of complement. For therapeutic purposes, the IgG1 format has been chosen, due to its greater stability and longer in vivo half-life. Binding of C1q to antigen-bound IgG1 leads to C2, C4 and C3 cleavage, which together form the C5 convertase (C2a-C4b-C3b complex) that binds covalently to the antibody molecule itself and to cell membrane components. Cleavage of C5 by the C5 convertase leads to deposition onto the cell membrane of C5b-C9 fragments polymers (or membrane attack complex) and lysis of the target cell. Not all hIgG1 antibodies activate complement however and it appears that several factors, including antigen density, closeness of the recognized epitope to the plasma membrane, conformation or positioning of bound antibodies and other undefined factors determine whether efficient C1q binding and complement activation does take place with any specific antibody. Indeed even different antibodies of the same Ig class and directed against the same antigen may show widely different capacities to activate complement, as exemplified by anti-CD20 antibodies.Citation94

The glycosylation pattern of the antibody may also significantly influence C1q binding and complement activation. Indeed fully deglycosylated hIgG1 antibodies are inactive in C1q binding and CDC. High mannose type molecules are also defective in C1q binding. Sialic acid is fully dispensable for classical complement pathway activation.Citation95 Galactose and bisecting GlcNAc residues however have a more controversial role.Citation96-Citation98 This may be due in part to the fact that IgG1 lacking galactose (G0) can bind to mannose binding lectin (MBL) and activate the lectin pathway of complement instead of the classical pathway. Nonetheless most studies suggest that removal of Gal leads to decreased C1q binding but increased MBL binding.Citation99 So overall, CDC by some molecules may have contributions from both the classical and lectin pathways.

The influence of Ig glycosylation pattern on complement activation implies that this aspect needs to be taken into account when producing antibodies in insect cells. It is possible either to engineer insect cells expressing specific enzymes that modify the glycosylation pattern of the antibodies produced, or perform in vitro enzymatic modifications of sugar moieties, in order to enable adequate glycosylation according to the functional activity required for the final product (such as introduction of GlcNAc-Gal for CDC or inhibition of fucosylation for ADCC). It is also necessary to carefully probe the effector functions of the antibodies produced in insect cells, in order to verify CDC activity, as required. There are few studies investigating CDC activity of IgG antibodies produced in insect cells.Citation23,Citation26,Citation27,Citation100 In several cases, no CDC activation could be demonstrated, but this was true also for the parent antibody produced in mammalian cells.Citation23,Citation26 In one study, insect cell produced antibodies could indeed induce C1q binding or CDC with human serum.Citation27 However comparison with the same antibody produced in CHO cells was not made, so that the role of a different glycosylation pattern could not be verified. Furthermore, no systematic study and comparison of the effect on CDC of different glycosylaled forms produced in insect cells has yet been performed.

Phagocytosis

Phagocytosis of opsonised tumor cells is thought to be one of the major immune mediated mechanisms for control of tumor cell growth by unconjugated hIgG1 antibodies such as rituximab or trastuzumab. Phagocytosis is performed mostly by macrophages that in man express the FcγRIIIA, FcγRIIA, and FcγRI activating receptors, all of which may mediate phagocytosis, and the inhibitory FcγRIIB molecule.Citation99,Citation101 As previously mentioned, removal of core fucose residue from hIgG1 specifically affects FcγRIIIA binding, as well as ADCC by NK cells that express only this receptor.Citation86 Defucosylation of anti-CD20 or anti-CEA antibodies has been shown however to have no significant effect on phagocytosis, suggesting that receptors other than FcγRIIIA, which are not similarly affected by fucose, play a predominant role in phagocytosis.Citation102,Citation103

Whereas several antibodies produced in insect cells have been probed for their capacity to activate ADCC, data on phagocytosis by insect cell produced antibodies are even more scarce, with only one report showing activation of monocytes by anti-colorectal carcinoma antigen antibody produced in Sf9 cells. Given the likely importance of this mechanism of action, future work should investigate more systematically the capacity of such antibodies to mediate phagocytosis.

Insect cells system vs. other expression systems

Most currently marketed antibodies are produced in mammalian cells; only two (ranibizumab and certolizumab pegol) are produced in non-mammalian cells (E. coli). By making use of host cell engineering to either overcome gene silencing and negative position effect or to control apoptotic response and cell metabolism, mammalian cells have surpassed all expression systems. After optimization of culture medium and culture processes, average production yields of up to 13 g/l of recombinant proteins are possible ().Citation104-Citation106 Rodent cells such as CHO cells or murine myeloma derived cell lines Sp2/0 and NS0 are the most frequently used. These cell lines have also been chosen because the glycosylation pattern of recombinant molecules produced in such systems is very similar to human. However, these glycosylation patterns are not identical to human ones. Twenty two per cent of patients treated with anti-EGFR mAb (cetuximab) approved for the treatment of cancers and produced in Sp2/0 cell line had a severe hypersensitivity reaction. This was in response to the presence of a highly immunogenic sugar (galactose α1,3 galactose) in the Fab portion of the antibody, which is specific of the Sp2/0 cell line.Citation107

Table 2. Comparison of the different expression systems used to produce antibodies

The major limitation of utilizing mammalian systems is the high cost. This is mainly due to the very time consuming process of isolating and characterizing the best producing clone, with good growing properties and that expresses the protein with an adequate glycosylation pattern. These systems may also suffer from instability of expression, such as decline in productivity and/or the alteration of the glycosylation pattern during the production process.Citation108 Indeed recently, it has been shown that the high heterogeneity observed between transformed clones can also be observed between genetically identical cells in the same environment, expression variation in a clone being larger than variation observed between clones.Citation109,Citation110

Another method of production that avoids some of the problems of stable clones uses transient gene expression systems (TGE). Indeed large-scale transfection protocols resulted in antibody titers up to 1 g/l for HEK cells, with the overall productivity in CHO cells being 3–5-fold lower.Citation111 Although this highly promising approach may be an answer to substantially reduce the time necessary for the development of a therapeutic molecule, further research is necessary to enable this technology to be used for the production of very large amounts of therapeutic antibodies.

The demand for improved and less expensive antibody production systems led researchers to explore new expression hosts. These are summarized in . Yeasts seem to be one of the most promising systems. Saccharomyces cerevisiae and Pichia pastoris, for example, have many advantages over other eukaryotic systems because their genome can be manipulated very easily and the cells do not require expensive growth media. In 1985, Wood et al.Citation112 reported the production and secretion of functional glycosylated antibodies in S. cerevisiae. Horwitz et al.Citation113 on the other hand, found the expression level to be very low with only 0.05 mg/l. While an efficient ADCC activity was observed, recombinant IgG1 failed to induce CDC.Citation113 A 180-fold improvement in secretion was obtained by using a mutant of α mating factor 1 leader peptide and by co-expressing protein disulfide isomerase (PDI).Citation114 Evans et al.Citation115 recently reported the production at high yield (40–90 mg/l) of scFv fused to human serum albumin. With regard to glysosylation patterns in yeasts, modifications have been achieved in particular removal of non-human O-glysosylation. However, these glycans being essential for cell survival, any attempt to completely eliminate O-glycosylation were unsuccessful.Citation116 More recently, by using specific inhibitors of protein-mannosyltransferase I (Pmt) catalyzing the initial reaction of O-glycosylation in yeasts, it is possible to lower the presence of such structures. However, even in these conditions, some yeast O-glycans are still present on recombinant proteins.Citation117

The methylotrophic yeast P. pastoris has been used to produce high yields of small recombinant proteins. This host presents many advantages over S. cerevisiae, as foreign genes are expressed under a strong tightly regulated promoter induced by methanol, the promoter of alcohol oxidase I gene (AOX1), and can be grown to higher density than S. cerevisiae. However, the glycosylation pattern of recombinant proteins such as those produced in S. cerevisiae is inappropriate, with extra O-glycosylation and the addition of high-mannose type oligosaccharides.Citation116,Citation117 Strains of P. pastoris have been re-engineered to humanize the glycosylation pathway.Citation118 As a result of this re-engineering of the secretion pathway, and in contrast to proteins produced by mammalian cells, these new yeast cell lines can produce highly uniform N-linked glycans, allowing the biosynthesis of specific glycoforms.Citation119-Citation121 Recombinant immunoglobulins, such as camelid VHH, have also been produced in P. pastoris with a yield of 10–15 mg/l of secreted and biologically active protein.Citation122 The quality and the high uniformity of the glycoforms generated by these new strains were used to analyze the impact of N-glycan structures on ADCC activity of anti-CD20 recombinant antibody.Citation123 All these improvements make this system potentially very attractive.

Other potential alternatives are filamentous fungi such as Aspergillus and Trichoderma spp Fab fragments and IgG have been produced at high titer in the range of 1 g/l for IgG and Fab.Citation124,Citation125 No difference was observed in antigen binding affinity, pharmacokinetic or ADCC. However immunoglobulins were produced as a mixture of glycosylated and aglycosylated forms, with 50% of N297 glycosylation site unoccupied on heavy chains.Citation124 Glycans produced by these filamentous fungi are of high mannose-type with terminal galactose in the furanose form (Galf), with Galf2Man7GlcNac2 as the most abundant structure. This isomer of galactose is absent in mammals and may be highly immunogenic in humans.Citation126

For a long time, plants were considered low-cost alternative for the production of recombinant proteins. In 1989, Hiatt and colleaguesCitation127 were the first to demonstrate the production of a functional recombinant IgG1 in transgenic tobacco. Since then, many groups have expressed recombinant antibodies in transgenic plants.Citation128 However, these transgenic expression systems have some disadvantages: (i) yield is low, usually in the range 1–40 mg of recombinant antibody per kg of fresh biomass and (ii) the process of generating and selecting the best transgenic strain is time consuming and may require more than 2 y.Citation129 More recently, very efficient transient expression systems have been developed based on A. tumefaciens-mediated delivery (leaf infiltration) or on plant-virus vectors. In contrast with the stably transformed plants, transient expression systems lead to high-yield of functional recombinant proteins 200–500 mg/kg of fresh tissue in only several days (< 2weeks).Citation130-Citation132 However, the presence of non-human immunogenic glycans, such as β1,2 xylose and core α1,3 fucose constituted the major limitation to their use for the production of therapeutics.Citation133 Knocked-out lines for xylose transferase (Xylt1) and fucose transferases (Fuct1) of Arabidosis thaliana were also generated, allowing the biosynthesis of recombinant antibodies with homogeneous mammalian-like complex glycans.Citation134 Moreover, transgenic plants with an engineered glycan synthesis pathway have been constructed e.g., by coexpressing human β1,4 galactosyltransferase or by expressing enzymes of the sialic acid synthesis pathway.Citation135,Citation136 Application of these new transient expression technologies on an industrial scale and the improvement in glycosylation make this system a highly appealing alternative over mammalian systems.

The small aquatic higher plant Lemna minor is also considered an interesting alternative.Citation137 By co-expressing H and L chains genes and RNAi targeting Xylt1 and Fuct1, a single major N-glycan species GlcNac2Man3GlcNac2 was produced. Antibodies produced in these conditions lacking fucose presented a higher affinity for FcγRIIIA and a 20–35-fold increase in the ADCC activity.Citation138 It should be noted that, in contrast to mammalian cells, the glycan profiles are highly reproducible even under different growth conditions.

The moss Physcomitrella patens can be grown in vitro in simple culture medium of inorganic salts. The high degree of homologous recombination observed in its nuclear DNA enables a precise integration of foreign genes and efficient targeted knockouts, such as Xylt1 and Fuct1.Citation139 Specific inducible promoters and signal peptide sequences have also been identified. Moreover, human genes can be expressed in moss without previous codon adaptation.

Other candidate hosts to reduce production time include microalgae. For example, the unicellular alga Chlamydomonas reinhardtii is able to produce antibodies after complete transformation of the chloroplasts genome.Citation140 Chloroplast expression presents unique attributes over the transformation of the nuclear genome: the integration of the transgene by homologous recombination, high expression rates, the ability to process polycistronic transcripts and no gene silencing. Soluble recombinant proteins are accumulated in the chloroplast but are not secreted. Dimerization by disulfide bond formation and multimeric protein complexes can be achieved, leading to the production of functional immunoglobulins. Even though, the chloroplast does not possess any glycosylation machinery.Citation141

As for mammalian cells and all stable expression systems such as yeasts or plants, the expression levels may be highly affected by the integration site, gene sequence and the copy number of the foreign gene and it may be a very time consuming and expensive to select the best cellular clone that will give the optimal production rate and glycosylation. However, by using new baculovirus/insect cell technologies involving linearized viral DNA or bacmid-derived systems, highly stable and homogeneous recombinant virus population expressing or co-expressing 1 or several foreign genes can be generated in three days. Consequently, the selection of a viral clone is very fast and efficient, the expression rate and glycosylation patterns depending only on the genetic construct inserted into the viral genome.

Surprisingly, E. coli, which is traditionally used for the production of small proteins, could be back in competition with the production of non-glycosylated full-size antibodies. Among the protein expression systems, E. coli has always been one of the most attractive hosts. It is a rapid and inexpensive method to produce recombinant proteins and the scale-up is easy and cheap. Antibody-derived fragments such as Fab directed against vEGF (ranibizumab) and TNFα (certolizumab pegol), as well as a protein/peptide fused to the Fc domain of human IgG1 (romiplostim) have been successfully developed and approved as therapeutics in human. Furthermore, some researchers have been working on the expression of full-length immunoglobulins in E. coli. First attempts were performed by expressing full-length H and L chains as insoluble aggregates. However refolding steps required to reconstitute an active immunoglobulin were very tedious and challenging, and led to very low production rates.Citation142,Citation143 Today, by using separated cistrons, adjusting the translation levels of the two chains, removing the rare codons and RNA secondary structure, it is possible to secrete full-length IgG with titer as high as 1–2 g/l.Citation144,Citation145 The main limitation of this system is that it produces aglycosylated antibodies and these lack immune effector activities. Moreover, aglycosylated IgG were shown to be not well-folded and more sensitive to proteases.Citation98,Citation146 As mentioned above, the presence and the composition of the ubiquitous N297 glycans exert a major effect on binding of the antibody to FcγRs; their complete removal abolishes the binding to FcγRIIIa and FcγRII and reduces the interaction with FcγRI. However, recent studies demonstrated that some aglycosylated variants of IgG1 are capable of binding to FcγRs, inducing a high ADCC activity.Citation147,Citation148 These observations open new avenues in the design of therapeutic antibodies. However, it is worth noting that N297 glycans affect the overall conformation of the immunoglobulin, altering the “open” conformation observed with the fully galactosylated IgG to a “closed” conformation, potentially leading to the unmasking of epitopes or to the induction of new conformational epitopes, which again raise the question of the potential immunogenicity of such molecules.Citation149

Discussion

The therapeutic use of monoclonal antibodies in the treatment of a wide range of pathologies such as cancer, infections and inflammatory diseases has exploded over the past 15 y. First treatments using murine monoclonal antibodies rapidly demonstrated the immunogenicity of these molecules, limiting their use in humans. Recombinant chimeric, humanized or human monoclonal antibodies have then been produced using mammalian expression systems. Whereas a significant decrease in the incidence of human anti-mouse antibodies (HAMA) response was evident with these recombinant molecules, some anti-glycan response was observed when the antibodies were produced in murine cell lines expressing non-human glycan epitopes such as NS0 or Sp2/0. In addition to these problems, current systems are not sufficient to meet the demand and call for the developments for new, less expensive, simpler and faster systems.

The ideal expression system for the production of recombinant antibodies for human therapy has to meet the following criteria: (1) the molecules produced have to be biologically functional, correctly glycosylated and non-immunogenic, (2) the production rate has to be high enough to meet increasing demand, (3) the selection and characterization steps for each new molecule have to be fast, reliable and cheap and (4) the cost of the whole process, from the establishment of the expression system to the purification steps has to be as contained as possible. The baculovirus/insect cell system may have all the qualities required to produce fully active glycosylated antibodies or antibody-derived molecules. The construction of the recombinant virus is very fast, with only a few days needed to generate stable recombinant viruses. Both generation of new molecules and their production are reliable and very cheap compared with the mammalian system, including the possible use of very inexpensive serum free culture medium. Indeed the main characteristic of the BEVS is the speed with which it is possible to obtain very stable recombinant viruses that are capable of producing fully functional glycoproteins with a human glycan pattern. These specificities may ultimately make a difference, by enabling the development of therapeutic antibodies at a much lower cost. Finally, baculoviruses have a highly restricted host range limited to some invertebrates which makes this system particularly safe. Two vaccines produced in insect cells, Cervarix (GlaxoSmithKline, cervical cancer) and Provenge (Dendreon, prostate cancer) have received EMA and FDA approval and are commercially available. Many others e.g., FluBlok (Protein Sciences Corporation, Seasonal influenza vaccine) and Diamyd (Diamyd Inc., Type 1 diabetes) are in late stages of clinical trials, holding promise for this type of system also for therapeutic antibody production.Citation150 Given the importance of the therapeutic antibody market, what needs to be established is whether this system is capable of producing very high amounts of recombinant proteins. Efforts are still needed to increase production rates and thus make the system commercially viable for the production of these drugs.

Abbreviations:
AcMNPV=

Autographa californica multiple nuclear polyhedrosis virus

BmMNPV=

Bombyx mori multiple nuclear polyhedrosis virus

Ig=

Immunoglobulin

H=

heavy chain

L=

light chain

scFv=

single chain fragment variable

Fab=

fragment antibody

PDI=

Protein Disulfide Isomerase

HSP70=

Heat Shock Protein 70

BiP=

Binding immunoglobulin Protein

GNT-I=

N acetylglucosaminyltransferase I

ER=

endoplasmic reticulum

Pfu=

Plaque forming unit

Acknowledgments

M.C. was supported by Centre National de la Recherche Scientifique. J.G. was supported by the “Associzione Italiana per le Leucemie—Linfoma e Mieloma” (AIL, Rome, Italy). This publication has been funded with support from the French National Research Agency under the program “Investissements d’avenir” Grant Agreement LabEx MAbImprove: ANR-10-LABX-53 and the Italian Association for Cancer Research (AIRC, IG grant to J.G.).

References

  • Smith GE, Summers MD, Fraser MJ. Production of human beta interferon in insect cells infected with a baculovirus expression vector. Mol Cell Biol 1983; 3:2156 - 65; PMID: 6318086
  • Hasemann CA, Capra JD. High-level production of a functional immunoglobulin heterodimer in a baculovirus expression system. Proc Natl Acad Sci U S A 1990; 87:3942 - 6; http://dx.doi.org/10.1073/pnas.87.10.3942; PMID: 2111022
  • zu Putlitz J, Kubasek WL, Duchêne M, Marget M, von Specht B-U, Domdey H. Antibody production in baculovirus-infected insect cells. Biotechnology (N Y) 1990; 8:651 - 4; http://dx.doi.org/10.1038/nbt0790-651; PMID: 1367456
  • Ayres MD, Howard SC, Kuzio J, Lopez-Ferber M, Possee RD. The complete DNA sequence of Autographa californica nuclear polyhedrosis virus. Virology 1994; 202:586 - 605; http://dx.doi.org/10.1006/viro.1994.1380; PMID: 8030224
  • Miller LK. Baculoviruses as gene expression vectors. Annu Rev Microbiol 1988; 42:177 - 99; http://dx.doi.org/10.1146/annurev.mi.42.100188.001141; PMID: 3059993
  • Kitts PA, Possee RD. A method for producing recombinant baculovirus expression vectors at high frequency. Biotechniques 1993; 14:810 - 7; PMID: 8512707
  • Luckow VA, Lee SC, Barry GF, Olins PO. Efficient generation of infectious recombinant baculoviruses by site-specific transposon-mediated insertion of foreign genes into a baculovirus genome propagated in Escherichia coli.. J Virol 1993; 67:4566 - 79; PMID: 8392598
  • Noad RJ, Stewart M, Boyce M, Celma CC, Willison KR, Roy P. Multigene expression of protein complexes by iterative modification of genomic Bacmid DNA. BMC Mol Biol 2009; 10:87 - 99; http://dx.doi.org/10.1186/1471-2199-10-87; PMID: 19725957
  • Sokolenko S, George S, Wagner A, Tuladhar A, Andrich JM, Aucoin MG. Co-expression vs. co-infection using baculovirus expression vectors in insect cell culture: Benefits and drawbacks. Biotechnol Adv 2012; 30:766 - 81; http://dx.doi.org/10.1016/j.biotechadv.2012.01.009; PMID: 22297133
  • Poul M-A, Cérutti M, Chaabihi H, Devauchelle G, Kaczorek M, Lefranc M-P. Design of cassette baculovirus vectors for the production of therapeutic antibodies in insect cells. Immunotechnology 1995; 1:189 - 96; http://dx.doi.org/10.1016/1380-2933(95)00019-4; PMID: 9373347
  • Liang M, Dübel S, Li D, Queitsch I, Li W, Bautz EKF. Baculovirus expression cassette vectors for rapid production of complete human IgG from phage display selected antibody fragments. J Immunol Methods 2001; 247:119 - 30; http://dx.doi.org/10.1016/S0022-1759(00)00322-7; PMID: 11150543
  • Kamita SG, Maeda S, Hammock BD. High-frequency homologous recombination between baculoviruses involves DNA replication. J Virol 2003; 77:13053 - 61; http://dx.doi.org/10.1128/JVI.77.24.13053-13061.2003; PMID: 14645562
  • Crouch EA, Passarelli AL. Genetic requirements for homologous recombination in Autographa californica nucleopolyhedrovirus. J Virol 2002; 76:9323 - 34; http://dx.doi.org/10.1128/JVI.76.18.9323-9334.2002; PMID: 12186915
  • Mikhailov VS, Okano K, Rohrmann GF. Baculovirus alkaline nuclease possesses a 5′-->3′ exonuclease activity and associates with the DNA-binding protein LEF-3. J Virol 2003; 77:2436 - 44; http://dx.doi.org/10.1128/JVI.77.4.2436-2444.2003; PMID: 12551981
  • Roelvink PW, van Meer MMM, de Kort CAD, Possee RD, Hammock BD, Vlak JM. Dissimilar expression of Autographa californica multiple nucleocapsid nuclear polyhedrosis virus polyhedrin and p10 genes. J Gen Virol 1992; 73:1481 - 9; http://dx.doi.org/10.1099/0022-1317-73-6-1481; PMID: 1607866
  • Chaabihi H, Ogliastro MH, Martin M, Giraud C, Devauchelle G, Cérutti M. Competition between baculovirus polyhedrin and p10 gene expression during infection of insect cells. J Virol 1993; 67:2664 - 71; PMID: 8474166
  • Feige MJ, Groscurth S, Marcinowski M, Shimizu Y, Kessler H, Hendershot LM, et al. An unfolded CH1 domain controls the assembly and secretion of IgG antibodies. Mol Cell 2009; 34:569 - 79; http://dx.doi.org/10.1016/j.molcel.2009.04.028; PMID: 19524537
  • Furuta T, Ogawa T, Katsuda T, Fujii I, Yamaji H. Efficient production of an antibody Fab fragment using the baculovirus-insect cell system. J Biosci Bioeng 2010; 110:577 - 81; http://dx.doi.org/10.1016/j.jbiosc.2010.06.001; PMID: 20591732
  • Hashimoto Y, Zhang S, Blissard GW. Ao38, a new cell line from eggs of the black witch moth, Ascalapha odorata (Lepidoptera: Noctuidae), is permissive for AcMNPV infection and produces high levels of recombinant proteins. BMC Biotechnol 2010; 10:50 - 65; http://dx.doi.org/10.1186/1472-6750-10-50; PMID: 20602790
  • Hancock K, Narang S, Pattabhi S, Yushak ML, Khan A, Lin S-C, et al. False positive reactivity of recombinant, diagnostic, glycoproteins produced in High Five insect cells: effect of glycosylation. J Immunol Methods 2008; 330:130 - 6; http://dx.doi.org/10.1016/j.jim.2007.08.002; PMID: 17868684
  • Hasemann CA, Capra JD. Mutational analysis of arsonate binding by a CRIA+ antibody. VH and VL junctional diversity are essential for binding activity. J Biol Chem 1991; 266:7626 - 32; PMID: 2019590
  • Hasemann CA, Capra JD. Mutational analysis of the cross-reactive idiotype of the A strain mouse. J Immunol 1991; 147:3170 - 9; PMID: 1717589
  • Nesbit M, Fu ZF, McDonald-Smith J, Steplewski Z, Curtis PJ. Production of a functional monoclonal antibody recognizing human colorectal carcinoma cells from a baculovirus expression system. J Immunol Methods 1992; 151:201 - 8; http://dx.doi.org/10.1016/0022-1759(92)90118-D; PMID: 1629610
  • Hsu T-A, Eiden JJ, Bourgarel P, Meo T, Betenbaugh MJ. Effects of co-expressing chaperone BiP on functional antibody production in the baculovirus system. Protein Expr Purif 1994; 5:595 - 603; http://dx.doi.org/10.1006/prep.1994.1082; PMID: 7858430
  • Ailor E, Betenbaugh MJ. Overexpression of a cytosolic chaperone to improve solubility and secretion of a recombinant IgG protein in insect cells. Biotechnol Bioeng 1998; 58:196 - 203; http://dx.doi.org/10.1002/(SICI)1097-0290(19980420)58:2/3<196::AID-BIT12>3.0.CO;2-B; PMID: 10191390
  • Poul M-A, Cérutti M, Chaabihi H, Ticchioni M, Deramoudt F-X, Bernard A, et al. Cassette baculovirus vectors for the production of chimeric, humanized, or human antibodies in insect cells. Eur J Immunol 1995; 25:2005 - 9; http://dx.doi.org/10.1002/eji.1830250731; PMID: 7542600
  • Hu P, Glasky MS, Yun A, Alauddin MM, Hornick JL, Khawli LA, et al. A human-mouse chimeric Lym-1 monoclonal antibody with specificity for human lymphomas expressed in a baculovirus system. Hum Antibodies Hybridomas 1995; 6:57 - 67; PMID: 7492752
  • Troadec S, Bès C, Chentouf M, Nguyen B, Briant L, Jacquet C, et al. Biological activities on T lymphocytes of a baculovirus-expressed chimeric recombinant IgG1 antibody with specificity for the CDR3-like loop on the D1 domain of the CD4 molecule. Clin Immunol 2006; 119:38 - 50; http://dx.doi.org/10.1016/j.clim.2005.11.013; PMID: 16426893
  • Huang Y-X, Han J, Dong C-F, Sun L, Gao C, Wang X-F, et al. Generation of genetic engineering monoclonal antibodies against prion protein. Med Microbiol Immunol 2007; 196:241 - 6; http://dx.doi.org/10.1007/s00430-007-0049-y; PMID: 17486363
  • Jar AM, Osorio FA, López OJ. Mouse x pig chimeric antibodies expressed in Baculovirus retain the same properties of their parent antibodies. Biotechnol Prog 2009; 25:516 - 23; http://dx.doi.org/10.1002/btpr.113; PMID: 19301248
  • Edelman L, Margaritte C, Chaabihi H, Monchâtre E, Blanchard D, Cardona A, et al. Obtaining a functional recombinant anti-rhesus (D) antibody using the baculovirus-insect cell expression system. Immunology 1997; 91:13 - 9; http://dx.doi.org/10.1046/j.1365-2567.1997.00219.x; PMID: 9203960
  • Lieby P, Soley A, Levallois H, Hugel B, Freyssinet J-M, Cérutti M, et al. The clonal analysis of anticardiolipin antibodies in a single patient with primary antiphospholipid syndrome reveals an extreme antibody heterogeneity. Blood 2001; 97:3820 - 8; http://dx.doi.org/10.1182/blood.V97.12.3820; PMID: 11389022
  • Lieby P, Soley A, Knapp A-M, Cérutti M, Freyssinet J-M, Pasquali J-L, et al. Memory B cells producing somatically mutated antiphospholipid antibodies are present in healthy individuals. Blood 2003; 102:2459 - 65; http://dx.doi.org/10.1182/blood-2003-01-0180; PMID: 12791657
  • Koch J, Liang M, Queitsch I, Kraus AA, Bautz EKF. Human recombinant neutralizing antibodies against hantaan virus G2 protein. Virology 2003; 308:64 - 73; http://dx.doi.org/10.1016/S0042-6822(02)00094-6; PMID: 12706090
  • Liang M, Mahler M, Koch J, Ji Y, Li D, Schmaljohn C, et al. Generation of an HFRS patient-derived neutralizing recombinant antibody to Hantaan virus G1 protein and definition of the neutralizing domain. J Med Virol 2003; 69:99 - 107; http://dx.doi.org/10.1002/jmv.10259; PMID: 12436484
  • Lieby P, Poindron V, Roussi S, Klein C, Knapp A-M, Garaud J-C, et al. Pathogenic antiphospholipid antibody: an antigen-selected needle in a haystack. Blood 2004; 104:1711 - 5; http://dx.doi.org/10.1182/blood-2004-02-0462; PMID: 15166038
  • Duan T, Wang X-F, Xiao S-Y, Gu S-Y, Liang M-F. Recombinant human IgG antibodies against human cytomegalovirus. Biomed Environ Sci 2008; 21:372 - 80; http://dx.doi.org/10.1016/S0895-3988(08)60057-4; PMID: 19133610
  • Sun L, Lu X, Li C, Wang M, Liu Q, Li Z, et al. Generation, characterization and epitope mapping of two neutralizing and protective human recombinant antibodies against influenza A H5N1 viruses. PLoS One 2009; 4:e5476; http://dx.doi.org/10.1371/journal.pone.0005476; PMID: 19421326
  • Thurner L, Müller A, Cérutti M, Martin T, Pasquali J-L, Gross WL, et al. Wegener’s granuloma harbors B lymphocytes with specificities against a proinflammatory transmembrane protein and a tetraspanin. J Autoimmun 2011; 36:87 - 90; http://dx.doi.org/10.1016/j.jaut.2010.09.002; PMID: 20951001
  • Carayannopoulos L, Max EE, Capra JD. Recombinant human IgA expressed in insect cells. Proc Natl Acad Sci U S A 1994; 91:8348 - 52; http://dx.doi.org/10.1073/pnas.91.18.8348; PMID: 8078886
  • Potter KN, Li Y, Mageed RA, Jefferis R, Capra JD. Anti-idiotypic antibody D12 and superantigen SPA both interact with human VH3-encoded antibodies on the external face of the heavy chain involving FR1, CDR2 and FR3. Mol Immunol 1998; 35:1179 - 87; http://dx.doi.org/10.1016/S0161-5890(98)00103-5; PMID: 10199392
  • Fukushima N, Nalbandian G, Van De Water J, White K, Ansari AA, Leung P, et al. Characterization of recombinant monoclonal IgA anti-PDC-E2 autoantibodies derived from patients with PBC. Hepatology 2002; 36:1383 - 92; PMID: 12447863
  • Vangelista L, Laffer S, Turek R, Grönlund H, Sperr WR, Valent P, et al. The immunoglobulin-like modules Cepsilon3 and α2 are the minimal units necessary for human IgE-FcepsilonRI interaction. J Clin Invest 1999; 103:1571 - 8; http://dx.doi.org/10.1172/JCI6551; PMID: 10359566
  • Björklund JEM, Schmidt M, Magnusson CGM. Characterisation of recombinant human IgE-Fc fragments expressed in baculovirus-infected insect cells. Mol Immunol 2000; 37:169 - 77; http://dx.doi.org/10.1016/S0161-5890(00)00028-6; PMID: 10865116
  • Vernersson M, Pejler G, Kristersson T, Alving K, Hellman L. Cloning, structural analysis, and expression of the pig IgE ε chain. Immunogenetics 1997; 46:461 - 8; http://dx.doi.org/10.1007/s002510050306; PMID: 9321425
  • Rindisbacher L, Cottet S, Wittek R, Kraehenbuhl JP, Corthésy B. Production of human secretory component with dimeric IgA binding capacity using viral expression systems. J Biol Chem 1995; 270:14220 - 8; http://dx.doi.org/10.1074/jbc.270.23.14220; PMID: 7775483
  • Laroche Y, Demaeyer M, Stassen J-M, Gansemans Y, Demarsin E, Matthyssens G, et al. Characterization of a recombinant single-chain molecule comprising the variable domains of a monoclonal antibody specific for human fibrin fragment D-dimer. J Biol Chem 1991; 266:16343 - 9; PMID: 1885569
  • Kretzschmar T, Aoustin L, Zingel O, Marangi M, Vonach B, Towbin H, et al. High-level expression in insect cells and purification of secreted monomeric single-chain Fv antibodies. J Immunol Methods 1996; 195:93 - 101; http://dx.doi.org/10.1016/0022-1759(96)00093-2; PMID: 8814324
  • Lemeulle C, Chardès T, Montavon C, Chaabihi H, Mani JC, Pugnière M, et al. Anti-digoxin scFv fragments expressed in bacteria and in insect cells have different antigen binding properties. FEBS Lett 1998; 423:159 - 66; http://dx.doi.org/10.1016/S0014-5793(98)00029-5; PMID: 9512350
  • Demangel C, Zhou J, Choo ABH, Shoebridge G, Halliday GM, Britton WJ. Single chain antibody fragments for the selective targeting of antigens to dendritic cells. Mol Immunol 2005; 42:979 - 85; http://dx.doi.org/10.1016/j.molimm.2004.09.034; PMID: 15829289
  • Yoshida S, Kobayashi T, Matsuoka H, Seki C, Gosnell WL, Chang SP, et al. T-cell activation and cytokine production via a bispecific single-chain antibody fragment targeted to blood-stage malaria parasites. Blood 2003; 101:2300 - 6; http://dx.doi.org/10.1182/blood-2002-03-0831; PMID: 12411309
  • Abrams C, Deng Y-J, Steiner B, O’Toole T, Shattil SJ. Determinants of specificity of a baculovirus-expressed antibody Fab fragment that binds selectively to the activated form of integrin α IIb β 3. J Biol Chem 1994; 269:18781 - 8; PMID: 7518445
  • Bès C, Cérutti M, Briant-Longuet L, Bresson D, Peraldi-Roux S, Pugnière M, et al. The chimeric mouse-human anti-CD4 Fab 13B8.2 expressed in baculovirus inhibits both antigen presentation and HIV-1 promoter activation. Hum Antibodies 2001; 10:67 - 76; PMID: 11673661
  • Yamaji H, Manabe T, Watakabe K, Muraoka M, Fujii I, Fukuda H. Production of functional antibody Fab fragment by recombinant insect cells. Biochem Eng J 2008; 41:203 - 9; http://dx.doi.org/10.1016/j.bej.2008.04.017
  • Johnson GA, Hansen TR, Austin KJ, Van Kirk EA, Murdoch WJ. Baculovirus-insect cell production of bioactive choriogonadotropin-immunoglobulin G heavy-chain fusion proteins in sheep. Biol Reprod 1995; 52:68 - 73; http://dx.doi.org/10.1095/biolreprod52.1.68; PMID: 7711185
  • Bei R, Schlom J, Kashmiri SVS. Baculovirus expression of a functional single-chain immunoglobulin and its IL-2 fusion protein. J Immunol Methods 1995; 186:245 - 55; http://dx.doi.org/10.1016/0022-1759(95)00149-5; PMID: 7594624
  • Hu P, Hornick JL, Glasky MS, Yun A, Milkie MN, Khawli LA, et al. A chimeric Lym-1/interleukin 2 fusion protein for increasing tumor vascular permeability and enhancing antibody uptake. Cancer Res 1996; 56:4998 - 5004; PMID: 8895756
  • Muraki M, Honda S. Efficient production of human Fas receptor extracellular domain-human IgG1 heavy chain Fc domain fusion protein using baculovirus/silkworm expression system. Protein Expr Purif 2010; 73:209 - 16; http://dx.doi.org/10.1016/j.pep.2010.05.007; PMID: 20576530
  • Maeda S, Kawai T, Obinata M, Fujiwara H, Horiuchi T, Saeki Y, et al. Production of human α-interferon in silkworm using a baculovirus vector. Nature 1985; 315:592 - 4; http://dx.doi.org/10.1038/315592a0; PMID: 2989694
  • Reis U, Blum B, von Specht B-U, Domdey H, Collins J. Antibody production in silkworm cells and silkworm larvae infected with a dual recombinant Bombyx mori nuclear polyhedrosis virus. Biotechnology (N Y) 1992; 10:910 - 2; http://dx.doi.org/10.1038/nbt0892-910; PMID: 1368987
  • Sakamoto S, Pongkitwitoon B, Nakamura S, Maenaka K, Tanaka H, Morimoto S. Efficient silkworm expression of single-chain variable fragment antibody against ginsenoside Re using Bombyx mori nucleopolyhedrovirus bacmid DNA system and its application in enzyme-linked immunosorbent assay for quality control of total ginsenosides. J Biochem 2010; 148:335 - 40; http://dx.doi.org/10.1093/jb/mvq072; PMID: 20592135
  • Park EY, Ishikiriyama M, Nishina T, Kato T, Yagi H, Kato K, et al. Human IgG1 expression in silkworm larval hemolymph using BmNPV bacmids and its N-linked glycan structure. J Biotechnol 2009; 139:108 - 14; http://dx.doi.org/10.1016/j.jbiotec.2008.09.013; PMID: 18984019
  • Dojima T, Nishina T, Kato T, Uno T, Yagi H, Kato K, et al. Improved secretion of molecular chaperone-assisted human IgG in silkworm, and no alterations in their N-linked glycan structures. Biotechnol Prog 2010; 26:232 - 8; PMID: 19918885
  • O’Connell KP, Kovaleva E, Campbell JH, Anderson PE, Brown SG, Davis DC, et al. Production of a recombinant antibody fragment in whole insect larvae. Mol Biotechnol 2007; 36:44 - 51; http://dx.doi.org/10.1007/s12033-007-0014-4; PMID: 17827537
  • McCarroll L, King LA. Stable insect cell cultures for recombinant protein production. Curr Opin Biotechnol 1997; 8:590 - 4; http://dx.doi.org/10.1016/S0958-1669(97)80034-1; PMID: 9353223
  • Farrell PJ, Behie LA, Iatrou K. Transformed Lepidopteran insect cells: new sources of recombinant human tissue plasminogen activator. Biotechnol Bioeng 1999; 64:426 - 33; http://dx.doi.org/10.1002/(SICI)1097-0290(19990820)64:4<426::AID-BIT5>3.0.CO;2-#; PMID: 10397881
  • Tessier DC, Thomas DY, Khouri HE, Laliberté F, Vernet T. Enhanced secretion from insect cells of a foreign protein fused to the honeybee melittin signal peptide. Gene 1991; 98:177 - 83; http://dx.doi.org/10.1016/0378-1119(91)90171-7; PMID: 2016060
  • Yokoyama N, Hirata M, Ohtsuka K, Nishiyama Y, Fujii K, Fujita M, et al. Co-expression of human chaperone Hsp70 and Hsdj or Hsp40 co-factor increases solubility of overexpressed target proteins in insect cells. Biochim Biophys Acta 2000; 1493:119 - 24; PMID: 10978513
  • Nakajima M, Kato T, Kanamasa S, Park EY. Molecular chaperone-assisted production of human α-1,4-N-acetylglucosaminyltransferase in silkworm larvae using recombinant BmNPV bacmids. Mol Biotechnol 2009; 43:67 - 75; http://dx.doi.org/10.1007/s12033-009-9174-8; PMID: 19418270
  • Feige MJ, Hendershot LM, Buchner J. How antibodies fold. Trends Biochem Sci 2010; 35:189 - 98; http://dx.doi.org/10.1016/j.tibs.2009.11.005; PMID: 20022755
  • Douris V, Swevers L, Labropoulou V, Andronopoulou E, Georgoussi Z, Iatrou K. Stably transformed insect cell lines: tools for expression of secreted and membrane-anchored proteins and high-throughput screening platforms for drug and insecticide discovery. Adv Virus Res 2006; 68:113 - 56; http://dx.doi.org/10.1016/S0065-3527(06)68004-4; PMID: 16997011
  • Guttieri MC, Bookwalter C, Schmaljohn C, Schmaljohn CS. Expression of a human, neutralizing monoclonal antibody specific to puumala virus G2-protein in stably-transformed insect cells. J Immunol Methods 2000; 246:97 - 108; http://dx.doi.org/10.1016/S0022-1759(00)00299-4; PMID: 11121551
  • Guttieri MC, Sinha T, Bookwalter C, Liang M, Schmaljohn CS. Cassette vectors for conversion of Fab fragments into full-length human IgG1 monoclonal antibodies by expression in stably transformed insect cells. Hybrid Hybridomics 2003; 22:135 - 45; http://dx.doi.org/10.1089/153685903322286548; PMID: 12954098
  • Liang M, Guttieri M, Lundkvist A, Schmaljohn C. Baculovirus expression of a human G2-specific, neutralizing IgG monoclonal antibody to Puumala virus. Virology 1997; 235:252 - 60; http://dx.doi.org/10.1006/viro.1997.8695; PMID: 9281505
  • Shields RL, Lai J, Keck R, O’Connell LY, Hong K, Meng YG, et al. 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 - 40; http://dx.doi.org/10.1074/jbc.M202069200; PMID: 11986321
  • Marchal I, Mir AM, Kmiécik D, Verbert A, Cacan R. Use of inhibitors to characterize intermediates in the processing of N-glycans synthesized by insect cells: a metabolic study with Sf9 cell line. Glycobiology 1999; 9:645 - 54; http://dx.doi.org/10.1093/glycob/9.7.645; PMID: 10362833
  • Altmann F, Kornfeld G, Dalik T, Staudacher E, Glössl J. Processing of asparagine-linked oligosaccharides in insect cells. N-acetylglucosaminyltransferase I and II activities in cultured lepidopteran cells. Glycobiology 1993; 3:619 - 25; http://dx.doi.org/10.1093/glycob/3.6.619; PMID: 8130393
  • Lopez M, Coddeville B, Langridge J, Lemoine J, Plancke H, Chaabihi H, et al. Microheterogeneity of the oligosaccharides carried by the N-glycosylation sites of the bovine lactotransferrin expressed in Mamestra brassicae using a baculovirus vector. Glycobiology 1997; 7:635 - 51; http://dx.doi.org/10.1093/glycob/7.5.635; PMID: 9254046
  • Altmann F, Schwihla H, Staudacher E, Glössl J, März L. Insect cells contain an unusual, membrane-bound beta-N-acetylglucosaminidase probably involved in the processing of protein N-glycans. J Biol Chem 1995; 270:17344 - 9; http://dx.doi.org/10.1074/jbc.270.29.17344; PMID: 7615537
  • Staudacher E, Kubelka V, März L. Distinct N-glycan fucosylation potentials of three lepidopteran cell lines. Eur J Biochem 1992; 207:987 - 93; http://dx.doi.org/10.1111/j.1432-1033.1992.tb17134.x; PMID: 1499571
  • Prenner C, Mach L, Glössl J, März L. The antigenicity of the carbohydrate moiety of an insect glycoprotein, honey-bee (Apis mellifera) venom phospholipase A2. The role of alpha 1,3-fucosylation of the asparagine-bound N-acetylglucosamine. Biochem J 1992; 284:377 - 80; PMID: 1376112
  • Song M, Park D-Y, Kim Y, Lee K-J, Lu Z, Ko K, et al. Characterization of N-glycan structures and biofunction of anti-colorectal cancer monoclonal antibody CO17-1A produced in baculovirus-insect cell expression system. J Biosci Bioeng 2010; 110:135 - 40; http://dx.doi.org/10.1016/j.jbiosc.2010.01.013; PMID: 20547339
  • Aumiller JJ, Mabashi-Asazuma H, Hillar A, Shi X, Jarvis DL. A new glycoengineered insect cell line with an inducibly mammalianized protein N-glycosylation pathway. Glycobiology 2012; 22:417 - 28; http://dx.doi.org/10.1093/glycob/cwr160; PMID: 22042767
  • Wagner R, Liedtke S, Kretzschmar E, Geyer H, Geyer R, Klenk HD. Elongation of the N-glycans of fowl plague virus hemagglutinin expressed in Spodoptera frugiperda (Sf9) cells by coexpression of human beta 1,2-N-acetylglucosaminyltransferase I. Glycobiology 1996; 6:165 - 75; http://dx.doi.org/10.1093/glycob/6.2.165; PMID: 8727789
  • Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood 2002; 99:754 - 8; http://dx.doi.org/10.1182/blood.V99.3.754; PMID: 11806974
  • Niwa R, Natsume A, Uehara A, Wakitani M, Iida S, Uchida K, et al. IgG subclass-independent improvement of antibody-dependent cellular cytotoxicity by fucose removal from Asn297-linked oligosaccharides. J Immunol Methods 2005; 306:151 - 60; http://dx.doi.org/10.1016/j.jim.2005.08.009; PMID: 16219319
  • Niwa R, Shoji-Hosaka E, Sakurada M, Shinkawa T, Uchida K, Nakamura K, et al. Defucosylated chimeric anti-CC chemokine receptor 4 IgG1 with enhanced antibody-dependent cellular cytotoxicity shows potent therapeutic activity to T-cell leukemia and lymphoma. Cancer Res 2004; 64:2127 - 33; http://dx.doi.org/10.1158/0008-5472.CAN-03-2068; PMID: 15026353
  • Junttila TT, Parsons K, Olsson C, Lu Y, Xin Y, Theriault J, et al. Superior in vivo efficacy of afucosylated trastuzumab in the treatment of HER2-amplified breast cancer. Cancer Res 2010; 70:4481 - 9; http://dx.doi.org/10.1158/0008-5472.CAN-09-3704; PMID: 20484044
  • Herbst R, Wang Y, Gallagher S, Mittereder N, Kuta E, Damschroder M, et al. B-cell depletion in vitro and in vivo with an afucosylated anti-CD19 antibody. J Pharmacol Exp Ther 2010; 335:213 - 22; http://dx.doi.org/10.1124/jpet.110.168062; PMID: 20605905
  • Ferrara C, Grau S, Jäger C, Sondermann P, Brünker P, Waldhauer I, et al. Unique carbohydrate-carbohydrate interactions are required for high affinity binding between FcgammaRIII and antibodies lacking core fucose. Proc Natl Acad Sci U S A 2011; 108:12669 - 74; http://dx.doi.org/10.1073/pnas.1108455108; PMID: 21768335
  • Jefferis R. Recombinant antibody therapeutics: the impact of glycosylation on mechanisms of action. Trends Pharmacol Sci 2009; 30:356 - 62; http://dx.doi.org/10.1016/j.tips.2009.04.007; PMID: 19552968
  • Arnold JN, Wormald MR, Sim RB, Rudd PM, Dwek RA. The impact of glycosylation on the biological function and structure of human immunoglobulins. Annu Rev Immunol 2007; 25:21 - 50; http://dx.doi.org/10.1146/annurev.immunol.25.022106.141702; PMID: 17029568
  • Iizuka M, Ogawa S, Takeuchi A, Nakakita S, Kubo Y, Miyawaki Y, et al. Production of a recombinant mouse monoclonal antibody in transgenic silkworm cocoons. FEBS J 2009; 276:5806 - 20; http://dx.doi.org/10.1111/j.1742-4658.2009.07262.x; PMID: 19740109
  • Cragg MS, Morgan SM, Chan HT, Morgan BP, Filatov AV, Johnson PW, et al. Complement-mediated lysis by anti-CD20 mAb correlates with segregation into lipid rafts. Blood 2003; 101:1045 - 52; http://dx.doi.org/10.1182/blood-2002-06-1761; PMID: 12393541
  • Raju TS. Terminal sugars of Fc glycans influence antibody effector functions of IgGs. Curr Opin Immunol 2008; 20:471 - 8; http://dx.doi.org/10.1016/j.coi.2008.06.007; PMID: 18606225
  • Wright A, Morrison SL. Effect of C2-associated carbohydrate structure on Ig effector function: studies with chimeric mouse-human IgG1 antibodies in glycosylation mutants of Chinese hamster ovary cells. J Immunol 1998; 160:3393 - 402; PMID: 9531299
  • Hodoniczky J, Zheng YZ, James DC. Control of recombinant monoclonal antibody effector functions by Fc N-glycan remodeling in vitro.. Biotechnol Prog 2005; 21:1644 - 52; http://dx.doi.org/10.1021/bp050228w; PMID: 16321047
  • Raju ST. Impact of Fc glycosylation on monoclonal antibody effector functions and degradation by proteases. Current Trends in Monoclonal Antibody Development and Manufacturing Biotechnology: Pharmaceutical Aspects. Edited by Shire SJ, Gombotz W, Bechtold-Peters K, Andya J. New-York: Springer 2010; 11: 249-69.
  • Nimmerjahn F, Ravetch JV. Fcgamma receptors as regulators of immune responses. Nat Rev Immunol 2008; 8:34 - 47; http://dx.doi.org/10.1038/nri2206; PMID: 18064051
  • Troadec S, Chentouf M, Cérutti M, Nguyen B, Olive D, Bès C, et al. In vitro antitumoral activity of baculovirus-expressed chimeric recombinant anti-CD4 antibody 13B8.2 on T-cell lymphomas. J Immunother 2007; 30:190 - 202; http://dx.doi.org/10.1097/01.cji.0000211331.61019.26; PMID: 17471166
  • Swanson JA, Hoppe AD. The coordination of signaling during Fc receptor-mediated phagocytosis. J Leukoc Biol 2004; 76:1093 - 103; http://dx.doi.org/10.1189/jlb.0804439; PMID: 15466916
  • Bologna L, Gotti E, Manganini M, Rambaldi A, Intermesoli T, Introna M, et al. Mechanism of action of type II, glycoengineered, anti-CD20 monoclonal antibody GA101 in B-chronic lymphocytic leukemia whole blood assays in comparison with rituximab and alemtuzumab. J Immunol 2011; 186:3762 - 9; http://dx.doi.org/10.4049/jimmunol.1000303; PMID: 21296976
  • Ashraf SQ, Umana P, Mössner E, Ntouroupi T, Brünker P, Schmidt C, et al. Humanised IgG1 antibody variants targeting membrane-bound carcinoembryonic antigen by antibody-dependent cellular cytotoxicity and phagocytosis. Br J Cancer 2009; 101:1758 - 68; http://dx.doi.org/10.1038/sj.bjc.6605355; PMID: 19904275
  • Birch JR, Racher AJ. Antibody production. Adv Drug Deliv Rev 2006; 58:671 - 85; http://dx.doi.org/10.1016/j.addr.2005.12.006; PMID: 16822577
  • Jefferis R. Glycosylation as a strategy to improve antibody-based therapeutics. Nat Rev Drug Discov 2009; 8:226 - 34; http://dx.doi.org/10.1038/nrd2804; PMID: 19247305
  • Olivier S, Jacoby M, Brillon C, Bouletreau S, Mollet T, Nerriere O, et al. EB66 cell line, a duck embryonic stem cell-derived substrate for the industrial production of therapeutic monoclonal antibodies with enhanced ADCC activity. MAbs 2010; 2:405 - 15; PMID: 20562528
  • Arnold DF, Misbah SA. Cetuximab-induced anaphylaxis and IgE specific for galactose-alpha-1,3-galactose. N Engl J Med 2008; 358:2735 - , author reply 2735-6; http://dx.doi.org/10.1056/NEJMc080834; PMID: 18565869
  • Hossler P, Khattak SF, Li ZJ. Optimal and consistent protein glycosylation in mammalian cell culture. Glycobiology 2009; 19:936 - 49; http://dx.doi.org/10.1093/glycob/cwp079; PMID: 19494347
  • Kaern M, Elston TC, Blake WJ, Collins JJ. Stochasticity in gene expression: from theories to phenotypes. Nat Rev Genet 2005; 6:451 - 64; http://dx.doi.org/10.1038/nrg1615; PMID: 15883588
  • Pilbrough W, Munro TP, Gray P. Intraclonal protein expression heterogeneity in recombinant CHO cells. PLoS One 2009; 4:e8432; http://dx.doi.org/10.1371/journal.pone.0008432; PMID: 20037651
  • Geisse S. reflections on more than 10 years of TGE (Transient Gene Expression) approaches. Protein Expr Purif 2009; 64:99 - 107; http://dx.doi.org/10.1016/j.pep.2008.10.017; PMID: 19027070
  • Wood CR, Boss MA, Kenten JH, Calvert JE, Roberts NA, Emtage JS. The synthesis and in vivo assembly of functional antibodies in yeast. Nature 1985; 314:446 - 9; http://dx.doi.org/10.1038/314446a0; PMID: 3920532
  • Horwitz AH, Chang CP, Better M, Hellstrom KE, Robinson RR. Secretion of functional antibody and Fab fragment from yeast cells. Proc Natl Acad Sci U S A 1988; 85:8678 - 82; http://dx.doi.org/10.1073/pnas.85.22.8678; PMID: 3054890
  • Rakestraw JA, Sazinsky SL, Piatesi A, Antipov E, Wittrup KD. Directed evolution of a secretory leader for the improved expression of heterologous proteins and full-length antibodies in Saccharomyces cerevisiae. Biotechnol Bioeng 2009; 103:1192 - 201; http://dx.doi.org/10.1002/bit.22338; PMID: 19459139
  • Evans L, Hughes M, Waters J, Cameron J, Dodsworth N, Tooth D, et al. The production, characterisation and enhanced pharmacokinetics of scFv-albumin fusions expressed in Saccharomyces cerevisiae. Protein Expr Purif 2010; 73:113 - 24; http://dx.doi.org/10.1016/j.pep.2010.05.009; PMID: 20546898
  • 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; http://dx.doi.org/10.1038/nprot.2008.213; PMID: 19131957
  • Kuroda K, Kobayashi K, Kitagawa Y, Nakagawa T, Tsumura H, Komeda T, et al. Efficient antibody production upon suppression of O mannosylation in the yeast Ogataea minuta.. Appl Environ Microbiol 2008; 74:446 - 53; http://dx.doi.org/10.1128/AEM.02106-07; PMID: 18039826
  • Choi B-K, Bobrowicz P, Davidson RC, Hamilton SR, Kung DH, Li H, et al. 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 - 7; http://dx.doi.org/10.1073/pnas.0931263100; PMID: 12702754
  • Hamilton SR, Bobrowicz P, Bobrowicz B, Davidson RC, Li H, Mitchell T, et al. Production of complex human glycoproteins in yeast. Science 2003; 301:1244 - 6; http://dx.doi.org/10.1126/science.1088166; PMID: 12947202
  • Bobrowicz P, Davidson RC, Li H, Potgieter TI, Nett JH, Hamilton SR, 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 - 66; http://dx.doi.org/10.1093/glycob/cwh104; PMID: 15190003
  • Hamilton SR, Gerngross TU. Glycosylation engineering in yeast: the advent of fully humanized yeast. Curr Opin Biotechnol 2007; 18:387 - 92; http://dx.doi.org/10.1016/j.copbio.2007.09.001; PMID: 17951046
  • Rahbarizadeh F, Rasaee MJ, Forouzandeh M, Allameh AA. Over expression of anti-MUC1 single-domain antibody fragments in the yeast Pichia pastoris. Mol Immunol 2006; 43:426 - 35; http://dx.doi.org/10.1016/j.molimm.2005.03.003; PMID: 16337485
  • Li H, Sethuraman N, Stadheim TA, Zha D, Prinz B, Ballew N, et al. Optimization of humanized IgGs in glycoengineered Pichia pastoris. Nat Biotechnol 2006; 24:210 - 5; http://dx.doi.org/10.1038/nbt1178; PMID: 16429149
  • Ward M, Lin C, Victoria DC, Fox BP, Fox JA, Wong DL, et al. Characterization of humanized antibodies secreted by Aspergillus niger.. Appl Environ Microbiol 2004; 70:2567 - 76; http://dx.doi.org/10.1128/AEM.70.5.2567-2576.2004; PMID: 15128505
  • Nyyssönen E, Penttilä M, Harkki A, Saloheimo A, Knowles JKC, Keränen S. Efficient production of antibody fragments by the filamentous fungus Trichoderma reesei. Biotechnology (N Y) 1993; 11:591 - 5; http://dx.doi.org/10.1038/nbt0593-591; PMID: 7763606
  • Wallis GLF, Easton RL, Jolly K, Hemming FW, Peberdy JF. Galactofuranoic-oligomannose N-linked glycans of α-galactosidase A from Aspergillus niger. Eur J Biochem 2001; 268:4134 - 43; http://dx.doi.org/10.1046/j.1432-1327.2001.02322.x; PMID: 11488905
  • Hiatt A, Cafferkey R, Bowdish K. Production of antibodies in transgenic plants. Nature 1989; 342:76 - 8; http://dx.doi.org/10.1038/342076a0; PMID: 2509938
  • Peters J, Stoger E. Transgenic crops for the production of recombinant vaccines and anti-microbial antibodies. Hum Vaccin 2011; 7:367 - 74; http://dx.doi.org/10.4161/hv.7.3.14303; PMID: 21346415
  • Komarova TV, Baschieri S, Donini M, Marusic C, Benvenuto E, Dorokhov YL. Transient expression systems for plant-derived biopharmaceuticals. Expert Rev Vaccines 2010; 9:859 - 76; http://dx.doi.org/10.1586/erv.10.85; PMID: 20673010
  • Pogue GP, Vojdani F, Palmer KE, Hiatt E, Hume S, Phelps J, et al. Production of pharmaceutical-grade recombinant aprotinin and a monoclonal antibody product using plant-based transient expression systems. Plant Biotechnol J 2010; 8:638 - 54; http://dx.doi.org/10.1111/j.1467-7652.2009.00495.x; PMID: 20514694
  • Giritch A, Marillonnet S, Engler C, van Eldik G, Botterman J, Klimyuk V, et al. Rapid high-yield expression of full-size IgG antibodies in plants coinfected with noncompeting viral vectors. Proc Natl Acad Sci U S A 2006; 103:14701 - 6; http://dx.doi.org/10.1073/pnas.0606631103; PMID: 16973752
  • Huang Z, Phoolcharoen W, Lai H, Piensook K, Cardineau G, Zeitlin L, et al. High-level rapid production of full-size monoclonal antibodies in plants by a single-vector DNA replicon system. Biotechnol Bioeng 2010; 106:9 - 17; PMID: 20047189
  • Cabanes-Macheteau M, Fitchette-Lainé A-C, Loutelier-Bourhis C, Lange C, Vine ND, Ma JK, et al. N-Glycosylation of a mouse IgG expressed in transgenic tobacco plants. Glycobiology 1999; 9:365 - 72; http://dx.doi.org/10.1093/glycob/9.4.365; PMID: 10089210
  • Schähs M, Strasser R, Stadlmann J, Kunert R, Rademacher T, Steinkellner H. Production of a monoclonal antibody in plants with a humanized N-glycosylation pattern. Plant Biotechnol J 2007; 5:657 - 63; http://dx.doi.org/10.1111/j.1467-7652.2007.00273.x; PMID: 17678502
  • Bakker H, Bardor M, Molthoff JW, Gomord V, Elbers I, Stevens LH, et al. Galactose-extended glycans of antibodies produced by transgenic plants. Proc Natl Acad Sci U S A 2001; 98:2899 - 904; http://dx.doi.org/10.1073/pnas.031419998; PMID: 11226338
  • Paccalet T, Bardor M, Rihouey C, Delmas F, Chevalier C, D’Aoust MA, et al. Engineering of a sialic acid synthesis pathway in transgenic plants by expression of bacterial Neu5Ac-synthesizing enzymes. Plant Biotechnol J 2007; 5:16 - 25; http://dx.doi.org/10.1111/j.1467-7652.2006.00211.x; PMID: 17207253
  • Gasdaska J, Spencer D, Dickey L. Advantages of therapeutic protein production in the aquatic plant lemna. Bioprocess J 2003; 2:49 - 56
  • Cox KM, Sterling JD, Regan JT, Gasdaska JR, Frantz KK, Peele CG, et al. Glycan optimization of a human monoclonal antibody in the aquatic plant Lemna minor. Nat Biotechnol 2006; 24:1591 - 7; http://dx.doi.org/10.1038/nbt1260; PMID: 17128273
  • Koprivova A, Stemmer C, Altmann F, Hoffmann A, Kopriva S, Gorr G, et al. Targeted knockouts of Physcomitrella lacking plant-specific immunogenic N-glycans. Plant Biotechnol J 2004; 2:517 - 23; http://dx.doi.org/10.1111/j.1467-7652.2004.00100.x; PMID: 17147624
  • Tran M, Zhou B, Pettersson PL, Gonzalez MJ, Mayfield SP. Synthesis and assembly of a full-length human monoclonal antibody in algal chloroplasts. Biotechnol Bioeng 2009; 104:663 - 73; PMID: 19562731
  • Specht E, Miyake-Stoner S, Mayfield S. Micro-algae come of age as a platform for recombinant protein production. Biotechnol Lett 2010; 32:1373 - 83; http://dx.doi.org/10.1007/s10529-010-0326-5; PMID: 20556634
  • Boss MA, Kenten JH, Wood CR, Emtage JS. Assembly of functional antibodies from immunoglobulin heavy and light chains synthesised in E. coli. Nucleic Acids Res 1984; 12:3791 - 806; http://dx.doi.org/10.1093/nar/12.9.3791; PMID: 6328437
  • Cabilly S, Riggs AD, Pande H, Shively JE, Holmes WE, Rey M, et al. Generation of antibody activity from immunoglobulin polypeptide chains produced in Escherichia coli. Proc Natl Acad Sci U S A 1984; 81:3273 - 7; http://dx.doi.org/10.1073/pnas.81.11.3273; PMID: 6374653
  • Simmons LC, Reilly D, Klimowski L, Raju TS, Meng G, Sims P, et al. Expression of full-length immunoglobulins in Escherichia coli: rapid and efficient production of aglycosylated antibodies. J Immunol Methods 2002; 263:133 - 47; http://dx.doi.org/10.1016/S0022-1759(02)00036-4; PMID: 12009210
  • Reilly DE, Yansura DG. Production of monoclonal antibodies in E.coli. In current trends in monoclonal antibody development and manufacturing. Edited by Shire SJ, Gombotz W, Bechtold-Peters K, Andya J. New-York: Springer; 2010: 295-308.
  • Raju TS, Scallon BJ. Glycosylation in the Fc domain of IgG increases resistance to proteolytic cleavage by papain. Biochem Biophys Res Commun 2006; 341:797 - 803; http://dx.doi.org/10.1016/j.bbrc.2006.01.030; PMID: 16442075
  • Sazinsky SL, Ott RG, Silver NW, Tidor B, Ravetch JV, Wittrup KD. Aglycosylated immunoglobulin G1 variants productively engage activating Fc receptors. Proc Natl Acad Sci U S A 2008; 105:20167 - 72; http://dx.doi.org/10.1073/pnas.0809257105; PMID: 19074274
  • 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 - 67; http://dx.doi.org/10.1016/j.copbio.2011.03.002; PMID: 21420850
  • Krapp S, Mimura Y, Jefferis R, Huber R, Sondermann P. Structural analysis of human IgG-Fc glycoforms reveals a correlation between glycosylation and structural integrity. J Mol Biol 2003; 325:979 - 89; http://dx.doi.org/10.1016/S0022-2836(02)01250-0; PMID: 12527303
  • Mena JA, Kamen AA. Insect cell technology is a versatile and robust vaccine manufacturing platform. Expert Rev Vaccines 2011; 10:1063 - 81; http://dx.doi.org/10.1586/erv.11.24; PMID: 21806400

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.