Download as pdf or txt
Download as pdf or txt
You are on page 1of 38

Glycoengineered Antibodies: Towards the Next-Generation of

Immunotherapeutics

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Renato Mastrangeli1, Wolf Palinsky2, Horst Bierau1

Author affiliations:

1
Biotech Development Programme, CMC Science & Intelligence, Merck Serono SpA, an affiliate of Merck

KgaA, Darmstadt, Germany. Via Luigi Einaudi, 11. 00012 Guidonia Montecelio (Roma) – Italy

2
Biotech Development Programme, Merck Biopharma, an affiliate of Merck KgaA, Darmstadt, Germany.

Zone Industrielle de l’Ouriettaz, Aubonne 1170, Switzerland

Corresponding author: Horst Bierau, Biotech Development Programme, CMC Science & Intelligence, Merck

Serono SpA, Via Luigi Einaudi, 11. 00012 Guidonia Montecelio (Roma), Italy. E-mail

horst.bierau@merckgroup.com, Phone number: +39 0774 350834.

Keywords: monoclonal antibody, effector functions, homogeneous glycosylation, glycoengineering,

biobetter

© The Author 2018. Published by Oxford University Press. All rights reserved. For permissions, please e-mail:
journals.permissions@oup.com
1
Abstract

Monoclonal antibodies (mAbs) are currently the largest and fastest growing class of biopharmaceuticals,

and they address unmet medical needs e.g. in oncology and in auto-immune diseases. Their clinical efficacy

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


and safety is significantly affected by the structure and composition of their glycosylation profile which is

commonly heterogeneous, heavily dependent on the manufacturing process, and thus susceptible to

variations in the cell culture conditions. Glycosylation is therefore considered a critical quality attribute

(CQA) for mAbs. Commonly, in currently marketed therapeutic mAbs, the glycosylation profile is

suboptimal in terms of biological properties such as antibody-dependent cell-mediated cytotoxicity (ADCC)

or may give rise to safety concerns due to the presence of non-human glycans.

This article will review recent innovative developments in chemo-enzymatic glycoengineering, which allow

generating mAbs carrying single, well-defined, uniform Fc glycoforms, which confers the desired biological

properties for the target application. This approach offers significant benefits such as enhanced Fc effector

functions, improved safety profiles, higher batch-to-batch consistency, decreased risks related to

immunogenicity and manufacturing process changes, and the possibility to manufacture mAbs, in an

economical manner, in non-mammalian expression systems. Overall, this approach could facilitate and

reduce mAb manufacturing costs which in turn would translate into tangible benefits for both patients and

manufacturers. The first glycoengineered mAbs are about to enter clinical trials and it is expected that,

once glycoengineering reagents are available at affordable costs, and in-line with regulatory requirements,

that targeted remodelling of antibody Fc glycosylation will become an integral part in manufacturing the

next-generation of immunotherapeutics.

2
Introduction

Recombinant monoclonal antibodies (mAbs) are one of the most important class of biotherapeutics. Rising

incidences of cancer and other chronic diseases are engendering a high demand for biologics, which is

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


serving as the key contributing factor for the growth of this industry. Since 1984, more than 60 mAbs and

fusion molecules have been approved by the US Food and Drug Administration (FDA) for over 30 targets in

cancer, autoimmune, and cardiovascular diseases (Shepard et al. 2017). In a recent market research study,

the global monoclonal antibody market was valued at USD 85.4 billion in 2015 and is expected to reach a

value of USD 138.6 billion by 2024 (http://www.grandviewresearch.com/pressrelease/global-monoclonal-

antibodies-market).

Historically, monoclonal antibody development has been driven by efforts to provide patients with safer

and more efficacious products through gradual improvements over time in terms of both reduced

immunogenicity and increased potency. The first generation mAbs were 100% murine and immunogenic in

human, and therefore not suited for chronic therapy. Successively, chimeric (approximately 35% murine),

humanized (approximately 7% murine) and fully human (0% murine) antibodies were developed over time

in order to decrease mAb immunogenicity. However, despite such efforts, even fully human antibodies

such as adalimumab, the best-selling antibody drug on the market, were still reported to be immunogenic

(Matucci et al. 2016). Moreover, a recent review on the evidence of benefits on the overall survival and the

quality of life of approved cancer drugs, including a number of mAbs, concluded that most of the drugs that

entered the market between 2009 and 2013 showed only marginal gains in terms of overall survival and

quality of life (Davis et al. 2017). A similar study on cancer treatments, approved by the FDA, concluded

that only a small proportion of these drugs unequivocally show benefits on the survival and the quality of

life (Kim and Prasad 2016). These studies therefore underscore the necessity to introduce new oncology

mAbs with maximized efficacy and safety profile which in turn translate into clear benefits such as the

survival and/or the quality of life of patients.

3
Most of the mAb therapeutics are glycosylated, although non-glycosylated forms exist, e.g. when effector

functions are not desired. The present paper focusses on glycosylated mAbs.

IgG Fc glycosylation – structural and functional properties

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Monoclonal antibodies are mainly based on the prototypic antibody structure of IgG1, the most abundant

serum subclass of -immunoglobulins. An IgG1 antibody is composed of two identical heavy (H) chains

(50 kDa) and two identical κ or λ light (L) chains (25 kDa), linked together by inter-molecular disulphide

bonds. Each heavy chain consists of an N-terminal variable domain (VH) followed by three constant

domains (CH1, CH2, CH3). The so-called hinge region, linking the CH1 and the CH2 domains, contains the

cysteines residues making up the four intermolecular disulphide bonds (two between the two heavy chains

H=H and two between the heavy chain and light chain 2H-L), and confers the typical Y-shaped quaternary

structure of IgG1s (Figure 1).

Each of the two CH2 domains of the heavy chains contains a conserved IgG-Fc N-glycan site (position Asn297

according to the EU numbering). The IgG-Fc overall shape resembles that of a horseshoe. Most of the

internal Fc space between the two CH2 domains is filled by the two oligosaccharide chains (Figure 1) which

engage in glycan-glycan and glycan-protein interactions (Figure 2).

The Fc-glycans dictate stability (Bowden et al. 2012), conformation and spacing between the two CH2 low

hinge regions. Consequently, they are implicated in the binding properties with complement factor (C1q)

and Fc gamma receptors (FcRs, including the activating receptors FcγRI, FcγRIIa, FcγRIIc, FcγRIIIa and

FcγRIIIb, and the inhibitory receptor FcγRIIb) (Idusogie et al. 2000, Krapp et al. 2003, Schneider and

Zacharias 2012). Furthermore, they impact on the opening/closure of the CH2-CH3 pivot region responsible

for the binding to the neonatal Fc receptor (FcRn) and the type II receptors (including DC-SIGN and CD23)

(Pincetic et al. 2014), and the susceptibility to enzymatic degradation (Falck et al. 2015, Zheng et al. 2014).

By masking hydrophobic residues, they also decrease aggregation propensity which was found to be higher

in non-glycosylated mAbs (Kayser et al. 2011). The manner in which the Fc glycans are embedded in the

protein structure gives rise to structural fluctuations which are more dynamic than previously assumed

4
(Meier and Duus 2011); they allow various Fc conformational states (Frank et al. 2014) with the potential to

expose or shield large parts of the protein surface, and making a buried glycan accessible to processing

enzymes or binding proteins (Meier and Duus 2011). The Fc glycans are therefore crucial for the Fc

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


structure and the resulting multiple Fc effector functions (Jefferis 2012). They can have a direct influence,

when involved in binding (e.g. with FcRIIIa), or indirect influence, when involved in allosteric modulation of

the FcγRs/C1q/FcRn/type II receptor binding sites. The innate effector cells, namely NK, monocytes,

macrophages, dendritic cells, mast cells, neutrophils, eosinophils and B cells express one or several FcγRs as

well as complement factors and immune-lectin-like receptors (Euler and Alter 2015). The dynamic

modulation of the two CH2 low hinge regions, induced by the Fc glycans, plays a key role in the antibody

interaction with the Fc receptors and the C1q, allowing a fine-tuned communication with the immune

system cells, e.g. providing instructions to the innate immune system on how (by antibody-dependent cell-

mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), or complement-

dependent cytotoxicity (CDC) it should destroy the cells to which that antibody is bound. These effector

functions play a pivotal role in the mAbs used in oncology and contribute to the clearance of cancer cells.

For example, the abovementioned effector functions are all part of the mode of action (MoA) of rituximab,

the first mAb approved (1997) for cancer treatments (Jaglowski et al. 2010) .

The glycan residues on the 1-6 and 1-3 arm of each Fc-N-glycan are characterized by differential

interactions and mobility (Fortunato and Colina 2014, Krapp et al. 2003). The 1-6 arm is more rigid, while

the 1-3 arm is more flexible (Figure 2).

The galactose on the 1-3 arm has a high mobility and is more accessible to human alpha2-6 sialyltransferase

(ST6), leading to an innate 1-3 arm preferential sialylation also on released glycans (Barb, Brady, and

Prestegard 2009). On the contrary, galactose on the 1-6 arm is involved in four hydrogen bonds which

decrease the mobility and further stabilize the Fc structure (Raju 2008). It has been reported that

galactosylated mAbs have different properties in terms of Fab orientation (distribution of directional and

rotational vectors) as well as distribution of radii of gyration in comparison with agalactosylated mAbs

5
(Fortunato and Colina 2014). This may further translate in a differential impact on the functional properties

of galactosylated in comparison with agalactosylated antibodies.

The Fc oligosaccharides and their 1-3 and 1-6 arm combination represent a special case among

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


glycoproteins with almost each of the oligosaccharide components directly or indirectly eliciting structural

or functional effects. The binding stoichiometry between mAbs and FcRs or C1q is 1:1, while between

mAbs and FcRn or type II receptors it is 1:2 (Pincetic et al. 2014).

Functional impact of sialylation and galactosylation. An intriguing aspect of Fc-glycans is the differential

impact of sialic acid linkage 2,6 versus 2,3 on the anti-inflammatory properties of antibodies(Anthony,

Nimmerjahn, et al. 2008, Anthony, Wermeling, et al. 2008, Sondermann et al. 2013). Human endogenous

Fc-glycans contain exclusively 2,6-linked sialic acid residues. Fc-2,6 sialylated glycans allow the Fc domain to

interconvert between an open and a closed conformation, thus increasing its conformational plasticity and

introducing novel functional properties, e.g. the capacity of the closed conformation to bind to the type II

receptors DC-SIGN (Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin) and

CD23 (Pincetic et al. 2014, Sondermann et al. 2013). In this manner, the 2,6 sialic acid confers anti-

inflammatory properties to IgG (Anthony and Ravetch 2010, Anthony, Wermeling, et al. 2008, Anthony,

Wermeling, and Ravetch 2012, Scanlan, Burton, and Dwek 2008). On the contrary, CHO cell lines (unless

genetically modified (El et al. 2013, Yang et al. 2015) cannot produce 2,6 sialylated glycans. Instead, they

produce only glycans with 2,3 sialic acid linkages, which however do not have anti-inflammatory properties

(Anthony, Nimmerjahn, et al. 2008, Anthony, Wermeling, et al. 2008). For intravenous immunoglobulins

(IVIGs), which are used for the treatment of different auto-immune diseases, and which commonly contain

comparatively low sialylation levels (<10%) (Huang et al. 2012), 2,6 sialylation has been reported to

improve anti-inflammatory properties (Anthony and Ravetch 2010).

Galactosylation is also implicated with anti-inflammatory properties. Highly galactosylated, as opposed to

agalactosylated immune-complexes, bind to the FcγRIIB and suppress the complement component 5a

receptor 1-related (C5aR) pro-inflammatory properties during complement activation (Karsten et al. 2012).

6
Consequently, both galactosylated and 2,6 sialylated Fc-glycoforms can modulate the suppression of the

inflammatory responses. On the contrary, immune-complexes formed by agalactosylated Fc-glycoforms are

pro-inflammatory as they can only activate the activating FcγRs (Karsten et al. 2012, Nimmerjahn and

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Ravetch 2010, Collin and Ehlers 2013) but not the inhibitory FcγRIIB. Fc-glycan removal ablates both anti-

and pro-inflammatory responses (Kaneko, Nimmerjahn, and Ravetch 2006, Anthony and Nimmerjahn 2011)

(Albert et al. 2008). This underscores the relevance of the Fc-glycans in fine-tuning the interactions of the

Fc domain with its cognate receptors and thus, in their regulation (Pincetic et al. 2014). Consequently, both

2,6 sialylated (S2(α2,6)G2F) and galactosylated (G2F) Fc-glycans are contributing factors to the anti-

inflammatory properties of antibodies.

Differently from endogenous antibodies, which generally contain higher levels of galactosylated and 2,6

sialylated glycans, commercial antibody therapeutics commonly contain mainly agalactosylated glycans

(Batra and Rathore 2016, Jefferis 2012), and this can be expected to clearly differentiate their anti-

inflammatory and functional properties.

Generally, the Fc glycosylation is considered not to directly affect the mAb’s interaction with the FcRn

(Pawlowski et al. 2018). However, indirectly, specific glycan chains may indeed have a potential role on the

FcRn-mediated clearance given the Fab interaction with the FcRn (Jensen et al. 2017), the impact of the Fc-

glycan residues on the Fab orientation (Fortunato and Colina 2014), as well as the potential modulation of

opening/closure of the FcRn binding site (Chen et al. 2017). Also, patient-related factors influence the

impact of the Fc glycans on efficacy, safety and immunogenicity of therapeutic mAbs. There will be a

differential impact depending on the type (soluble or membrane-bound), and specificity of the target, the

indication, the patient’s FcγRs genetic polymorphism, and the potential presence of pre-existing antibodies

against certain glycoforms in a given patient population, e.g. anti-agalactosyl, anti-galactose-α-1,3-

galactose (anti--gal), anti-N-Glycolylneuraminic acid (anti-NGNA) (Lu et al. 2007, Zhu and Hurst 2002,

Macher and Galili 2008). Given the relatively high content of agalactosylated glycans in commercial, fully

human mAbs, such as adalimumab (Raju and Jordan 2012), it might be speculated that their observed

immunogenicity can, in part, be attributed to the presence of pre-existing antibodies against the
7
agalactosylated glycoforms in RA patients (Lu et al. 2007), or to the immunogenic potential of the

agalactosylated/high-mannose glycoforms which have the ability to engage with mannose receptors on

dendritic cells (Dong, Storkus, and Salter 1999).

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Regarding ADCC, controversial results have been reported on the impact of galactosylation, e.g. no impact

(Boyd, Lines, and Patel 1995, Shinkawa et al. 2003), or positive impact (Houde et al. 2010). The

controversial ADCC results may be explained by the differential impact of galactose on the two arms (1-6

and/or 1-3) of the Fc-glycan; the galactose on the 1-6 arm has been associated with the enhanced ADCC,

while the opposite has been observed when present on the 1-3 arm (Chung et al. 2014). Of interest, Houde

clearly showed the significantly higher binding to FcRIIIa, compared to the wild-type, when the mAb was

non-fucosylated and hypergalactosylated (approximately an 80-fold increase) in comparison to the non-

fucosylated agalactosylated glycoform (approximately a 40-fold increase) (Houde et al. 2010). Furthermore,

it has been suggested that a particular pairing of oligosaccharides is necessary for the optimum binding of

IgG to C1q, FcγRs. The apparent contradiction among some glycosylation data may thus partially be due to

the lack of information of the mode of pairing of the two oligosaccharides. The Fc portion can be

asymmetric as well as symmetric with respect to glycosylation, and the different pairing adds further

complexity to the functional studies. The information concerning the pairing of the Fc-glycan is also

relevant in understanding the biological significance of the heterogeneities of the IgG glycans (Masuda et

al. 2000).

The level of galactosylation was also found to be correlated with ADCP with higher galactosylation levels

resulting in stronger ADCP (Chung et al. 2014).

An enzymatic approach to study the functional impact of glycovariants has been reported by Roche in

which the same starting material can be degalactosylated, hypergalactosylated, and sialylated by using

commercially available enzymes and activated sugars (Thomann et al. 2015). The above study clearly shows

the relevance of galactosylation on ADCC which is higher in fucosylated hypergalactosylated glycans.

8
Endogenous human IgG1 and differences with commercial, cell culture-derived mAbs. Regarding the overall

Fc-glycan composition, the endogenous IgG1 glycan structures are almost completely biantennary complex

glycans (99.4% complex, 0.6% hybrid, <0.1% HM) with an extensive micro-heterogeneity due to the

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


presence or absence of different terminal sugars: Sialic acid, Gal, GlcNAc, core Fuc and bisecting GlcNAc.

Total agalactosylated glycoforms (G0+G0F+G0FB) were found at a level of approximately ~ 16% (Flynn et al.

2010). However, elevated levels of agalactosylated glycans are found in humans under specific conditions

such as aging, inflammatory, and autoimmune diseases (Dall'Olio et al. 2013). Moreover, in rheumatoid

arthritis (RA) patients, elevated levels of anti-agalactosylated IgG and Rheumatoid Factors (RF) are present,

which bind preferentially to the agalactosylated glycoforms (Ichikawa et al. 1998).

During pregnancy, changes in the maternal immune adaptation are required to tolerate the foetus, and to

initiate and maintain a successful pregnancy. Changes include an increased level of galactosylation in the Fc

glycan profile of IgG in both healthy individuals and RA patients (Ruhaak et al. 2014). RA patients

experience amelioration of symptoms during pregnancy, followed by a worsening after delivery, which

leads to a decrease in the galactosylation level. In RA patients during pregnancy, galactosylation, but not

sialylation, is associated with lower disease activity (Bondt et al. 2013).

In the serum of patients suffering from rheumatoid arthritis and other autoimmune diseases,

agalactosylated IgGs are present in abundance. This, together with the observation that arthritis-like

symptoms could be induced by the infusion of agalactosylated IgG, but not by G2 IgG, into non-diseased

murine recipients, lead to the hypothesis that agalactosylated IgG play a potential role in the disease

pathogenesis (Dong, Storkus, and Salter 1999). A potential explanation suggested the involvement of the

mannose receptor, which is expressed on macrophages and dendritic cells, which binds glycans containing

exposed fucose, mannose, and N-acetylglucosamine residues (e.g. agalactosylated glycoforms). Ligand

uptake is continuous, due to receptor recycling, allowing the intracellular processing of large quantities of

ligands (Dong, Storkus, and Salter 1999). The dendritic cell uptake of agalactosylated IgG, but not G2 IgG,

was highly efficient (within 15 min after exposure of ligands), and was inhibited by mannose (Dong, Storkus,

and Salter 1999). The processing and the presentation of agalactosylated IgG-derived peptides could
9
stimulate CD4+ T-cell responses. Therefore, the agalactosylated and the high mannose IgG glycoforms may

potentially alter function, safety, catabolism and antigenicity.

As mentioned above, most commercial monoclonal antibodies contain high levels of agalactosylated

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


glycans (Batra and Rathore 2016, Jefferis 2012) as well as a discrete amount of high mannose glycans. This

is presumably attributable to decreased glycan processing, culture process parameters, e.g. low dissolved

oxygen (DO) concentration, or a reduced culture reduction potential (CRP), which have been reported to

decrease galactosylation (Dionne, Mishra, and Butler 2017). In addition, the presence of sialidases (Dorai

and Ganguly 2014) and other glycosidases (Gramer and Goochee 1993) in the cell culture medium may

further decrease the galactosylation/sialylation level of mAbs.

In commercial mAbs, the agalactosylated and the high-mannose glycans level may potentially confer an

inflammatory and immunogenic potential especially in autoimmune diseases. The inflammatory potential

may further be increased in the non-fucosylated agalactosylated component. In such conditions, it is likely

that mAb therapeutics with increased levels of galactosylation and 2,6 sialic acid contents, would exhibit

superior profiles in terms of safety, efficacy, PK, and immunogenicity.

In addition to the Fc-glycosylation, approximately 10-20% of endogenous antibodies are also glycosylated in

the variable domain of the Fab region, with glycans commonly having a different glycosylation profile than

the Fc-glycans (Mimura et al. 2007, Spiegelberg et al. 1970). The fact that the glycans in the variable

domain are generally more exposed than the Fc-glycans raises safety concerns when non-human glycan

residues such as N-Glycolylneuraminic acid (NGNA) and galactose-α-1,3-galactose (-gal) are involved

(Spiegelberg et al. 1970). Therefore, mAb glycosylation is a relevant CQA (Reusch and Tejada 2015) to be

tightly monitored in the NBE development, in comparability studies following manufacturing changes, and

in the biosimilar development (comparability with the reference medicinal product).

In summary, several concerns associated with commercial mAb preparations could be addressed by closer

mimicking physiological, human-like glycosylation patterns.

Therapeutic mAb manufacturing - current state of the art

10
The development of therapeutic mAbs is a lengthy and expensive process. Manufacturers are facing

increasing demands in product quality and affordable drugs by authorities. Moreover, the emergence of

biosimilars exerts additional cost pressure. In conventional biopharmaceutical manufacturing processes, it

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


is generally accepted that the process itself defines the product which is characterized by numerous quality

attributes. Among these, glycosylation is an important subset as it has profound effects in terms of efficacy

and safety.

Mammalian host systems are the preferred expression platforms because they allow for the highest level of

posttranslational processing and functional activity of the protein. The most prominent host cell lines

currently used for marketed products are Chinese Hamster Ovary (CHO) or mouse myeloma (NS0, SP2/0)

cell lines (Dumont et al. 2016, Kunert and Reinhart 2016). However, as a consequence, their glycosylation

profiles may differ significantly from those produced in human cells, raising concerns regarding

immunogenicity and safety, e.g. due to the potential presence of non-human α-Gal or NGNA residues

introduced by mouse cells (Chung et al. 2008, Jefferis 2009) and, at lower levels, also in CHO (Bosques et al.

2010, Montjovent et al. 2017). For example, administration of mAbs containing α-Gal in patients with pre-

existing IgE antibodies directed against the α-Gal epitope can lead to hypersensitivity reactions (Platts-Mills

et al. 2015).

When antibodies are produced at industrial scale, the N-glycosylation machinery of the host cell lines

define the key Fc-glycan structure (Dell and Morris 2001). The glycosylation processing within the cells

involves a complex series of enzymatic reactions catalyzed by membrane-bound glycosidases (stepwise

trimming) and glycosyltransferases (stepwise addition of new sugar) (Helenius and Aebi 2001). Attached

key glycans depend on the cell type and the physiological status of the cell; the expression and the activity

of the cellular enzymes are exquisitely sensitive to events which take place within the cell. Depending on

the extension of processing, the N-glycans are diversified in the Golgi apparatus in high mannose, hybrid

and complex type (Shi and Goudar 2014) (Figure 3), and the transition from hybrid- to complex-type further

stabilizes the Fc region (Bowden et al. 2012).

11
Culture media components and process parameters such as pH, temperature, etc. may also affect glycan

composition (Batra and Rathore 2016, Liu, Nowak, et al. 2016). In addition, once a glycoprotein is secreted,

glycosidases (e.g. sialidases) released by lysed cells, especially at a later stage of the bioprocess, may

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


remove terminal glycan residues (Dorai and Ganguly 2014, Gramer and Goochee 1993).

Depending on the cell type and process, the principle structures emerging are high mannose, hybrid, and

complex type (see Figure 3). Within complex type glycans, the overall composition and structure may vary

significantly, e.g. through the presence/absence of core fucose, the degree of terminal galactosylation (G0,

G1, G2), the presence/absence and type of terminal sialic acid substitution N-Acetylneuraminic acid

(NANA), NGNA, the presence/absence of -gal. The resulting glycan profile may lead to a differential

impact on structure, stability, biological functions, pharmacokinetics, immunogenicity, and safety.

One way to improve the antibody properties is to introduce a better control on the composition of the Fc-

glycosylation, in particular, the reduction of fucosylation with the aim to increase ADCC. Current

optimization efforts focus on host cell engineering and the cell culture conditions (El et al. 2013, Imai-

Nishiya et al. 2007, Yang et al. 2015, Bruhlmann et al. 2017, Bruhlmann et al. 2015). For example, to

improve ADCC, a significant improvement through cell-based glycoengineering has been previously

reported with the first approved mAbs mogamulizumab and obinutuzumab. Mogamulizumab (POTELIGEO®,

KW-0761) is a humanized mAb derived from Kyowa Hakko Kirin's POTELLIGENT (®) technology which uses a

α-1,6-fucosyltransferase (FUT8) knockout CHO cell line to produce mAbs with non-fucosylated glycan

mixtures (Beck and Reichert 2012). Obinutuzumab (Gazyva™, GA-101) is derived from Roche GlycoMAb®

technology which overexpresses β1,4-Nacetylglucosaminyltransferase III (GnTIII). Once the GnT-III adds a

bisecting GlcNAc to an oligosaccharide, the core-fucosylation is inhibited (Cameron and McCormack 2014).

Both technologies produce therapeutic mAbs with enhanced ADCC activity. Furthermore, the use of

fluorinated fucose analogues as a specific inhibitor of fucosyltransferase was reported to substantially

reduce core fucosylation of N-linked glycans in CHO cells (Brühlmann et al. 2015, Rillahan et al. 2012) and

can therefore be expected to enhance ADCC activity of mAbs through rational media design.

12
However, there are still some limitations with regards to maximized ADCC properties that can be obtained

for oncology mAbs. Although this approach allows more control over the fucosylation level, these platforms

still produce heterogeneous mixtures of the non-fucosylated glycoforms containing predominantly

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


agalactosylated glycoforms, as reported for mogamulizumab. Elevated levels of the agalactosylated glycan

variants result from impaired glycan processing since processing largely depends on the presence of core

fucose and, consequently, non-fucosylated forms tend to be less processed (Castilho et al. 2015, Mimura et

al. 2018).

From a process economic point of view, the volumetric yields of mammalian cells are comparatively low

compared to expression in other eukaryotic systems such as yeast. Yeast expression systems (e.g.

Saccharomyces cerevisiae and Pichia pastoris) achieve rapid cell growth and high-protein yields with facile

production scalability (Gerngross 2004), and have most of the subcellular machinery for posttranslational

modification present in higher eukaryotes. However, a key challenge associated with yeast expression is

their production of high mannose glycans which may confer a short half-life and render proteins less

efficacious and even immunogenic in humans (Dean 1999, Gemmill and Trimble 1999). Therefore, cell line

development efforts in yeast have focused on genetic modifications to reduce the high mannose content,

and the production of glycoproteins, with humanized sialylated glycans, has been reported (Hamilton and

Gerngross 2007).

In summary, the glycan distribution defines to a large extent, the functional (with regards to effector

functions) and safety profile of a manufactured mAb. The pain points resulting from current manufacturing

processes may be summarized as follows. For patients, the efficacy and safety may be suboptimal, and

manufacturers struggle with time-consuming process development, and inefficient manufacturing

processes prone to process changes and batch-to-batch variability.

Current approaches to in-vitro glycoengineering. Enzymatic glycoengineering refers to an in-vitro

modification of glycans with the aim to reduce glycan heterogeneity. The protocols typically employ

enzymes targeting terminal sugar residues (neuraminidase, β(1,4) galactosidase, β(1,4)

13
galactosyltransferase, and (2,6) sialyltransferase) and suitable activated sugars (e.g. UDP-gal, CMP-NANA)

to increase the galactosylation and/or sialylation level on mAbs. Thomann et al. describes the workflow of

the in-solution modification of a monoclonal antibody at medium scale (Thomann et al. 2015). Similarly,

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Tayi reports the sequential enzymatic transformation on a solid-state matrix, e.g. protein A and G columns

(Tayi et al 2018). The immobilisation of the antibody in the latter approach facilitates the processing by

allowing the enzymes and buffers to be washed out in repeated cycles of enzymatic reactions. The use of

affinity resins commonly used in mAb purification may be further optimized for the implementation in

large-scale manufacturing processes. However, although the end products may be derived from the same

starting material, and despite the possibility to introduce “human-like” sialic acid with 2,6 linkages, such in-

vitro glycoengineering approaches are not able to deliver a truly homogeneous glycosylation. For example,

this approach does not allow modifying high mannose glycans or transforming hybrid into complex glycans.

Therefore, although enrichment of a desired complex glycan variant can be achieved, a certain overall

heterogeneity arising from the presence of high-mannose and hybrid variants will remain in the final

product. As a consequence, this methodology, in its current format, is not applicable to proteins expressed

in wild type yeast due to the high content of high mannose glycans. As a further limitation, although the

target glycan is enriched, the final products contain also still heterogeneous mixtures of monosialylated and

disialylated glycans and several rounds of enzymatic reactions may be required in order to reach the

desired degree of sialylation (Thomann et al. 2015, Tagy et al. 2018). Furthermore, some enzymatic

reactions may lead to an increased level of Asn deamidation and Asp isomerization (Thomann et al. 2015).

Homogeneous glycans and the “universal” glycan

The functional properties (e.g. ADCC) in therapeutic mAbs may be enhanced through Fc-glycoforms that are

both non-fucosylated and hyper-galactosylated. However, the enhanced potency carries also the potential

risk of an immunogenic reaction towards non-diseased cells also bearing the antibody target, underlining

the importance of target specificity (Dekkers et al. 2017).

14
Wong et al. coined the term “glycoantibody”, referring to a homogeneous population of mAbs having a

single, uniform glycoform on the Fc region. The same group also coined the term “universal” glycan for the

non-fucosylated, fully sialylated biantennary glycan chain (S2(α2,6)G2) which, when homogeneously added

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


to a mAbs, confers several improved properties in terms of humanization of sialic acid linkage, significantly

improved binding to the FcγRIIIa (both VV and FF polymorphisms), increased ADCC (while maintaining the

binding properties to other receptors such as FcγRs and C1q), increased ADCC of S2(α2,6)G2 in comparison

to S2(α2,3)G2, and a higher conformational plasticity between the open and closed Fc conformation (Chen

et al. 2017, Lin et al. 2015). The closed conformation is suggested to optimize the binding properties to

FcRn (Chen et al. 2017), this in turn could result in a decreased clearance rate of mAbs. The latter feature

can represent a differentiating factor between the glycan chains (S2(α2,6)G2) and G2. Further studies

would be necessary to establish if the closed conformation of non-fucosylated (S2(α2,6)G2) enhance the

binding properties with both the FcRn and the type II Fc receptors. The type II Fc receptors are known to

confer anti-inflammatory properties to the closed conformation of fucosylated (S2(α2,6)G2F) antibodies

(Sondermann et al. 2013).

It is worth noting that non-fucosylated glycoengineered mAbs not only maximize ADCC “in vivo”, where a

high amount of approximately 10mg/mL of endogenous antibodies compete for the FcRs but they can also

enhance macrophage/monocyte induced ADCP activity (Herter et al. 2014). ADCP is a well-recognized and

relevant mechanism of action of therapeutic antibodies used in oncology (Weiskopf and Weissman 2015).

Chemoenzymatic glycoengineering to obtain homogeneously glycosylated antibodies. Chemoenzymatic

glycoengineering refers to an in-vitro remodelling of glycans, employing enzymes such as endoglycosidases

and glycosynthase. Details on such a platform can be found in the recent review by Mimura (Mimura et al.

2018) as well as in the protocol by Tang (Tang, Wang, and Huang 2017) on chemoenzymatic synthesis of

glycoengineered IgG antibodies and glycosite-specific antibody-drug conjugates (ADCs). Briefly,

chemoenzymatic glycoengineering is based on two enzymatic steps: the trimming of all the heterogeneous

N-glycans by an endoglycosidase leaving the first GlcNac residue and the adding back a well-defined N-

glycan with desired properties en bloc by glycosyntase mutants which, as a substrate, use a synthetic N-
15
glycan core oxazoline. By co-expressing endoglycosidases (EndoH), the first step can potentially be omitted,

as the deglycosylation occurs in-vivo (Bennett et al. 2018). This platform has a high flexibility with the

potential to produce non-fucosylated, fucosylated, and functionalized Fc glycans.

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


The platform may be applied to both mAbs, antibody mixtures such as IVIGs (e.g. to obtain highly sialylated

(>90%) IVGs), and as a functionalization platform to produce ADCs or other labelling modifications. The

feasibility of this approach has been reported by several groups (see next section). Furthermore, it has been

shown that this approach, apart from the specific transformation of the mAb glycans, does not affect other

posttranslational modifications (Liu, Giddens, et al. 2016). Until now, this platform has mainly been

employed to generate symmetric and homogeneous glycosylation populations for structural and functional

studies (Kurogochi et al. 2015, Lin et al. 2015).

Case studies. Chemoenzymatic glycoengineering platforms were successfully employed by several groups,

amongst which Academia Sinica, National Taiwan University, CHO Pharma Inc. (Taiwan) and the University

of Maryland, Rockefeller University (USA) have produced pivotal papers (Huang et al. 2012, Li, Zhu, et al.

2017, Lin et al. 2015, Tang, Wang, and Huang 2017, Tang et al. 2016, Zou et al. 2011). A few of these

published studies use commercial mAbs as reference models, e.g. Rituxan (rituximab) (Li, DiLillo, et al.

2017, Lin et al. 2015), Herceptin (trastuzumab) (Chen et al. 2017, Kurogochi et al. 2015, Li, DiLillo, et al.

2017, Lin et al. 2015, Liu et al. 2018) and Humira (adalimumab) (Tsai et al. 2017).

A common important finding reported in these studies, although the actual fold increase differed, was that

the glycoengineered mAbs with a homogeneous glycosylation showed clearly enhanced FcRIIIa binding

compared to heterogeneously glycosylated reference samples (wild type), (Li, DiLillo, et al. 2017, Lin et al.

2015). Moreover, FcRIIIa binding was found to be consistently higher in the non-fucosylated form of the

molecule (both G2 and S2(α2,6)G2) in comparison not only to wildtype but also in comparison to the

glycoengineered mAbs which contain homogeneous non-fucosylated agalactosylated and M3 glycoforms

(Lin et al. 2015) (Figure 4).

Some patients may become resistant to rituximab due to high dosage and long-term use. It has been shown

“in vitro” that rituximab resistant cells are, however, still sensitive to glycoengineered mAb (S2(α2,6)G2)
16
presumably due to their higher affinity (Lin et al. 2015). The S2(α2,6)G2 and the G2 glycoengineered mAbs

maximize ADCC but can also enhance ADCP (Herter et al. 2014). The benefits of glycoengineered mAbs in

terms of enhanced ADCC and ADCP activity in presence of competitive endogenous antibodies due to their

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


enhanced binding to FcRIII are worth underlining; in addition, Chen demonstrated the differential binding

activity to FcRIIIa of trastuzumab in absence/presence of the target (Chen et al. 2017). In presence of the

target, a strong decrease in affinity was observed for wildtype trastuzumab (Kd = 24 nM  Kd = 169 nM),

while the decrease was minimal for the glycoengineered trastuzumab (S2(α2,6)G2) (Kd = 10 nM Kd = 20

nM). Due to the Fc conformational plasticity (open/closed conformation) induced by S2(α2,6)G2, and/or

differential impact of G2 vs. G0 on Fabs orientation (distribution of directional and rotational vectors), one

can speculate that, similarly to the FcRIII binding site, the binding sites for the other FcRs and C1q, may

also be less affected by the presence of the target ligands. If confirmed, this may translate in higher effector

function (ADCC, ADCP and CDC) in the tumor microenvironment of S2(α2,6)G2and G2 glycoengineered

mAbs.

Glycoengineering was also applied to IVIGs with the aim of increasing the 2,6-sialic acid content (from <10%

to >90%). Furthermore, the platform is also applicable to the generation of functionalized glycans, suitable

for click-chemistry for the targeted introduction of payloads, e.g. cytotoxic drug development (e.g.

antibody-drug conjugates, ADCs), or for labelling and tracers for in-vitro and in-vivo cell imaging,

immunohistochemistry and western blotting (Huang et al. 2012, Tang, Wang, and Huang 2017).

Finally, first clinical trials are underway applying glycoengineered mAbs. CHO Pharma Inc., a Taiwan-based

start-up company, has applied its proprietary glycan-based technology platforms to develop truly

homogeneously glycosylated therapeutics that are intended as "biobetters" of currently marketed

antibodies. A Phase I open-label, multiple-dose study of a glycoengineered anti-CD20 antibody (Trial ID:

NCT03221348) has been initiated (March 2018) for Refractory or Relapsed Follicular Lymphoma indication

while others are in pre-clinical development phases.

Some suppliers already commercialize reagents that are required for glycan engineering. For example,

Fushimi Pharmaceutical Co. Ltd. (Japan) offers enzymes and substrates for the targeted remodelling of

17
protein glycans at lab scale. Genovis (Sweden) commercializes soluble and immobilized enzymes required

for deglycosylation of mAbs (up to 100 mg scale), as well as kits with broader applications such as cytotoxic

drug development, in-vitro and in-vivo cell imaging, immunohistochemistry and western blotting.

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Chemoenzymatic glycoengineering – advantages. The pain points associated with current manufacturing

processes, i.e. the suboptimal safety and efficacy, and batch-to-batch inconsistencies, could collectively be

addressed by having maximum control over the Fc-glycosylation of the end products. For example, instead

of relying on the host cell machinery, the chemoenzymatic glycoengineering approach is a radically novel

effort in which the glycans that are attached during the cell culture phase are entirely remodelled by

replacing them with those optimal for the target application, e.g. humanized N-glycans, maximized efficacy.

In this manner, the final product can be produced with precisely defined and desired glycan profiles, e.g.

homogeneous, human-like glycans with enhanced biological properties. Moreover, the resulting glycan

profile is also uncoupled from the process, i.e. independent from the expression host, and unaffected by

process variations and changes.

The chemoenzymatic glycoengineering approach offers thus several significant advantages.

 Maximized efficacy – Studying homogeneous glycan populations in Rituximab, Lin et al. (Lin et al.

2015) has shown that non-fucosylated and completely galactosylated (G2) or human-like sialylated

mAbs (S2(α2,6)G2) are superior (approx. 4-6 fold higher FcγRIIIa binding compared with non-

fucosylated agalactosylated glycans (G0) and high mannose variants. Compared to the

heterogeneous wildtype glycan population, an increase in terms of the FcγRIIIa binding of up to 20-

fold was observed for the homogeneous S2(α2,6)G2 variant. Analogous to Li et al. 2017, who

observed a correlation between in-vitro binding and enhanced in-vivo activity (Li et al. 2017), it can

be expected that the maximized binding translates into maximised in vivo efficacy. This emphasizes

that an antibody, which is chemo-enzymatically engineered for a particular homogeneous glycan

population, is not only superior to currently marketed mAbs carrying a conventional,

18
heterogeneous glycan population, but also superior compared with low-fucose products derived

from processes with glycosylation improvements on the cell-line level.

 Maximized safety – Chemoenzymatic glycoengineering allows precise control over the glycan

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


structures present in a product. Therefore, any potential presence of non-human variants, high

mannose, and undesired agalactosylated glycans can be remodelled. Moreover, the precise tuning

of the glycan content allows avoiding immunogenicity and adverse reactions due to pre-existing

antibodies in patients, e.g. anti--Gal, anti-NGNA, anti-agalactosyl (Ichikawa et al. 1998, Nishijima,

Sato, and Takehara 2001, Lu et al. 2007, Zhu and Hurst 2002, Macher and Galili 2008, Teranishi et

al. 2002).

 Increased process flexibility and efficiency –

o Facilitated process development – Development is shortened timewise because less time

would be required to fine-tune the fermentation conditions to achieve a desired

glycosylation profile. The pivotal role of the intracellular functions of N-linked glycans in

terms of protein folding, oligomerization, quality control, sorting, and transport is retained

with this platform (Helenius and Aebi 2001). Differently glycosylated end products can be

produced from the same intermediate bulk. Furthermore, the approach can facilitate also

functional studies when applied to current products.

o Free choice of expression host – The production of glycosylated mAbs is no longer

restricted to mammalian cells. Instead, other eukaryotic expression hosts or even plant

cells could thus be selected for maximum productivity (Bennett et al. 2018, Liu et al. 2018).

o More robust processes and increased batch-to-batch consistency – During the re-

modelling of the glycan profile, after the fermentation, any change in glycosylation,

because of a process change, will be normalized.

Chemoenzymatic glycoengineering – challenges and limitations. Current manufacturing plants and

analytical methods are not optimised for the application of this platform and should be redesigned to apply

this procedure in the most effective way. Additional processing steps, glycan removal and re-attachment

19
will be required for glycan remodeling. The additional use of enzymes and oligosaccharide reagents poses

the risk to introduce additional HCPs or process related impurities. This will necessitate additional analytical

methods to monitor their clearance and may require additional purification steps to remove newly

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


introduced process related impurities. The handling times, additional equipment and reagents will add to

the overall manufacturing costs, although implementation of chemoenzymatic glycoengineering may be

facilitated by the integration of additional processing steps into existing operations, e.g. enzymatic

reactions on solid-phases like protein A columns (Tayi et al 2018).

Currently, enzymes and oligosaccharide reagents are not yet commercially available at relevant production

scales. However, once dedicated equipment and reagents will be produced for mAb manufacturing, their

unit costs will go down with scale.

Conclusions and perspectives

Chemoenzymatic glycoengineering is emerging as a radical and attractive approach to gain unprecedented

control over the glycosylation profile of mAbs. It therefore offers the possibility to generate mAbs with

precisely defined and desired glycans, resulting in superior product quality and safety, or in other targeted

functionalisation such as ADCs.

Additional processing costs will be offset by a significant cost-saving potential, due to the possibility of

more economic process formats with significantly higher volumetric productivities (Kurogochi et al. 2015,

Liu et al. 2018).

Currently, this platform is focused on Fc-glycans, however, by using suitable reagents and enzymes, it may

become generally applicable to glycoproteins as a platform for glycan remodelling to generate superior

products. For example, it has been suggested that among interferon-beta-1a glycoforms those with higher-

antennarity could better sustain bioactivity over time (Mastrangeli et al. 2014).

20
Once the first glycoengineered antibody, obtained by this approach, will demonstrate to be superior in the

clinic use, and customized reagents and equipment will be commercially available at affordable costs and

in-line with regulatory requirements, the platform is likely to become a breakthrough, paving the way for

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


the development and production of the next-generation of immunotherapeutics.

Acknowledgements: The authors would like to thank Maria Concetta Audino for support regarding

literature searches and Hervé Broly for critically reviewing the manuscript.

21
References

Albert H, Collin M, Dudziak D, Ravetch JV, Nimmerjahn F. 2008. In vivo enzymatic modulation of IgG

glycosylation inhibits autoimmune disease in an IgG subclass-dependent manner. Proc. Natl. Acad. Sci. U. S.

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


A 105 (39):15005-15009.

Anthony RM, Nimmerjahn F. 2011. The role of differential IgG glycosylation in the interaction of antibodies

with FcgammaRs in vivo. Curr Opin Organ Transplant 16 (1):7-14.

Anthony RM, Nimmerjahn F, Ashline DJ, Reinhold VN, Paulson JC, Ravetch JV. 2008. Recapitulation of IVIG

anti-inflammatory activity with a recombinant IgG Fc. Science 320 (5874):373-376.

Anthony RM, Ravetch JV. 2010. A novel role for the IgG Fc glycan: the anti-inflammatory activity of

sialylated IgG Fcs. J Clin Immunol 30 Suppl 1:S9-14.

Anthony, RM, Wermeling F, Karlsson MC, and Ravetch JV. 2008. Identification of a receptor required for the

anti-inflammatory activity of IVIG. Proc. Natl. Acad. Sci. U. S. A 105 (50):19571-19578.

Anthony RM, Wermeling F, Ravetch JV. 2012. Novel roles for the IgG Fc glycan. Ann. N. Y. Acad. Sci

1253:170-180.

Barb AW, Brady EK, Prestegard JH. 2009. Branch-specific sialylation of IgG-Fc glycans by ST6Gal-I.

Biochemistry 48 (41):9705-9707.

Batra J, Rathore AS. 2016. Glycosylation of monoclonal antibody products: Current status and future

prospects. Biotechnol. Prog 32 (5):1091-1102.

Beck A, Reichert JM. 2012. Marketing approval of mogamulizumab: a triumph for glyco-engineering. MAbs

4 (4):419-425.

Bennett LD, Yang Q, Berquist BR, Giddens JP, Ren Z, Kommineni V, Murray RP, White EL, Holtz BR, Wang LX,

Marcel S. 2018. Implementation of Glycan Remodeling to Plant-Made Therapeutic Antibodies. Int J Mol Sci

19 (2).

22
Bondt A, Selman MH, Deelder AM, Hazes JM, Willemsen SP, Wuhrer M, Dolhain RJ. 2013. Association

between galactosylation of immunoglobulin G and improvement of rheumatoid arthritis during pregnancy

is independent of sialylation. J Proteome. Res 12 (10):4522-4531.

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Bosques CJ, Collins BE, Meador III JW, Sarvaiya H, Murphy JL, Dellorusso G, Bulik DA, Hsu IH, Washburn N,

Sipsey SF, Myette JR, Raman R, Shriver Z, Sasisekharan R, Venkataraman G. 2010. Chinese hamster ovary

cells can produce galactose-alpha-1,3-galactose antigens on proteins. Nat. Biotechnol 28 (11):1153-1156.

Bowden TA, Baruah K, Coles CH, Harvey DJ, Yu X, Song BD, Stuart DI, Aricescu AR, Scanlan CN, Jones EY,

Crispin M. 2012. Chemical and structural analysis of an antibody folding intermediate trapped during glycan

biosynthesis. J Am. Chem. Soc 134 (42):17554-17563.

Boyd PN, Lines AC, Patel AK. 1995. The effect of the removal of sialic acid, galactose and total carbohydrate

on the functional activity of Campath-1H. Mol. Immunol 32 (17-18):1311-1318.

Brühlmann D, Jordan M, Hemberger J, Sauer M, Stettler M, Broly H. 2015. Tailoring recombinant protein

quality by rational media design. Biotechnol Prog. 31(3):615-29.

Brühlmann D, Muhr A, Parker R, Vuillemin T, Bucsella B, Kalman F, Torre S, La Neve F, Lembo A, Haas T,

Sauer M, Souquet J, Broly H, Hemberger J, Jordan M. 2017. Cell culture media supplemented with raffinose

reproducibly enhances high mannose glycan formation. J Biotechnol 252:32-42.

Cameron F, McCormack PL. 2014. Obinutuzumab: first global approval. Drugs 74 (1):147-154.

Castilho A, Gruber C, Thader A, Oostenbrink C, Pechlaner M, Steinkellner H, Altmann F. 2015. Processing of

complex N-glycans in IgG Fc-region is affected by core fucosylation. MAbs 7 (5):863-870.

Chen CL, Hsu JC, Lin CW, Wang CH, Tsai MH, Wu CY, Wong CH, Ma C. 2017. Crystal Structure of a

Homogeneous IgG-Fc Glycoform with the N-Glycan Designed to Maximize the Antibody Dependent Cellular

Cytotoxicity. ACS Chem. Biol. 12 (5), 1335–1345.

23
Chung AW, Crispin M, Pritchard L, Robinson H, Gorny MK, Yu X, Bailey-Kellogg C, Ackerman ME, Scanlan C,

Zolla-Pazner S, Alter G. 2014. Identification of antibody glycosylation structures that predict monoclonal

antibody Fc-effector function. AIDS.

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Chung CH, Mirakhur B, Chan E, Le QT, Berlin J, Morse M, Murphy BA, Satinover SM, Hosen J, Mauro A,

Slebos RJ, Zhou Q, Gold D, Hatley T, Hicklin DJ, Platts-Mills TA. 2008. Cetuximab-induced anaphylaxis and

IgE specific for galactose-alpha-1,3-galactose. N Engl J Med 358 (11):1109-17.

Collin M, Ehlers M. 2013. The carbohydrate switch between pathogenic and immunosuppressive antigen-

specific antibodies. Exp. Dermatol 22 (8):511-514.

Dall'Olio F, Vanhooren V, Chen CC, Slagboom PE, Wuhrer M, Franceschi C. 2013. N-glycomic biomarkers of

biological aging and longevity: a link with inflammaging. Ageing Res Rev 12 (2):685-698.

Davis C, Naci H, Gurpinar E, Poplavska E, Pinto A, Aggarwal A. 2017. Availability of evidence of benefits on

overall survival and quality of life of cancer drugs approved by European Medicines Agency: retrospective

cohort study of drug approvals 2009-13. BMJ 359:j4530.

Dean N. 1999. Asparagine-linked glycosylation in the yeast Golgi. Biochim Biophys Acta 1426 (2):309-22.

Dekkers G, Treffers L, Plomp R, Bentlage AEH, de Boer M, Koeleman CAM, SLissenberg-Thunnissen SN,

Visser R, Brouwer M, Mok JY, Matlung H, van den Berg TK, van Esch WJE, Kuijpers TW, Wouters D, Rispens

T, Wuhrer M, Vidarsson G. 2017. Decoding the Human Immunoglobulin G-Glycan Repertoire Reveals a

Spectrum of Fc-Receptor- and Complement-Mediated-Effector Activities. Front Immunol 2;8:877.

Dell A, Morris HR. 2001. Glycoprotein structure determination by mass spectrometry. Science 291

(5512):2351-2356.

Dionne B, Mishra N, Butler M. 2017. A low redox potential affects monoclonal antibody assembly and

glycosylation in cell culture. J Biotechnol 246:71-80.

Dong X, Storkus WJ, Salter RD. 1999. Binding and uptake of agalactosyl IgG by mannose receptor on

macrophages and dendritic cells. J Immunol 163 (10):5427-5434.


24
Dorai, H, Ganguly S. 2014. Mammalian cell-produced therapeutic proteins: heterogeneity derived from

protein degradation. Curr Opin Biotechnol 30:198-204.

Dumont J, Euwart D, Mei B, Estes S, Kshirsagar R. 2016. Human cell lines for biopharmaceutical

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


manufacturing: history, status, and future perspectives. Crit Rev Biotechnol 36 (6):1110-1122.

El Mai N, Donadio-Andrei S, Iss C, Calabro V, RoninC. 2013. Engineering a human-like glycosylation to

produce therapeutic glycoproteins based on 6-linked sialylation in CHO cells. Methods Mol. Biol 988:19-29.

Euler Z, Alter G. 2015. Exploring the potential of monoclonal antibody therapeutics for HIV-1 eradication.

AIDS Res Hum Retroviruses 31 (1):13-24.

Falck D, Jansen BC, Plomp R, Reusch D, Haberger M, Wuhrer M. 2015. Glycoforms of Immunoglobulin G

Based Biopharmaceuticals Are Differentially Cleaved by Trypsin Due to the Glycoform Influence on Higher-

Order Structure. J Proteome. Res 14 (9):4019-4028.

Flynn GC, Chen X, Liu YD, Shah B, Zhang Z. 2010. Naturally occurring glycan forms of human

immunoglobulins G1 and G2. Mol. Immunol 47 (11-12):2074-2082.

Fortunato ME, Colina CM. 2014. Effects of galactosylation in immunoglobulin G from all-atom molecular

dynamics simulations. J Phys. Chem. B 118 (33):9844-9851.

Frank M, Walker RC, Lanzilotta WN, Prestegard JH, Barb AW. 2014. Immunoglobulin G1 Fc domain motions:

implications for Fc engineering. J Mol. Biol 426 (8):1799-1811.

Gemmill TR, Trimble RB. 1999. Overview of N- and O-linked oligosaccharide structures found in various

yeast species. Biochim Biophys Acta 1426 (2):227-37.

Gerngross TU. 2004. Advances in the production of human therapeutic proteins in yeasts and filamentous

fungi. Nat Biotechnol 22 (11):1409-14.

Gramer MJ, Goochee CF. 1993. Glycosidase activities in Chinese hamster ovary cell lysate and cell culture

supernatant. Biotechnol Prog 9 (4):366-73.

25
Hamilton SR, Gerngross TU. 2007. Glycosylation engineering in yeast: the advent of fully humanized yeast.

Curr Opin Biotechnol 18 (5):387-92.

Helenius A, Aebi M. 2001. Intracellular functions of N-linked glycans. Science 291 (5512):2364-2369.

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Herter S, Birk MC, Klein C, Gerdes C, Umana p, Bacac M. 2014. Glycoengineering of therapeutic antibodies

enhances monocyte/macrophage-mediated phagocytosis and cytotoxicity. J Immunol 192 (5):2252-2260.

Houde D, Peng Y, Berkowitz SA, Engen JR. 2010. Post-translational modifications differentially affect IgG1

conformation and receptor binding. Mol. Cell Proteomics 9 (8):1716-1728.

Huang W, Giddens J, Fan SQ, Toonstra C, Wang LX. 2012. Chemoenzymatic glycoengineering of intact IgG

antibodies for gain of functions. J Am. Chem. Soc 134 (29):12308-12318.

Ichikawa Y, Yamada C, Horiki T, Hoshina Y, Uchiyama M, Yamada Y, Toumatu J. 1998. Anti-agalactosyl IgG

antibodies and isotype profiles of rheumatoid factors in Sjogren's syndrome and rheumatoid arthritis. Clin

Exp. Rheumatol 16 (6):709-715.

Idusogie EE, Presta LG, Gazzano-Santoro H, Totpal K, Wong PY, Ultsch M, Meng YG, MulkerrinMG. 2000.

Mapping of the C1q binding site on rituxan, a chimeric antibody with a human IgG1 Fc. J. Immunol 164

(8):4178-4184.

Imai-Nishiya H, Mori K, Inoue M, Wakitani M, Iida S, Shitara K, Satoh M. 2007. Double knockdown of

alpha1,6-fucosyltransferase (FUT8) and GDP-mannose 4,6-dehydratase (GMD) in antibody-producing cells:

a new strategy for generating fully non-fucosylated therapeutic antibodies with enhanced ADCC. BMC.

Biotechnol 7:84.

Jaglowski SM, Alinari L, Lapalombella R, Muthusamy N, Byrd JC. 2010. The clinical application of monoclonal

antibodies in chronic lymphocytic leukemia. Blood 116 (19):3705-3714.

Jefferis R. 2009. Glycosylation as a strategy to improve antibody-based therapeutics. Nat. Rev. Drug Discov

8 (3):226-234.

26
Jefferis R. 2012. Isotype and glycoform selection for antibody therapeutics. Arch. Biochem. Biophys 526

(2):159-166.

Jensen PF, Schoch A, Larraillet V, Hilger M, Schlothauer T, Emrich T, Rand KD. 2017. A Two-pronged Binding

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Mechanism of IgG to the Neonatal Fc Receptor Controls Complex Stability and IgG Serum Half-life. Mol Cell

Proteomics 16 (3):451-456.

Kaneko Y, Nimmerjahn F, Ravetch JV. 2006. Anti-inflammatory activity of immunoglobulin G resulting from

Fc sialylation. Science 313 (5787):670-673.

Karsten CM, Pandey MK, Figge J, Kilchenstein R, Taylor PR, Rosas M, McDonald JU, Orr SJ, Berger M, Petzold

D, Blanchard V, Winkler A, Hess C, Reid DM, Majoul IV, Strait RT, Harris NL, Kohl G, Wex E, Ludwig R,

Zillikens D, Nimmerjahn F, Finkelman FD, Brown GD, Ehlers M, Kohl J. 2012. Anti-inflammatory activity of

IgG1 mediated by Fc galactosylation and association of FcgammaRIIB and dectin-1. Nat. Med 18 (9):1401-

1406.

Kayser V, Chennamsetty N, Voynov V, Forrer K, Helk B, Trout BL. 2011. Glycosylation influences on the

aggregation propensity of therapeutic monoclonal antibodies. Biotechnol. J 6 (1):38-44.

Kim C, Prasad V. 2016. Strength of Validation for Surrogate End Points Used in the US Food and Drug

Administration's Approval of Oncology Drugs. Mayo Clin Proc. May 10. pii: S0025-6196(16)00125-7.

Krapp S, Mimura Y, Jefferis R, Huber R, Sondermann P. 2003. Structural analysis of human IgG-Fc

glycoforms reveals a correlation between glycosylation and structural integrity. J. Mol. Biol 325 (5):979-989.

Kunert R, Reinhart D. 2016. Advances in recombinant antibody manufacturing. Appl Microbiol Biotechnol

100 (8):3451-61.

Kurogochi M, Mori M, Osumi K, Tojino M, Sugawara S, Takashima S, Hirose Y, Tsukimura W, Mizuno M,

Amano J, Matsuda A, Tomita M, Takayanagi A, Shoda S, Shirai T. 2015. Glycoengineered Monoclonal

Antibodies with Homogeneous Glycan (M3, G0, G2, and A2) Using a Chemoenzymatic Approach Have

27
Different Affinities for FcgammaRIIIa and Variable Antibody-Dependent Cellular Cytotoxicity Activities.

PLoS. One 10 (7):e0132848.

Li T, DiLillo DJ, Bournazos S, Giddens JP, Ravetch JV, Wang LX. 2017. Modulating IgG effector function by Fc

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


glycan engineering. Proc. Natl. Acad. Sci. U. S. A 114 (13):3485-3490.

Li, W., Z. Zhu, W. Chen, Y. Feng, and D.S. Dimitrov. 2017. Crystallizable Fragment Glycoengineering for

Therapeutic Antibodies Development. Front Immunol 8:1554.

Lin CW, Tsai MH, Li ST, Tsai TI, Chu KC, Liu YC, Lai MY, Wu CY, Tseng YC, Shivatare SS, Wang CH, Chao P,

Wang SY, Shih HW, Zeng YF, You TH, Liao JY, Tu YC, Lin YS, Chuang HY, Chen CL, Tsai CS, Huang CC, Lin NH,

Ma C., Wu CY, Wong CH. 2015. A common glycan structure on immunoglobulin G for enhancement of

effector functions. Proc. Natl. Acad. Sci. U. S. A 112 (34):10611-10616.

Liu CP, Tsai TI, Cheng T, Shivatare VS, Wu CY, Wu CY, Wong CH. 2018. Glycoengineering of antibody

(Herceptin) through yeast expression and in vitro enzymatic glycosylation. Proc. Natl. Acad. Sci. U. S. A 115

(4):720-725

Liu H, Nowak C, Shao M, Ponniah G, Neill A. 2016. Impact of cell culture on recombinant monoclonal

antibody product heterogeneity. Biotechnol. Prog 32 (5):1103-1112.

Liu R, Giddens J, McClung CM, Magnelli PE, Wang LX, Guthrie EP. 2016. Evaluation of a glycoengineered

monoclonal antibody via LC-MS analysis in combination with multiple enzymatic digestion. MAbs 8 (2):340-

346.

Lu MC, Hsieh SC, Lai NS, Li KJ, Wu CH, Yu CL. 2007. Comparison of anti-agalactosyl IgG antibodies,

rheumatoid factors, and anti-cyclic citrullinated peptide antibodies in the differential diagnosis of

rheumatoid arthritis and its mimics. Clin Exp. Rheumatol 25 (5):716-721.

Macher B A, Galili U. 2008. The Galalpha1,3Galbeta1,4GlcNAc-R (alpha-Gal) epitope: a carbohydrate of

unique evolution and clinical relevance. Biochim Biophys Acta 1780 (2):75-88.

28
Mastrangeli R, Rossi M, Mascia M, Palinsky W, Datola A, Terlizzese M, Bierau H. 2014. In vitro Biological

Characterization of IFN-beta-1a major Glycoforms. Glycobiology. (1):21-9.

Masuda K, Yamaguchi Y, Kato K, Takahashi N, Shimada I, Arata Y. 2000. Pairing of oligosaccharides in the Fc

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


region of immunoglobulin G. FEBS Lett 473 (3):349-357.

Matucci A, Cammelli D, Cantini F, Goletti D, Marino V, Milano GM, Scarpa R, Tocci G, Maggi E, Vultaggio A.

2016. Influence of anti-TNF immunogenicity on safety in rheumatic disease: a narrative review. Expert.

Opin. Drug Saf 15 (sup1):3-10.

Meier S, Duus J. 2011. Carbohydrate dynamics: Antibody glycans wiggle and jiggle. Nat. Chem. Biol 7

(3):131-132.

Mimura Y, Ashton PR, Takahashi N, Harvey DJ, Jefferis R. 2007. Contrasting glycosylation profiles between

Fab and Fc of a human IgG protein studied by electrospray ionization mass spectrometry. J Immunol.

Methods 326 (1-2):116-126.

Mimura Y, Katoh T, Saldova R, O'Flaherty R, Izumi T, Mimura-Kimura Y, Utsunomiya T, Mizukami Y,

Yamamoto K, Matsumoto T, Rudd PM. 2018. Glycosylation engineering of therapeutic IgG antibodies:

challenges for the safety, functionality and efficacy. Protein Cell 9 (1):47-62.

Montjovent M O, Zou L, Faust H, Abdolzade-Bavil A, Liu PM, Struwe P. 2017. Method establishment for

discerned immunogenicity assessment of a recombinant glycoprotein containing nonhuman sialic acid

Neu5Gc residues. Bioanalysis 9 (18):1385-1393.

Nimmerjahn F, Ravetch JV. 2010. Antibody-mediated modulation of immune responses. Immunol Rev

236:265-75.

Nishijima C, Sato S, Takehara K. 2001. Anti-agalactosyl IgG antibodies in sera from patients with systemic

sclerosis. J Rheumatol 28 (8):1847-1851.

Pawlowski JW, Bajardi-Taccioli A, Houde D, Feschenko M, Carlage T, Kaltashov IA. 2018. Influence of glycan

modification on IgG1 biochemical and biophysical properties. J Pharm. Biomed. Anal 151:133-144
29
Pincetic A, Bournazos S, DiLillo DJ, Maamary J, Wang TT, Dahan R, Fiebiger BM, Ravetch JV. 2014. Type I and

type II Fc receptors regulate innate and adaptive immunity. Nat. Immunol 15 (8):707-716.

Platts-Mills TAE, Schuyler Aj, Hoyt AEW, Scott P, Commins P. 2015. Delayed anaphylaxis involving IgE to

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


galactose-alpha-1,3-galactose Curr Allergy Asthma Rep 15(4):12.

Quast I, Peschke B, Lunemann JD. 2017. Regulation of antibody effector functions through IgG Fc N-

glycosylation. Cell Mol. Life Sci 74 (5):837-847.

Raju TS. 2008. Terminal sugars of Fc glycans influence antibody effector functions of IgGs. Curr. Opin.

Immunol 20 (4):471-478.

Raju TS, Jordan RE. 2012. Galactosylation variations in marketed therapeutic antibodies. MAbs 4 (3):385-

391.

Reusch D, Tejada ML. 2015. Fc glycans of therapeutic antibodies as critical quality attributes. Glycobiology

25 (12):1325-1334.

Rillahan CD, Antonopoulos A, Lefort CT, Sonon R, Azadi P, Ley K, Dell A, Haslam SM, Paulson JC. 2012.

Global metabolic inhibitors of sialyl- and fucosyltransferases remodel the glycome. Nat. Chem. Biol (8):661–

668.

Ruhaak LR, Uh HW, Deelder AM, Dolhain RE, Wuhrer M. 2014. Total plasma N-glycome changes during

pregnancy. J Proteome. Res 13 (3):1657-1668.

Scanlan CN, Burton DR, Dwek RA. 2008. Making autoantibodies safe. Proc. Natl. Acad. Sci. U. S. A 105

(11):4081-4082.

Schneider S, Zacharias M. 2012. Atomic resolution model of the antibody Fc interaction with the

complement C1q component. Mol. Immunol 51 (1):66-72.

Shepard HM, Phillips GL, Thanos D, Feldmann M. 2017. Developments in therapy with monoclonal

antibodies and related proteins. Clin Med. (Lond) 17 (3):220-232.

30
Shi HH, Goudar CT. 2014. Recent advances in the understanding of biological implications and modulation

methodologies of monoclonal antibody N-linked high mannose glycans. Biotechnol. Bioeng 111 (10):1907-

1919.

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Shinkawa T, Nakamura K, Yamane N, Shoji-Hosaka E, Kanda Y, Sakurada M, Uchida K, Anazawa H, Satoh M,

Yamasaki M, Hanai N, Shitara K. 2003. The absence of fucose but not the presence of galactose or bisecting

N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing

antibody-dependent cellular cytotoxicity. J Biol. Chem 278 (5):3466-3473.

Sondermann P, Pincetic A, Maamary J, Lammens K, Ravetch JV. 2013. General mechanism for modulating

immunoglobulin effector function. Proc. Natl. Acad. Sci. U. S. A 110 (24):9868-9872.

Spiegelberg HL, Abel CA, Fishkin BG, Grey HM. 1970. Localization of the carbohydrate within the variable

region of light and heavy chains of human gamma g myeloma proteins. Biochemistry 9 (21):4217-4223.

Tang F, Wang LX, Huang W. 2017. Chemoenzymatic synthesis of glycoengineered IgG antibodies and

glycosite-specific antibody-drug conjugates. Nat. Protoc 12 (8):1702-1721.

Tang F, Yang Y, Tang Y, Tang S, Yang L, Sun B, Jiang B, Dong J, Liu H, Huang M, Geng MY, Huang W. 2016.

One-pot N-glycosylation remodeling of IgG with non-natural sialylglycopeptides enables glycosite-specific

and dual-payload antibody-drug conjugates. Org. Biomol. Chem 14 (40):9501-9518.

Tagyi VS, Butler M. 2018. Solid-Phase Enzymatic Remodeling Produces High Yields of Single Glycoform

Antibodies. Biotechnol. J 13(4): e1700381

Teranishi K, Manez R, Awwad M, Cooper DK. 2002. Anti-Gal alpha 1-3Gal IgM and IgG antibody levels in

sera of humans and old world non-human primates. Xenotransplantation 9 (2):148-54.

Thomann M, Schlothauer T, Dashivets T, Malik S, Avenal C, Bulau P, Ruger P, Reusch D. 2015. In vitro

glycoengineering of IgG1 and its effect on Fc receptor binding and ADCC activity. PLoS. One 10

(8):e0134949.

31
Tsai TI, Li ST, Liu CP, Chen KY, Shivatare SS, Lin CW, Liao SF, Lin CW, Hsu TL, Wu YT, Tsai MH, Lai MY, Lin NH,

Wu CY, Wong CH. 2017. An Effective Bacterial Fucosidase for Glycoprotein Remodeling. ACS Chem. Biol 12

(1):63-72.

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Vidarsson G, Dekkers G, Rispens T. 2014. IgG subclasses and allotypes: from structure to effector functions.

Front Immunol 5:520.

Weiskopf K, Weissman IL. 2015. Macrophages are critical effectors of antibody therapies for cancer. MAbs 7

(2):303-310.

Yang Z, Wang S, Halim A, Schulz MA, Frodin M, Rahman SH, Vester-Christensen MB, Behrens C, Kristensen

C, Vakhrushev SY, Bennett EP, Wandall HH, Clausen H. 2015. Engineered CHO cells for production of

diverse, homogeneous glycoproteins. Nat. Biotechnol 33 (8):842-844.

Zheng K, Yarmarkovich M, Bantog C, Bayer R, Patapoff TW. 2014. Influence of glycosylation pattern on the

molecular properties of monoclonal antibodies. MAbs 6 (3):649-658.

Zhu A, Hurst R. 2002. Anti-N-glycolylneuraminic acid antibodies identified in healthy human serum.

Xenotransplantation 9 (6):376-81.

Zou G, Ochiai H, Huang W, Yang Q, Li C, Wang LX. 2011. Chemoenzymatic synthesis and Fcgamma receptor

binding of homogeneous glycoforms of antibody Fc domain. Presence of a bisecting sugar moiety enhances

the affinity of Fc to FcgammaIIIa receptor. J Am. Chem. Soc 133 (46):18975-18991.

32
Legends to figures

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Figure 1. Human IgG1 3D structure

The heavy chains are shown in blue and light blue, the light chains are shown in red and light red,

respectively. The 2 N-glycans, present at each of the two heavy chains, are illustrated in a solid sphere

representation with different colors (grey and yellow). The illustration was generated by rendering the PDB

entry 1hzh by using the PyMOL Molecular Graphic System version 1.8.6.0. Schrödinger, LLC. Each light chain

consists of an N-terminal variable domain (VL) and a constant domain (CL). Two heavy chain–light chain

heterodimers combine into a single antibody molecule (H2L2). The light chain is associated to the VH and

the CH1 domains to form the fragment antigen binding (Fab) region. The C-terminal part of the antibody,

formed by (CH2-CH3)2 domains, is called fragment crystallisable (Fc) region. The variable region of the Fab

is responsible for the capability of immunoglobulins of binding to an antigen, while the Fc part, which

contains the binding sites for the complement component 1q (C1q) and the Fc--receptors (FcRs), is

responsible for the antibody effector functions such as the antibody-dependent cell-mediated cytotoxicity

(ADCC), the antibody-dependent cell-mediated phagocytosis (ADCP), and the complement-dependent

cytotoxicity (CDC). In addition, the Fc region contains two Fc neonatal receptor (FcRn) binding sites which

are responsible for the long antibody half-life (Quast et al. 2017).

Figure 2. Biantennary complex glycan structure found in the Fc region of human endogenous IgG.

Structural implications and differential interactions and mobility of α1,6 and α1,3 arms of the Fc glycan.

Figure 3. IgG1 Fc-glycan microheterogeneities

A) human IgG1, B) CHO, and C) Mouse cells (NS0/SP2). The double-dash (- -) symbol between residues

indicates a potential truncation contributing to the overall high Fc-glycan microheterogeneity. Complex
33
type glycans containing 0, 1 or 2 galactose residues are referred as G0F, G1F and G2F glycoform when core-

fucosylated and G0, G1 and G2 when core-fucose is absent. Non-human glycans are highly expressed by

mouse cells and at low levels, by CHO cells.

Downloaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 October 2018


Figure 4. Relative FcγRIIIa binding increase of non-fucosylated homogeneous Fc-glycans

The figure illustrates the fold increase, in terms of FcγRIIIa binding, observed for non-fucosylated,

homogeneous Fc glycan variants compared with the heterogenous wildtype population as a reference. The

homogeneous glycan populations, using rituximab and trastuzumab as model proteins, were obtained by

chemo-enzymatic glycoengineering (Lin et al. 2015).

34
cob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673
ob/advance-article-abstract/doi/10.1093/glycob/cwy092/511567
oaded from https://academic.oup.com/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bukkyo University user on 06 Octobe
om/glycob/advance-article-abstract/doi/10.1093/glycob/cwy092/5115673 by Bu

You might also like