14,719
Views
13
CrossRef citations to date
0
Altmetric
Review

The “less-is-more” in therapeutic antibodies: Afucosylated anti-cancer antibodies with enhanced antibody-dependent cellular cytotoxicity

, , &
Pages 693-711 | Received 24 Jan 2018, Accepted 16 Apr 2018, Published online: 07 May 2018

ABSTRACT

Therapeutic monoclonal antibodies are the fastest growing class of biological therapeutics for the treatment of various cancers and inflammatory disorders. In cancer immunotherapy, some IgG1 antibodies rely on the Fc-mediated immune effector function, antibody-dependent cellular cytotoxicity (ADCC), as the major mode of action to deplete tumor cells. It is well-known that this effector function is modulated by the N-linked glycosylation in the Fc region of the antibody. In particular, absence of core fucose on the Fc N-glycan has been shown to increase IgG1 Fc binding affinity to the FcγRIIIa present on immune effector cells such as natural killer cells and lead to enhanced ADCC activity. As such, various strategies have focused on producing afucosylated antibodies to improve therapeutic efficacy. This review discusses the relevance of antibody core fucosylation to ADCC, different strategies to produce afucosylated antibodies, and an update of afucosylated antibody drugs currently undergoing clinical trials as well as those that have been approved.

Introduction

Therapeutic antibodies represent the fastest growing group of biotherapeutics in recent years, both in the numbers of antibodies entering clinical trials and in global sales revenue.Citation1-Citation4 Many monoclonal antibodies are used for treatment of various malignancies and autoimmune disorders. Anti-cancer antibodies target cancer cells by triggering effector functions such as antibody-dependent cellular cytotoxicity (ADCC) upon engagement of immune complexes with FcγRIIIa present on natural killer (NK) cells, or direct induction of tumor cell apoptosis through blocking the binding of pro-survival ligands or inhibiting signal receptor dimerization. NK cells are a type of lymphocyte, representing about 10% of total lymphocytes. Unlike B and T lymphocytes, which are the important components of the adaptive immune system, NK cells are a critical component of the innate immune system. The Fc region of monoclonal antibodies acts as an important bridge between adaptive and innate immune response. When the antigens expressed on the surfaces of cancer cells, virus-infected cells or invading pathogens are recognized by specific antibodies, the cells or pathogens become coated with the antibodies. The Fc region of the antibodies bound to these surfaces assists in the elimination of the targets via different mechanisms. Firstly, it can interact with the C1 molecule of the complement system and trigger the activation of classical pathway of the complement system. It can also recruit phagocytes via Fc receptors and activate the phagocytosis pathway and, as mentioned above, activate ADCC mediated by NK cells. Among these mechanisms, studies on rituximab and trastuzumab have suggested that ADCC is the key mechanism of action to eliminate cancer cells.Citation5-Citation7

The FcγRIII binds the Fc region of IgG1 antibodies by interacting with the hinge region and the CH2 domain.Citation 8 , Citation 9 This Fc-FcγRIII interaction is significantly affected by the glycan present at the conserved N-glycosylation site Asn297 (N297) in each of the CH2 domains.Citation 10 Mutations in the CH2 domain that destroyed the conserved N-glycosylation motif and hence gave rise to aglycosylated Fc resulted in complete loss of binding to most FcγRs except FcγRI.Citation 11 Several approaches have been utilized to increase the affinity between antibody and the FcγRIII. These include engineering the Fc region through amino acid mutationsCitation 12 and glycoengineering the Fc N-glycan to reduce core fucose.Citation13-Citation15 It is now widely recognized that removal of the core fucose from Fc N-glycans represents the most effective approach to enhance ADCC activity.Citation 14 , Citation 15 A high-throughput study of the IgG glycome of three isolated human populations showed that most of the human plasma IgG antibodies are core fucosylated with levels of afucosylated IgG ranging from 1.3% to 19.3%, underlying the difference in ADCC efficacy of naturally occurring antibodies to protect against diseases.Citation 16 Dramatic shifts in IgG glycan profile towards reduced galactosylation and fucosylation have been observed in human immunodeficiency virus (HIV)-specific antibodies and are associated with improved antiviral activity and HIV control.Citation 17

There are two FcγRIII genes in the human genome, one encodes FcγRIIIa and the other encodes FcγRIIIb. These two proteins share 97% homology at the amino acid level. While the transmembrane protein FcγRIIIa is expressed in most effector cells of the immune system, FcγRIIIb is exclusively expressed by neutrophils as a glycosylphosphatidylinositol (GPI)-anchored protein. FcγRIIIb is not known to play a role in ADCC, but it may play a role in phagocytosis of IgG-coated pathogens. Two common alleles of the FcγRIIIa gene encode two variants that differ at position 158, either a Val (V158) or a Phe (F158).Citation 18 , Citation 19 Between the two variants, FcγRIIIa-V158 has a higher affinity to human IgG1. For example, under similar experimental conditions, FcγRIIIa-V158 demonstrated an approximately 10-fold higher affinity for IgG than FcγRIIIa-F158.Citation 20 Cells expressing the FcγRIIIa-V158 allele mediate ADCC more effectively.Citation 19 In anti-epidermal growth factor receptor (EGFR) antibody-treated colorectal cancer patients, the clinical outcome was strongly associated with the FcγRIIIa polymorphisms. Better clinical outcomes have been observed in patients expressing high affinity FcγRIIIa variant (V158) when they were treated with anti-CD20 or anti-EGFR antibodies.Citation 5 , Citation21-Citation23

Protein fucosylation in mammalian system

Fucose (6-deoxy-L-galactose) is a common component of many N- and O-linked glycans produced in mammalian cells. A total of 13 fucosyltransferases (FUT) that have been identified in the human genome transfer a fucose residue from GDP-fucose to an acceptor substrate.Citation 24 FUT1 and FUT2 transfer the fucose residue to the terminal galactose and form an α1,2 linkage. FUT3 has both α1,3- and α1,4-fucosyltransferase activities responsible for the synthesis of Lewisx- and Lewisa-related structures. FUT4 to FUT7 and FUT9 to FUT11 are all α1,3-fucosyltransferases. These transferases are responsible for the synthesis of the ABH and the Lewis antigens.Citation 25 , Citation 26 Lewis-related tri- or tetra-saccharides play critical roles in leukocyte adhesion during inflammatory response and lymphocyte homing.Citation 27 Based on the glycosidic linkages, the Lewis antigens can be divided into two types. Type I includes Lewisa (Lea), sialyl-Lewisa (SLea) and Lewisb (Leb). Type II includes Lewisx (Lex), sialyl-Lewisx (SLex) and Lewisy (Ley). Some of these Lewis antigens are found overexpressed on different types of cancer cells.Citation 28 , Citation 29 SLea or CA 19-9 (cancer antigen 19-9) is one of the commonly used tumor markers in clinics.Citation 28 , Citation 30 Lewis antigens may contribute to adhesion of cancer cells to vascular endothelium and promote hematogenous metastasis of cancer cells.Citation 31 , Citation 32 In the 1980s and early 1990s, many monoclonal antibodies were generated by whole-cell immunization of mice with different types of cancer cells. Many of these “anti-cancer” antibodies turned out to be specific for different Lewis antigens.Citation33-Citation36 Unfortunately, the development of these antibodies into anti-cancer therapeutics has been quite challenging, because many Lewis antigens are also expressed in several types of normal tissues, particularly in the mucosa of human gastrointestinal tract in the form of O-linked glycans attached to the mucins.Citation37-Citation48 For example, anti-Ley antibodies showed strong side effects including nausea and vomiting in Phase 1 clinical studies because the expression of Ley in the gastrointestinal tract.Citation 33

FUT8 is the only α1,6-fucosyltransferase that transfers fucose via an α1,6 linkage to the innermost N-acetylglucosamine on N-glycans for core fucosylation.Citation 49 FUT8 is widely expressed in various tissues except in the liver, but it is significantly upregulated in hepatocellular carcinoma (HCC) tissues. Alpha-fetoprotein (AFP) is the most abundant plasma protein found in the human fetus. The level of AFP begins to decrease after birth and reaches very low levels in adults. Serum AFP level is elevated in people with HCC, and it has therefore been a reliable biomarker for HCC. However, the serum level of AFP also increases slightly in some patients with chronic liver diseases, which makes it difficult to diagnose HCC at its early stage when serum AFP level is still low. Since FUT8 is overexpressed in HCC patients and therefore the AFP in HCC patients is core-fucosylated, but the AFP is not core-fucosylated in patients with chronic liver diseases. Therefore, elevated levels of core-fucosylated AFP have been used as a more accurate tumor biomarker.Citation 50 , Citation 51 The other two fucosyltransferases are POFUT1 and POFUT2. They are O-fucosyltransferases that mediate the direct attachment of fucose to Ser or Thr residues of proteins in the ER.Citation 52 , Citation 53 O-fucosylation of Notch protein is essential for Notch signaling which plays an important role in the regulation of embryonic development.Citation 54

The substrate for fucosylation reactions, GDP-β-L-fucose (GDP-fucose), is synthesized in the cytoplasm through the de novo and the salvage pathway. The de novo pathway, which generates the majority of GDP-fucose, involves the conversion of GDP-mannose to GDP-fucose by GDP-mannose 4,6 dehydratase (GMD) and GDP-keto-6-deoxymannose 3,5-epimerase/4 reductase (also known as FX).Citation 55 The salvage pathway, which accounts for only a small percentage of GDP-fucose production, utilizes free cytosolic fucose derived from degraded glycoproteins or glycolipids or exogenous fucose.Citation 24 The GDP-fucose synthesized in the cytosol must be transported into the Golgi apparatus or the endoplasmic reticulum (ER) by specific transporters in order to serve as the substrate for fucosylation reactions. The Golgi GDP-fucose transporter (GFT), encoded by the Slc35c1 gene, is a member of the solute carrier family 35 (SLC35).Citation 56 GFT is responsible for transporting GDP-fucose from the cytosol into the Golgi. Mutations in the Slc35c1 gene in humans lead to the development of leukocyte adhesion deficiency type II (LADII) or congenital disorder of glycosylation type IIc, characterized by severe immunodeficiency, mental retardation and slow growth.Citation57-Citation60

The effect of IgG core fucosylation on ADCC

The classic ADCC response is mediated by NK cells following the binding of the FcγRIIIa to the Fc region of antibody molecules. This binding triggers the NK cells to release cytokines and cytolytic agents that eventually kill the target cell. The ADCC activity is highly affected by the Fc N-glycan. In recombinant IgG therapeutics produced in Chinese hamster ovary (CHO) cells, the Fc N-glycans are heterogeneous biantennary complex type with a fucose residue attached to the core position. These N-glycans contain little to no sialic acid with zero (G0), one (G1) or two (G2) galactose residues. In the study by Shields et al., humanized IgG1 antibodies expressed in CHO Lec13 cells demonstrated a 50-fold improvement in binding affinity to human FcγRIIIa compared to the same antibodies produced in wild type CHO cells.Citation 14 Antibodies produced in Lec13 cells carry a significant amount of afucosylated N-glycans due to the mutated GMD gene in these cells.Citation 61 Importantly, the afucosylated IgG1 demonstrated significant improvement in ADCC in vitro using peripheral blood mononuclear cells (PBMCs) or NK cells in comparison to its fucosylated counterpart. Shinkawa et al. subsequently reported that the absence of fucose, but not the presence of galactose or bisecting GlcNAc, is critical for enhancing ADCC.Citation 15 Another study also suggested that the removal of core fucose from antibodies was sufficient to achieve maximal ADCC activity.Citation 62 It was shown that there was no significant difference in ADCC activity mediated by core fucose removal or amino acid mutations S229D/D298A/I332E, which was known to have higher binding affinity for FcγRIIIa.Citation 12 In addition, no additive effect was observed on B-cell depletion activity of anti-CD20 IgG1 in human blood using a combination of these techniques.Citation 62 Through the use of isothermal titration calorimetry, it was demonstrated that the IgG1-FcγRIIIa binding is driven by favorable binding enthalpy (ΔH), but opposed by unfavorable binding entropy change (ΔS).Citation 63 Fucose removal enhanced the favorable ΔH leading to an increase in the binding constant of IgG1 for the receptor by a factor of 20–30 fold, suggestive of an increase in non-covalent interactions upon complexation.Citation 63

Molecular mechanisms to account for the enhanced affinity of afucosylated antibodies to FcγRIIIa

The first crystal structure of FcγRIII-IgG1-Fc complex was reported in 2000.Citation 9 The FcγRIII used in the study was a soluble FcγRIIIb (sFcγRIIIb) produced in E. coli and the Fc was isolated from pooled human IgG1. The crystal structure revealed that the receptor is bound between the two CH2 domains and the hinge region asymmetrically through van der Waals contacts and hydrogen bonds. Only one N-glycan of the two CH2 domains makes contact with the receptor. The innermost GlcNAc residue of the Fc N-glycan was found to have the potential of forming hydrogen bonds with several amino acids of the FcγRIII. As the sFcγRIII preparation used in the study was unglycosylated, it was impossible to evaluate the impact of its N-linked glycan on the FcγRIII-Fc interaction. Nonetheless, the authors did highlight that Asn162 is a potential glycosylation site of FcγRIII that is close to a binding site and a larger carbohydrate moiety attached to this site may influence the affinity to IgG.Citation 9 Indeed, a subsequent study revealed that, compared to the unglycosylated form of FcγRIII (by mutating Asn162 to Gln162), the glycosylated FcγRIII (Asn162) showed reduced affinity for native (fucosylated) IgG antibodies, while antibodies with or without the core fucose showed a similar affinity for unglycosylated FcγRIII.Citation 20 However, when fucose-free antibody binds glycosylated FcγRIII (Asn162), the affinity increased significantly. The binding affinities of different glycoforms of IgG-FcγRIII pairs are in the following order: IgG-fucose-free/FcγRIIIa-Asn162 >> IgG-native glycan/FcγRIIIa-Gln162 > IgG-native glycan/FcγRIIIa-Asn162.Citation 20 The authors concluded that the carbohydrate moieties of both FcγRIIIa and IgG are important for the interaction. An N-glycan needs to be attached to FcγRIIIa Asn162 and enhanced binding affinity can be achieved if the antibody is afucosylated.Citation 20 In their proposed model, the fucose residue protrudes from the continuous surface of the Fc into open space, which prohibits close contact of the Fc receptor N-glycan core, thereby precluding additional productive interactions. Furthermore, the model predicts that only one of the two Fc-fucose residues needs to be absent for increased binding affinity toward FcγRIIIa.

Detailed X-ray crystallography studies on the Fc-FcγRIIIa complex confirmed this model. Ferrara et al. showed that a unique kind of carbohydrate–carbohydrate interaction coupled with increased number of newly formed hydrogen bonds and van der Waals contacts likely contribute to the increased binding affinity observed between afucosylated Fc and the Asn162-glycosylated receptor.Citation 64 However, in the crystal structure of fucosylated Fc in complex with FcγRIIIa, the core fucose is oriented toward the second GlcNAc of the N-glycan attached to Asn162 and has to accommodate in the interface between the interacting glycan chains.Citation 64 As a result, the whole oligosaccharide unit on Asn162 moves away from the Fc glycan, which leads to a weakened FcγRIIIa-IgG interaction.

Ferrara et al. demonstrated that the glycosylation at Asn162 of FcγRIII is not essential for the expression of the receptor; however, this glycosylation site is conserved among all FcγRIIIs (or the equivalent) in all mammals studied.Citation 19 Furthermore, in all FcγRs, the regions that interact with the antibody are highly conserved, yet all other receptors lack this glycosylation site.Citation 9 It is tempting to speculate that ADCC may be modulated by IgG core fucosylation because of the presence of the glycan at Asn162 of FcγRIII. Indeed, reduced core fucosylation of antibodies has been linked to enhanced immune response during an autoimmune disease and an infectious disease.Citation 65 , Citation 66

Fc galactosylation and sialylation also modulate IgG1 interaction with FcγRIIIa, but to a significantly lesser extent

Recent studies have indicated that Fc galactosylation leads to increased FcγRIIIa binding, although to a significantly lesser extent compared to the removal of core fucose.Citation 11 , Citation67-Citation69 By carrying out enzymatic hyper-galactosylation across four batches of monoclonal antibodies produced from standard manufacturing processes in CHO cells, Thomann et al. demonstrated that hyper-galactosylation of antibody samples consistently leads to improvement in FcγRIIIa binding and ADCC.Citation 68 However, addition of galactose to afucosylated antibodies did not confer additional improvements to ADCC efficacy, indicating that afucosylation remains the major determinant of ADCC activity. While afucosylation removes the steric hindrance for enhanced Fc-FcγRIIIa interaction, a more ‘bulky’ G2F N-glycan structure may help to keep the two CH2 domains of IgG Fc in a more open horseshoe conformation for FcγRIIIa to bind.Citation 10 These observations are particularly important as recombinant therapeutic antibodies produced in CHO cells exhibit heterogeneity in terms of galactosylation, with G0F as the most abundant and G2F as the least abundant N-glycan. Improving the percentage of G2F can be achieved by over-expressing appropriate galactosyltransferases in CHO cells. In recombinant antibodies produced in CHO cells, only a small portion of the N-glycans is sialylated. On the contrary, a recent report showed that increased sialylation of the Fc N-glycan decreased ADCC if core fucose is present. However, in the absence of fucosylation, sialylation did not make any difference.Citation 70 Therefore, core fucosylation plays a much more significant role in modulating ADCC than galactosylation or sialylation.

Modulating FcγRIIIa interaction through Fc engineering

In addition to glycoengineering of the Fc N-glycan, various strategies have been performed to engineer the Fc domain to improve the ADCC effector function. Through alanine scanning mutagenesis of individual solvent-exposed residues on the human IgG1 Fc domain, residues involved in the binding site for human FcR were mapped.Citation 12 IgG1 mutants with improved binding to FcγRIIIa – T256A, K290A, S298A, E333A, and K334A were identified. These Fc variants demonstrated up to 1-fold enhanced ADCC in vitro.Citation 12

With the use of computational structure-based design and high-throughput screening, a series of engineered Fc variants were generated.Citation 71 These Fc variants of either single (S239D or I332E), double (S239D/I332E) or triple (S239D/I332E/A330L) mutations demonstrated up to 169-fold enhanced interaction with human FcγRIIIa.Citation 71 The Fc variants also showed enhanced binding ratio between activating FcRγIIIa and inhibitory FcγRIIb of up to 9-fold. The double mutant (S239D/I332E) has been employed in the design of a humanized anti-CD19 antibody, XmAb5574, by Xencor. XmAb5574 was able to enhance ADCC activity against a wide range of B-lymphoma and leukemia cell lines and also that of patient-derived acute lymphoblastic leukemia and mantle cell lymphoma cells.Citation 72 In vivo, it showed enhanced anti-tumor effect in mouse lymphoma xenograft over the wild type analogue.Citation 72 XmAb5574 is currently in clinical trials against various forms of B cell lymphoma.

Functional genetic screen, through the use of yeast surface display, to identify Fc sites with enhanced binding to low affinity activating FcγRIIIa and reduced binding to the inhibitory FcγRIIb was performed.Citation 73 An Fc variant 18 with several mutations (F243L/R292P/Y300L/V305I/P396L) was identified and demonstrated about 100-fold enhanced ADCC activity.Citation 73 MGAH22, from Macrogenics, is a chimeric IgG1 anti-HER2 antibody, with similar affinity and specificity to trastuzumab, containing the engineered Fc domain (variant 18) except that V305I was replaced with L235V to reduce FcγRIIb binding.Citation 74 MGAH22 showed enhanced affinity to both FcγRIIIa variants (F158 and V158), but decreased affinity to inhibitory FcγRIIb.Citation 74 This translated into enhanced ADCC activity over the wild-type equivalent of MGAH22 antibody. In vivo, MGAH22 demonstrated enhanced anti-HER2 activity over HER2 positive tumor in transgenic mouse expressing the low affinity human FcγRIIIa F158 variant.Citation 74 MGH22 is currently being evaluated in clinical studies of patients with HER2-positive cancers.

Strategies to produce afucosylated antibodies

Biosynthetic enzymes of GDP-fucose

CHO Lec13 cells are naturally defective in GDP-fucose formation due to a deficiency in endogenous GDP-mannose 4,6-dehydratase (GMD).Citation 61 The enzyme is responsible for catalysing the first of three steps in the de novo GDP-fucose biosynthesis pathway. This has resulted in the application of Lec13 cells as the host cell line for the production of afucosylated antibodies.Citation 14 However studies have shown that single clones isolated from Lec13 cells display a wide variety of fucosylation range, with most clones producing 50–70% fucosylated antibody when cultured to confluence in a static flask.Citation 75 Further analysis revealed low-level expression of GMD at mRNA level as well as the presence of fucosylated oligosaccharides on cell surface using LCA-staining. Shields et al. also noted that the Lec13 cell line is not sufficiently robust to be utilized as a production cell line as expression levels of antibodies tested (anti-HER2Hu4D5 and anti-IgE HuE27) were lower than that produced in other CHO cells.Citation 14 A GDP-keto-6-deoxymannose 3,5-epimerase/4 reductase (FX)-knockout CHO cell line that can be used to produce antibodies with completely afucosylated N-glycans was recently reported.Citation 76

Fucosyltransferase – FUT8

Shinkawa et al. employed rat hybridoma YB2/0 cells to produce humanized anti-human interleukin-5 receptor (IL-5R) IgG1 antibody (KM8399) and compared it against the same antibody produced in CHO cells (KM8404).Citation 15 Although both antibodies showed similar levels of antigen binding, the ADCC activity of YB2/0-produced KM8399 was 50-fold higher than CHO-produced KM8404. Similar results were obtained when two other antibodies were produced in CHO cells and YB2/0 cells. Glycan analysis showed that lower level of core fucose in YB2/0 cells-produced antibodies was the main reason for the enhanced ADCC.Citation 15 Analysis showed that YB2/0 cells have significantly lower levels of the Fut8 mRNA than CHO cells.

Another strategy to produce afucosylated antibodies involves inactivating the FUT8 gene. In the study by Yamane-Ohnuki et al., the FUT8 gene in an anti-CD20 antibody-producing CHO DG44 cell line was targeted for disruption using sequential homologous recombination.Citation 77 In the resultant cell line, both FUT8 alleles were knocked out from the FUT8 genomic region. The FUT8−/− cell line was shown to express completely afucosylated antibodies with a two-fold increase in ADCC compared to the same antibody produced in the parental cell line. The FUT8−/− cell line also demonstrated similar growth kinetics and productivity compared to the parental cell line when cultured in 1 L bioreactors. The FUT8 gene has also been targeted for inactivation using the zinc finger nuclease platform.Citation 78 This also led to the production of completely afucosylated antibodies. Small interfering RNA (siRNA) was also used to target FUT8 in an antibody-producing CHO DG44 cell line, and stable clones that produced 60% afucosylated antibodies were isolated.Citation 79

GDP-fucose transporter (SLC35C1)

It has been shown that loss-of-function mutations in the Golgi GDP-fucose transporter (GFT) gene (Slc35c1) was able to eliminate fucosylation reactions that occur in the Golgi.Citation 59 , Citation 60 Our group inactivated the Slc35c1 gene in CHO cells first by zinc-finger nucleases (ZFNs), followed by transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeats-Cas9 (CRISPR-Cas9) techniques.Citation 80 , Citation 81 The mutant cells in the transfected pools were identified and isolated by fluorescence-activated cell sorting (FACS) using fluorescently labelled fucose-specific Aleuria aurantia lectin (AAL).Citation 80 CHO cells with inactivated Slc35c1 gene have been named as CHO-gmt3 (CHO-glycosylation mutant3) cells. Mass spectrometry analyses demonstrated the complete lack of core fucose on N-glycans attached to the EPO-Fc fusion protein and IgG1 antibodies produced in the CHO-gmt3 cells.Citation 80 The CHO-K1 transcriptome data have shown that among all Golgi fucosyltransferases, only FUT8 is expressed.Citation 82 Therefore, inactivating Fut8 or Slc35c1 should have similar effects on CHO-K1 cells. A potential advantage of knocking out Slc35c1 over Fut8 is that it eliminates the potential complications caused by the gain-of-function mutations of fucosyltransferase found in LEC11 and LEC12 cells.Citation 83 Using this approach, we have been able to establish stable Slc35c1 / lines from several pre-existing antibody-producing CHO cell lines in less than two months. Our data showed that inactivation of the Slc35c1 gene in the pre-existing antibody-producing CHO cell line does not alter cell growth rate, viable cell density and antibody productivity in serum-free suspension culture conditions.Citation 80 This strategy has been used to produce afucosylated antibodies in a few recent studies.Citation 84 , Citation 85

Generation of bisecting GlcNac

β-1,4-mannosyl-glycoprotein 4-β-N-acetylglucosaminyltransferase (GnT-III) is normally not expressed in CHO cells. GnT-III catalyzes the formation of a bisecting GlcNAc by attaching a GlcNAc in β1,4 linkage to the β-linked mannose of the trimannosyl core of N-glycans. It was shown that overexpression of GnT-III in CHO cells was able to reduce Fc core fucosylation. Ferrara et al. evaluated the overexpression of a series of Golgi resident enzymes in combination with GnT-III and showed that overexpression of GnT-III and Golgi α-mannosidase II (αManII) resulted in the highest level of bisecting and afucosylated glycans on IgG antibodies.Citation 86 CHO cells that overexpress both GnT-III and αManII have been successfully used as the host cell line to produce anti-CD20 antibody GA101.

Expression of bacterial RMD in the cytosol of CHO cells to disrupt the GDP-fucose de novo pathway

In the de novo pathway of GDP-fucose biosynthesis in mammalian systems, GDP-mannose is first converted to GDP-4-keto-6-deoxy mannose (GKDM) by GDP-mannose-4,6-dehydratase. GKDM is eventually converted to GDP-fucose by several downstream enzymatic reactions. In bacteria, however, GKDM can be reduced to form GDP-rhamnose by a GDP-4-keto-6-deoxy mannose reductase (RMD).Citation 87 GDP-rhamnose is a common component of bacterial cell surface glycans. Heterologous expression of bacterial RMD in the cytosol of CHO cells allowed the GDP-fucose de novo pathway to be efficiently bypassed and afucosylated IgG antibodies to be produced.Citation 88 The dead-end product GDP-rhamnose is likely to inhibit the activity of GMD as a competitive inhibitor.

Biochemical inhibitors of fucosylation

To complement existing platforms that involve genetic engineering of cell lines for the production of afucosylated antibodies, Okeley et al. utilized small molecules to inhibit antibody fucosylation.Citation 89 2-fluorofucose and 5-alkynylfucose were shown to generate afucosylated monoclonal antibodies. The mechanism of action of these inhibitors is likely due to the depletion of intracellular GDP-fucose with a subsequent block of the de novo pathway or the inhibition of FUT8.

Plant cells as expression platforms

In addition to CHO cells, alternative expression platforms such as plants have also been reported for production of recombinant antibodies.Citation 90 Unlike CHO cells, glycoproteins produced from plants lack α1,6-fucose, β1,4-galactose and α2,3-sialic acid. Plant N-glycans typically contains a Man3GlcNAc2 core modified with β1,2-xylose and α1,3-fucose. Large complex type N-glycans with mammalian Lea structure containing α1,4-fucose and β1,3-galactose residues were sometimes observed.Citation 91 Antibody N-glycans produced in plants are predominantly GnGnXF3 structures containing the unwanted residues β1,2-xylose and core α1,3-fucose.Citation 92 , Citation 93 These sugars are immunogenic to humans, and serum antibodies against core xylose and core α1,3-fucose have been detected in healthy human blood donors.Citation 94 Strategies to overcome this immunogenicity include use of RNAi knockdown of α1,3-fucosyltransferase (FucT) and β1,2-xylosyltransferase (XylT) in plantsCitation 95 , Citation 96 and FucT/XylT-knockout lines.Citation 97 , Citation 98 An afucosylated anti-CD30 monoclonal antibody with G0 structure was produced using glycoengineered aquatic plant Lemna minor and shown to have improved ADCC over the same CHO cell-produced antibody.Citation 95 Anti-HIV 2G12 produced in XylT/FucT-knockdown N. benthamiana was found to be homogeneous G0 structures with terminal N-acetylglucosamine and lacking both xylose and α1,3-fucose residues.Citation 96 Further glycoengineering in XylT/FucT knockdown N. benthamiana by expressing a modified human β1,4-galactosyltransferase was reported to produce anti-HIV monoclonal antibodies with fully β1,4-galactosylated N-glycans and improved virus neutralization potency.Citation 99

Chemoenzymatic remodelling strategy

Chemoenzymatic remodelling of antibodies represents another strategy for generating afucosylated antibodies. This chemical biology approach involves the use of an endo-β-N-acetylglucosamidase such as Endo S to remove the majority of N-glycans from antibodies, followed by treatment with an exoglycosidase such as fucosidase to remove the core fucose. The mono-GlcNac is then further extended by transglycosylation with Endo S-based glycosynthases in the presence of desialylated complex type glycan oxazoline, which serve as donor substrates to generate different homogenous afucosylated glycoforms.Citation 100 However, this method is not cost effective for producing afucosylated therapeutic antibodies.

Enhanced ADCC activities by afucosylated antibodies in in vivo studies

The efficacy of numerous afucosylated antibodies have been investigated in vivo using animal models. The studies that have been published are compiled into . The diseases targeted by these antibodies include cancers, viral infections and inflammatory disorders.

Table 1. Summary of glycoengineered antibodies that have been studied in vivo in animal models.

CD20 is one of the most promising targets for B cell malignancies. The treatment of B cell malignancies has evolved significantly after the US Food and Drug Administration (FDA) approved the first anti-CD20 monoclonal antibody to treat non-Hodgkin's lymphoma (NHL) in 1997. Rituximab (Rituxan®), a type I chimeric IgG1, is currently the best-selling therapeutic monoclonal antibodies marketed for the treatment of B cell malignancies and rheumatoid arthritis. An afucosylated rituximab was evaluated in animal models, and it showed enhanced B-cell depletion in cynomolgus monkeysCitation 101 and in human FcγR- and CD20-transgenic miceCitation 70 compared with fucosylated rituximab. The next-generation anti-CD20 antibody obinutuzumab (GA101 or Gazyva®) is a type II humanized Fc glycoengineered antibody with improved efficacy. This antibody, with reduced fucosylation (<30%, according to the manufacturer), showed superior tumor inhibition in NHL xenograft SCID mice and B-cell depletion in cynomolgus monkeys over rituximab.Citation 102

CD19 is another B cell marker that has been targeted by monoclonal antibodies. CD19 is particularly important because it is present on malignant B cells that have lost CD20 expression upon repeated rituximab treatment. Several groups have developed anti-CD19 antibodies that are afucosylated.Citation103-Citation107 These afucosylated antibodies generally showed enhanced B-cell depletion in murine and non-human primate models compared with the fucosylated counterparts. However, anti-CD19 monoclonal antibody MEDI-551 only showed a minor or insignificant improvement in tumor inhibition in CD19+ Raji and Daudi cell lymphoma xenograft SCID mouse models.Citation 104 The discrepancy in efficacy could be dependent on the level of CD19 on the target cell. In addition to ADCC, data suggested the importance of antibody-dependent cellular phagocytosis (ADCP) in MEDI-551-mediated B-cell depletion.Citation 105 , Citation 106

Overexpressed receptor tyrosine kinases are frequently implicated as oncogenes in a wide range of cancers. Antibodies with reduced fucosylation against receptors like EGFR, insulin-like growth factor 1 receptor and c-Met have been generated and tested in murine models.Citation108-Citation110 In addition to the anti-EGFR antibody imgatuzumab (GA201 or RG7160), bi-specific glycoengineered formats against two different receptors have also been developed.Citation 109 , Citation 110 In xenograft SCID mouse models, these afucosylated antibodies demonstrated enhanced tumor inhibition in vivo, which is probably dependent on their enhanced binding to FcγRIII on various effector cells.

Antibodies targeting several viruses that are associated with mortality have been developed as a possible means of passive immunization because no effective vaccines against these viruses are available yet. For example, respiratory syncytial virus (RSV) infection in high-risk young children and elderly is often associated with morbidity and mortality. Palivizumab, a humanized IgG1 against RSV, is suggested for preventive use in high-risk children where RSV can result in complications. Ebola virus (EBOV) is a single-stranded RNA virus that can cause hemorrhagic fever potentially leading to fatalities in humans.Citation 111 It is one of the most virulent and infectious agents known. ZMapp, a cocktail of three monoclonal antibodies produced in plants against the glycoproteins of EBOV, has been successful in passive immunization in nonhuman primates.Citation 112 HIV-1 is well known for its mortality and high rate of viral escape. Broadly neutralizing antibodies against HIV-1 gp120 have demonstrated efficacy in reducing viral load in animal studies and clinical trials.Citation 113 Antibodies against RSV, EBOV and HIV have been glycoengineered to become afucosylated to further improve their anti-viral activity.Citation 84 , Citation 114 , Citation 115 Enhanced binding to the FcγR by these afucosylated antibodies was correlated with enhanced efficacy in murine models.Citation 114 , Citation 115 For example, the afucosylated gp120-bispecific and hexavalent broadly neutralizing fusion protein – LSEVh-LS-F also showed potent inhibition of HIV-1 and simian-HIV infection in humanized mouse and macaque models through NK-cell mediated ADCC.Citation 84

In summary, the efficacies of the afucosylated antibodies have been tested in murine and non-human primates. The animal model data demonstrated enhanced in vivo efficacy, especially at lower doses, by the afucosylated antibodies. The exact in vivo mechanism of action can include a multitude of different effector functions (e.g., ADCC, ADCP). However, the significant improvement in ADCC by the afucosylated antibodies observed in the in vitro studies was often reduced in the animal models. This could be due to pharmacodynamic and pharmacokinetic effects, differences between the human and animal FcγR genotypes, and the characteristics and density of the antigens. Nevertheless, the enhanced efficacy and the tolerability of several of these glycoengineered drugs in animal studies supported progression into clinical trials.

Therapeutic afucosylated antibody drugs approved for market use and clinical trials

The encouraging results of the afucosylated antibodies in the animal models have led to their advancement into clinical trials. There are currently three afucosylated antibodies on the market and more than 20 are currently being evaluated in clinical trials (, source: https://clinicaltrials.gov/). We will discuss the approved drugs and a few selected differently glycoengineered antibodies.

Table 2. Current status of glycoengineered antibodies in clinical trials.

Obinutuzumab (GA101 or Gazyva®) is the first glycoengineered therapeutic anti-CD20 antibody approved by FDA in 2013 for the combination treatment of patients with CLL and follicular lymphoma. Reduced fucosylation is achieved through the co-expression of GnT-III and αManII in CHO cells. The antibody demonstrated an enhanced binding affinity for FcγRIIIa and consequently, an increased ADCC activity.Citation 102 Results from Phase1b/2 trials indicated that all patients with CLL experienced rapid and sustained removal of B cells in the peripheral blood.Citation116-Citation119 In Phase 3 trials, GA101 in combination with chlorambucil prolonged overall survival significantly, as well as progression-free survival and increased the complete response rate.Citation 120 In addition, this combination resulted in substantially increased time to next treatment.Citation 121

Mogamulizumab (POTELIGEO®) was first approved in 2012 in Japan for hematologic malignancies, and in 2014 for cutaneous T-cell lymphoma (CTCL). In November 2017, FDA granted it Breakthrough Therapy Designation status for the treatment of mycosis fungoides and Sézary syndrome in patients who have previously received at least one treatment. The antibody is produced in FUT8-knockout CHO cells (Biowa Potelligent Technology) to achieve afucosylation. Mogamulizumab has demonstrated effectiveness against CTCL in Phase 2 randomized controlled trials.Citation 122 Currently, it is in several clinical trials in combination with other drugs to target several forms of solid tumors. It is also in a Phase 3 clinical trial targeting human T-lymphotrophic virus 1 (HTLV1)-associated myelopathy.

Benralizumab (MEDI-563, Fasenra™) was approved by FDA in November 2017 for the treatment of severe eosinophil asthma. The antibody is produced in FUT8-knockout CHO cells (Biowa Potelligent Technology). It functions by blocking IL-5R signalling and ADCC-mediated depletion of IL-5Rα-expressing eosinophils.Citation 123 Benralizumab has completed seven Phase 3 studies for asthma treatment. Based on two published Phase 3 studies, benralizumab has reduced the annual exacerbation rate for patients having severe uncontrolled eosinophilic asthma despite treatment with medium to high dosage of inhaled corticosteroids and long-acting beta2-agonists.Citation 124 , Citation 125 Currently, it is being tested in several clinical trials against eosinophilic chronic rhinosinusitis and chronic obstructive pulmonary disease. A late-phase clinical trial is testing benralizumab for the treatment of patients with chronic allergic reaction to drugs or food, a condition known as chronic idiopathic urticaria, who are unresponsive to H1-antihistamines.

Ublituximab is a chimeric anti-CD20 IgG1 antibody produced in the YB2/0 cell line, which generates antibodies with low fucose and consequently higher ADCC.Citation 126 Ublituximab has completed several Phase 1 and 2 clinical trials against B cell malignancies. In a Phase 1/2 clinical trial in patients with B cell NHL or CLL previously treated with rituximab, ublituximab was well tolerated and efficacious.Citation 127 Currently, it is in several clinical trials in combination with other drugs for treatment of patients with CLL. It is also being tested in combination with the drug teriflunomide for safety and efficacy in patients with relapsing multiple sclerosis in Phase 3 clinical trials.

TrasGEX (GT-MAB7.3-GEX, Glycooptimized Trastuzumab-GEX) was developed by Glycotope. It is a humanized anti-HER2 IgG1 that is glycoengineered through the GlycoExpress Technology, which yields antibodies with humanized and optimized glycosylation pattern. It has completed a Phase 1 trial for dose-escalating and pharmacokinetic analysis in patients with HER2-positive cancers. In a female patient with metastatic HER2+ colorectal cancer against which all other options failed, the use of TrasGEX resulted in a 10-fold to 140-fold enhanced ADCC.Citation 128

SEA-CD40 is a humanized afucosylated anti-CD40 IgG1 developed by Seattle Genetics. The antibody is produced by the sugar-engineered antibody (SEA) technology to eliminate the fucose sugar group to enhance the ADCC activity. The SEA technology involves the use of modified sugars (fucosylation inhibitor, 2-fluorofucose) to inhibit fucosylation during cell culture. Currently, it is in a Phase 1 trial for a range of patients with cancer such as Hodgkin disease, non-small cell lung cancer and melanoma.

Concluding remarks

ADCC is one of the critical effector functions triggered when a therapeutic antibody is used to eliminate target cells. Antibodies specific for CD20 and CD19 have been used to treat B cell malignancies by triggering ADCC. Anti-CD20 antibody rituximab has also been used to deplete B cells in rheumatoid arthritis patients. Antibodies specific for EGFR have been used to target EGFR-positive tumors. Elevated levels of eosinophils in certain severe asthma patients can be removed by antibodies against IL-5Rα on eosinophils. Studies have shown that antibodies can eliminate HIV- or influenza virus-infected cells by the same mechanism. As discussed in this article, removal of fucose from all these antibodies has significantly improved their ADCC activity in in vitro and in vivo studies.

The enhanced ADCC activity by afucosylated antibodies was discovered by in vitro binding analyses and cell-based ADCC assays. The initial in vitro observations have been confirmed in in vivo animal models and clinical studies. The enhanced affinity is the result of a unique carbohydrate-carbohydrate interaction between the N-glycan of the IgG and the N-glycan of the FcγRIIIa at Asn162. This is the first example where two glycans from two binding partners interact and the carbohydrate-carbohydrate interaction significantly modulates the binding affinity between the two proteins. Because of this novel phenomenon, various approaches have been utilized to target the fucosylation machinery of the host cell lines. A more economically effective approach involves the glycoengineering of mammalian cell lines to produce afucosylated antibodies. As of today, at least 35 glycoengineered antibodies, with their Fc fucose partially or completely removed, have been investigated in animal models (), 26 of them have been studied in clinical trials and 3 have been approved for use in clinical practice (). We expect that more afucosylated antibodies will enter clinical trials and subsequently be approved for clinical use.

Abbreviations

AFP=

Alpha-fetoprotein

αManII=

α-mannosidase II

ADCC=

antibody-dependent cellular cytotoxicity

ADCP=

antibody-dependent cellular phagocytosis

EGFR=

epidermal growth factor receptor

FUT=

fucosyltransferase

GFT=

GDP-fucose transporter

GKDM=

GDP-4-keto-6-deoxy mannose

GnT-III=

β-1,4-mannosyl-glycoprotein 4-β-N-acetylglucosaminyltransferase

GMD=

GDP-mannose 4,6-dehydratase

HCC=

hepatocellular carcinoma

IL-5R=

interleukin-5 receptor

NK cells=

natural killer cells

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Acknowledgments

This work was supported by A*STAR BMRC Strategic Positioning Fund. The authors would like to thank Mr. Ryan Haryadi and Dr. Irene Kiess for careful review of the manuscript.

Additional information

Funding

Agency for Science, Technology and Research (A*STAR), Singapore.

References

  • Ecker DM, Jones SD, Levine HL. The therapeutic monoclonal antibody market. MAbs. 2015;7:9–14. doi:10.4161/19420862.2015.989042. PMID:25529996
  • Reichert JM. Antibodies to watch in 2015. MAbs. 2015;7:1–8. doi:10.4161/19420862.2015.988944. PMID:25484055.
  • Reichert JM. Antibodies to watch in 2016. MAbs. 2016;8:197–204. doi:10.1080/19420862.2015.1125583. PMID:26651519.
  • Reichert JM. Antibodies to watch in 2017. MAbs. 2017;9:167–81. doi:10.1080/19420862.2016.1269580. PMID:27960628.
  • Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, Watier H. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood. 2002;99:754–8. doi:10.1182/blood.V99.3.754. PMID:11806974.
  • Musolino A, Naldi N, Bortesi B, Pezzuolo D, Capelletti M, Missale G, Laccabue D, Zerbini A, Camisa R, Bisagni G, et al. Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol. 2008;26:1789–96. doi:10.1200/JCO.2007.14.8957. PMID:18347005.
  • Weng WK, Levy R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J Clin Oncol. 2003;21:3940–7. doi:10.1200/JCO.2003.05.013. PMID:12975461.
  • Radaev S, Motyka S, Fridman WH, Sautes-Fridman C, Sun PD. The structure of a human type III Fcgamma receptor in complex with Fc. J Biol Chem. 2001;276:16469–77. doi:10.1074/jbc.M100350200. PMID:11297532.
  • Sondermann P, Huber R, Oosthuizen V, Jacob U. The 3.2-A crystal structure of the human IgG1 Fc fragment-Fc gammaRIII complex. Nature. 2000;406:267–73. doi:10.1038/35018508. PMID:10917521.
  • 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. doi:10.1016/S0022-2836(02)01250-0. PMID:12527303.
  • Dashivets T, Thomann M, Rueger P, Knaupp A, Buchner J, Schlothauer T. Multi-angle effector function analysis of human monoclonal IgG Glycovariants. PLoS One. 2015;10:e0143520. doi:10.1371/journal.pone.0143520. PMID:26657484.
  • Shields RL, Namenuk AK, Hong K, Meng YG, Rae J, Briggs J, Xie D, Lai J, Stadlen A, Li B, et al. High resolution mapping of the binding site on human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the Fc gamma R. J Biol Chem. 2001;276:6591–604. doi:10.1074/jbc.M009483200. PMID:11096108.
  • Iida S, Misaka H, Inoue M, Shibata M, Nakano R, Yamane-Ohnuki N, Wakitani M, Yano K, Shitara K, Satoh M. Nonfucosylated therapeutic IgG1 antibody can evade the inhibitory effect of serum immunoglobulin G on antibody-dependent cellular cytotoxicity through its high binding to FcgammaRIIIa. Clin Cancer Res. 2006;12:2879–87. doi:10.1158/1078-0432.CCR-05-2619. PMID:16675584.
  • Shields RL, Lai J, Keck R, O'Connell LY, Hong K, Meng YG, Weikert SH, Presta LG. Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human Fcgamma RIII and antibody-dependent cellular toxicity. J Biol Chem. 2002;277:26733–40. doi:10.1074/jbc.M202069200. PMID:11986321.
  • Shinkawa T, Nakamura K, Yamane N, Shoji-Hosaka E, Kanda Y, Sakurada M, Uchida K, Anazawa H, Satoh M, Yamasaki M, et al. 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. 2003;278:3466–73. doi:10.1074/jbc.M210665200. PMID:12427744.
  • Pucic M, Knezevic A, Vidic J, Adamczyk B, Novokmet M, Polasek O, Gornik O, Supraha-Goreta S, Wormald MR, Redzic I, et al. High throughput isolation and glycosylation analysis of IgG-variability and heritability of the IgG glycome in three isolated human populations. Mol Cell Proteomics. 2011;10:M111 010090. doi:10.1074/mcp.M111.010090. PMID:21653738.
  • Ackerman ME, Crispin M, Yu X, Baruah K, Boesch AW, Harvey DJ, Dugast AS, Heizen EL, Ercan A, Choi I, et al. Natural variation in Fc glycosylation of HIV-specific antibodies impacts antiviral activity. J Clin Invest. 2013;123:2183–92. doi:10.1172/JCI65708. PMID:23563315.
  • Koene HR, Kleijer M, Algra J, Roos D, von dem Borne AE, de Haas M. Fc gammaRIIIa-158V/F polymorphism influences the binding of IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa-48L/R/H phenotype. Blood. 1997;90:1109–14. PMID:9242542.
  • Wu J, Edberg JC, Redecha PB, Bansal V, Guyre PM, Coleman K, Salmon JE, Kimberly RP. A novel polymorphism of FcgammaRIIIa (CD16) alters receptor function and predisposes to autoimmune disease. J Clin Invest. 1997;100:1059–70. doi:10.1172/JCI119616. PMID:9276722.
  • Ferrara C, Stuart F, Sondermann P, Brunker P, Umana P. The carbohydrate at FcgammaRIIIa Asn-162. An element required for high affinity binding to non-fucosylated IgG glycoforms. J Biol Chem. 2006;281:5032–6. doi:10.1074/jbc.M510171200. PMID:16330541.
  • Veeramani S, Wang SY, Dahle C, Blackwell S, Jacobus L, Knutson T, Button A, Link BK, Weiner GJ. Rituximab infusion induces NK activation in lymphoma patients with the high-affinity CD16 polymorphism. Blood. 2011;118:3347–9. doi:10.1182/blood-2011-05-351411. PMID:21768303.
  • Treon SP, Hansen M, Branagan AR, Verselis S, Emmanouilides C, Kimby E, Frankel SR, Touroutoglou N, Turnbull B, Anderson KC, et al. Polymorphisms in FcgammaRIIIA (CD16) receptor expression are associated with clinical response to rituximab in Waldenstrom's macroglobulinemia. J Clin Oncol. 2005;23:474–81. doi:10.1200/JCO.2005.06.059. PMID:15659493.
  • Calemma R, Ottaiano A, Trotta AM, Nasti G, Romano C, Napolitano M, Galati D, Borrelli P, Zanotta S, Cassata A, et al. Fc gamma receptor IIIa polymorphisms in advanced colorectal cancer patients correlated with response to anti-EGFR antibodies and clinical outcome. J Translational Med. 2012;10:232. doi:10.1186/1479-5876-10-232..
  • Becker DJ, Lowe JB. Fucose: biosynthesis and biological function in mammals. Glycobiology. 2003;13:41R–53R. doi:10.1093/glycob/cwg054. PMID:12651883.
  • Scharberg EA, Olsen C, Bugert P. The H blood group system. Immunohematology. 2016;32:112–118. PMID:27834485.
  • Marionneau S, Cailleau-Thomas A, Rocher J, Le Moullac-Vaidye B, Ruvoën N, Clément M, Le Pendu J. ABH and Lewis histo-blood group antigens, a model for the meaning of oligosaccharide diversity in the face of a changing world. Biochimie. 2001;83:565–73. PMID:11522384.
  • Lowe JB. Glycan-dependent leukocyte adhesion and recruitment in inflammation. Curr Opin Cell Biol. 2003;15:531–8. doi:10.1016/j.ceb.2003.08.002. PMID:14519387.
  • Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinical implications. Nat Rev Cancer. 2015;15:540–55. doi:10.1038/nrc3982. PMID:26289314.
  • Vasconcelos-Dos-Santos A, Oliveira IA, Lucena MC, Mantuano NR, Whelan SA, Dias WB, Todeschini AR. Biosynthetic machinery involved in aberrant Glycosylation: Promising targets for developing of drugs against cancer. Frontiers Oncol. 2015;5:138. doi:10.3389/fonc.2015.00138..
  • Hauselmann I, Borsig L. Altered tumor-cell glycosylation promotes metastasis. Frontiers Oncol. 2014;4:28. doi:10.3389/fonc.2014.00028..
  • Elola MT, Capurro MI, Barrio MM, Coombs PJ, Taylor ME, Drickamer K, Mordoh J. Lewis x antigen mediates adhesion of human breast carcinoma cells to activated endothelium. Possible involvement of the endothelial scavenger receptor C-type lectin. Breast Cancer Res Treatment. 2007;101:161–74. doi:10.1007/s10549-006-9286-9..
  • Takada A, Ohmori K, Yoneda T, Tsuyuoka K, Hasegawa A, Kiso M, Kannagi R. Contribution of carbohydrate antigens sialyl Lewis A and sialyl Lewis X to adhesion of human cancer cells to vascular endothelium. Cancer Res. 1993;53:354–61. PMID:7678075.
  • Dingjan T, Spendlove I, Durrant LG, Scott AM, Yuriev E, Ramsland PA. Structural biology of antibody recognition of carbohydrate epitopes and potential uses for targeted cancer immunotherapies. Mol Immunol. 2015;67:75–88. doi:10.1016/j.molimm.2015.02.028. PMID:25757815.
  • Manimala JC, Roach TA, Li Z, Gildersleeve JC. High-throughput carbohydrate microarray profiling of 27 antibodies demonstrates widespread specificity problems. Glycobiology. 2007;17:17C–23C. doi:10.1093/glycob/cwm047. PMID:17483136.
  • Padler-Karavani V. Aiming at the sweet side of cancer: aberrant glycosylation as possible target for personalized-medicine. Cancer Lett. 2014;352:102–12. doi:10.1016/j.canlet.2013.10.005. PMID:24141190.
  • Rabu C, McIntosh R, Jurasova Z, Durrant L. Glycans as targets for therapeutic antitumor antibodies. Future Oncol. 2012;8:943–60. doi:10.2217/fon.12.88. PMID:22894669.
  • Cordon-Cardo C, Lloyd KO, Sakamoto J, McGroarty ME, Old LJ, Melamed MR. Immunohistologic expression of blood-group antigens in normal human gastrointestinal tract and colonic carcinoma. Int J Cancer. 1986;37:667–76. doi:10.1002/ijc.2910370505. PMID:3516890.
  • Croce MV, Isla-Larrain M, Rabassa ME, Demichelis S, Colussi AG, Crespo M, Lacunza E, Segal-Eiras A. Lewis x is highly expressed in normal tissues: a comparative immunohistochemical study and literature revision. Pathol Oncol Res. 2007;13:130–8. doi:10.1007/BF02893488. PMID:17607374.
  • Davidson JS, Triadafilopoulos G. Blood group-related antigen expression in normal and metaplastic human upper gastrointestinal mucosa. Gastroenterology. 1992;103:1552–61. doi:10.1016/0016-5085(92)91177-6. PMID:1426874.
  • De Bolos C, Garrido M, Real FX. MUC6 apomucin shows a distinct normal tissue distribution that correlates with Lewis antigen expression in the human stomach. Gastroenterology. 1995;109:723–34. doi:10.1016/0016-5085(95)90379-8. PMID:7657100.
  • de Bolos C, Real FX, Lopez-Ferrer A. Regulation of mucin and glycoconjugate expression: from normal epithelium to gastric tumors. Frontiers Biosci. 2001;6:D1256–63.
  • Itzkowitz SH, Yuan M, Fukushi Y, Palekar A, Phelps PC, Shamsuddin AM, Trump BF, Hakomori S, Kim YS. Lewisx- and sialylated Lewisx-related antigen expression in human malignant and nonmalignant colonic tissues. Cancer Res. 1986;46:2627–32. PMID:3516383.
  • Kim YS, Yuan M, Itzkowitz SH, Sun QB, Kaizu T, Palekar A, Trump BF, Hakomori S. Expression of LeY and extended LeY blood group-related antigens in human malignant, premalignant, and nonmalignant colonic tissues. Cancer Res. 1986;46:5985–92. PMID:2428490.
  • Kirkeby S, Moe D. Expression of the carcinoma markers: the sialylated Lewis A and X carbohydrate antigens in normal laryngeal surface epithelium and submucosal glands from old humans. APMIS. 2013;121:182–8. doi:10.1111/j.1600-0463.2012.02954.x. PMID:23030724.
  • Kobayashi K, Sakamoto J, Kito T, Yamamura Y, Koshikawa T, Fujita M, Watanabe T, Nakazato H. Lewis blood group-related antigen expression in normal gastric epithelium, intestinal metaplasia, gastric adenoma, and gastric carcinoma. Am J Gastroenterol. 1993;88:919–24. PMID:8503388.
  • Lamblin G, Degroote S, Perini JM, Delmotte P, Scharfman A, Davril M, Lo-Guidice JM, Houdret N, Dumur V, Klein A, et al. Human airway mucin glycosylation: a combinatory of carbohydrate determinants which vary in cystic fibrosis. Glycoconjugate J. 2001;18:661–84. doi:10.1023/A:1020867221861..
  • Lopez-Ferrer A, de Bolos C, Barranco C, Garrido M, Isern J, Carlstedt I, Reis CA, Torrado J, Real FX. Role of fucosyltransferases in the association between apomucin and Lewis antigen expression in normal and malignant gastric epithelium. Gut. 2000;47:349–56. doi:10.1136/gut.47.3.349. PMID:10940270.
  • Robbe C, Capon C, Coddeville B, Michalski JC. Structural diversity and specific distribution of O-glycans in normal human mucins along the intestinal tract. Biochem J. 2004;384:307–16. doi:10.1042/BJ20040605. PMID:15361072.
  • Miyoshi E, Noda K, Yamaguchi Y, Inoue S, Ikeda Y, Wang W, Ko JH, Uozumi N, Li W, Taniguchi N. The alpha1-6-fucosyltransferase gene and its biological significance. Biochim Biophys Acta. 1999;1473:9–20. doi:10.1016/S0304-4165(99)00166-X. PMID:10580126.
  • Miyoshi E, Uozumi N, Noda K, Hayashi N, Hori M, Taniguchi N. Expression of alpha1-6 fucosyltransferase in rat tissues and human cancer cell lines. Int J Cancer. 1997;72:1117–21. doi:10.1002/(SICI)1097-0215(19970917)72:6%3c1117::AID-IJC29%3e3.0.CO;2-. PMID:9378548.
  • Noda K, Miyoshi E, Uozumi N, Yanagidani S, Ikeda Y, Gao C, Suzuki K, Yoshihara H, Yoshikawa K, Kawano K, et al. Gene expression of alpha1-6 fucosyltransferase in human hepatoma tissues: a possible implication for increased fucosylation of alpha-fetoprotein. Hepatology. 1998;28:944–52. doi:10.1002/hep.510280408. PMID:9755230.
  • Luo Y, Koles K, Vorndam W, Haltiwanger RS, Panin VM. Protein O-fucosyltransferase 2 adds O-fucose to thrombospondin type 1 repeats. J Biol Chem. 2006;281:9393–9. doi:10.1074/jbc.M511975200. PMID:16464857.
  • Wang Y, Shao L, Shi S, Harris RJ, Spellman MW, Stanley P, Haltiwanger RS. Modification of epidermal growth factor-like repeats with O-fucose. Molecular cloning and expression of a novel GDP-fucose protein O-fucosyltransferase. J Biol Chem. 2001;276:40338–45. doi:10.1074/jbc.M107849200. PMID:11524432.
  • Stanley P. Regulation of Notch signaling by glycosylation. Curr Opin Structural Biol. 2007;17:530–5. doi:10.1016/j.sbi.2007.09.007..
  • Tonetti M, Sturla L, Bisso A, Benatti U, De Flora A. Synthesis of GDP-L-fucose by the human FX protein. J Biol Chem. 1996;271:27274–9. doi:10.1074/jbc.271.44.27274. PMID:8910301.
  • Song Z. Roles of the nucleotide sugar transporters (SLC35 family) in health and disease. Mol Aspects Med. 2013;34:590–600. doi:10.1016/j.mam.2012.12.004. PMID:23506892.
  • Etzioni A, Frydman M, Pollack S, Avidor I, Phillips ML, Paulson JC, Gershoni-Baruch R. Brief report: recurrent severe infections caused by a novel leukocyte adhesion deficiency. N Engl J Med. 1992;327:1789–92. doi:10.1056/NEJM199212173272505. PMID:1279426.
  • Hirschberg CB, Robbins PW, Abeijon C. Transporters of nucleotide sugars, ATP, and nucleotide sulfate in the endoplasmic reticulum and Golgi apparatus. Annual Rev Biochem. 1998;67:49–69. doi:10.1146/annurev.biochem.67.1.49..
  • Lubke T, Marquardt T, Etzioni A, Hartmann E, von Figura K, Korner C. Complementation cloning identifies CDG-IIc, a new type of congenital disorders of glycosylation, as a GDP-fucose transporter deficiency. Nat Genet. 2001;28:73–6. doi:10.1038/ng0501-73. PMID:11326280.
  • Luhn K, Wild MK, Eckhardt M, Gerardy-Schahn R, Vestweber D. The gene defective in leukocyte adhesion deficiency II encodes a putative GDP-fucose transporter. Nat Genet. 2001;28:69–72. doi:10.1038/ng0501-69. PMID:11326279.
  • Ohyama C, Smith PL, Angata K, Fukuda MN, Lowe JB, Fukuda M. Molecular cloning and expression of GDP-D-mannose-4,6-dehydratase, a key enzyme for fucose metabolism defective in Lec13 cells. J Biol Chem. 1998;273:14582–7. doi:10.1074/jbc.273.23.14582. PMID:9603974.
  • Masuda K, Kubota T, Kaneko E, Iida S, Wakitani M, Kobayashi-Natsume Y, Kubota A, Shitara K, Nakamura K. Enhanced binding affinity for FcgammaRIIIa of fucose-negative antibody is sufficient to induce maximal antibody-dependent cellular cytotoxicity. Mol Immunol. 2007;44:3122–31. doi:10.1016/j.molimm.2007.02.005. PMID:17379311.
  • Okazaki A, Shoji-Hosaka E, Nakamura K, Wakitani M, Uchida K, Kakita S, Tsumoto K, Kumagai I, Shitara K. Fucose depletion from human IgG1 oligosaccharide enhances binding enthalpy and association rate between IgG1 and FcgammaRIIIa. J Mol Biol. 2004;336:1239–49. doi:10.1016/j.jmb.2004.01.007. PMID:15037082.
  • Ferrara C, Grau S, Jager C, Sondermann P, Brunker P, Waldhauer I, Hennig M, Ruf A, Rufer AC, Stihle M, 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. PMID:21768335.
  • Wang TT, Sewatanon J, Memoli MJ, Wrammert J, Bournazos S, Bhaumik SK, Pinsky BA, Chokephaibulkit K, Onlamoon N, Pattanapanyasat K, et al. IgG antibodies to dengue enhanced for FcgammaRIIIA binding determine disease severity. Science. 2017;355:395–8. PMID:28126818.
  • Wuhrer M, Porcelijn L, Kapur R, Koeleman CA, Deelder A, de Haas M, Vidarsson G. Regulated glycosylation patterns of IgG during alloimmune responses against human platelet antigens. J Proteome Res. 2009;8:450–6. PMID:18942870.
  • Thomann M, Schlothauer T, Dashivets T, Malik S, Avenal C, Bulau P, Ruger P, Reusch D. In vitro glycoengineering of IgG1 and its effect on Fc receptor binding and ADCC activity. PLoS One. 2015;10:e0134949. PMID:26266936.
  • Thomann M, Reckermann K, Reusch D, Prasser J, Tejada ML. Fc-galactosylation modulates antibody-dependent cellular cytotoxicity of therapeutic antibodies. Mol Immunol. 2016;73:69–75. PMID:27058641.
  • Houde D, Peng Y, Berkowitz SA, Engen JR. Post-translational modifications differentially affect IgG1 conformation and receptor binding. Mol Cell Proteomics. 2010;9:1716–28. PMID:20103567.
  • Li T, DiLillo DJ, Bournazos S, Giddens JP, Ravetch JV, Wang LX. Modulating IgG effector function by Fc glycan engineering. Proc Natl Acad Sci U S A. 2017;114:3485–90. PMID:28289219.
  • Lazar GA, Dang W, Karki S, Vafa O, Peng JS, Hyun L, Chan C, Chung HS, Eivazi A, Yoder SC, et al. Engineered antibody Fc variants with enhanced effector function. Proc Natl Acad Sci U S A. 2006;103:4005–10. PMID:16537476.
  • Horton HM, Bernett MJ, Pong E, Peipp M, Karki S, Chu SY, Richards JO, Vostiar I, Joyce PF, Repp R, et al. Potent in vitro and in vivo activity of an Fc-engineered anti-CD19 monoclonal antibody against lymphoma and leukemia. Cancer Res. 2008;68:8049–57. PMID:18829563.
  • Stavenhagen JB, Gorlatov S, Tuaillon N, Rankin CT, Li H, Burke S, Huang L, Vijh S, Johnson S, Bonvini E, et al. Fc optimization of therapeutic antibodies enhances their ability to kill tumor cells in vitro and controls tumor expansion in vivo via low-affinity activating Fcgamma receptors. Cancer Res. 2007;67:8882–90. PMID:17875730.
  • Nordstrom JL, Gorlatov S, Zhang W, Yang Y, Huang L, Burke S, Li H, Ciccarone V, Zhang T, Stavenhagen J, et al. Anti-tumor activity and toxicokinetics analysis of MGAH22, an anti-HER2 monoclonal antibody with enhanced Fcgamma receptor binding properties. Breast Cancer Res. 2011;13:R123. PMID:22129105.
  • Kanda Y, Yamane-Ohnuki N, Sakai N, Yamano K, Nakano R, Inoue M, Misaka H, Iida S, Wakitani M, Konno Y, et al. Comparison of cell lines for stable production of fucose-negative antibodies with enhanced ADCC. Biotechnol Bioeng. 2006;94:680–8. PMID:16609957.
  • Louie S, Haley B, Marshall B, Heidersbach A, Yim M, Brozynski M, Tang D, Lam C, Petryniak B, Shaw D, et al. FX knockout CHO hosts can express desired ratios of fucosylated or afucosylated antibodies with high titers and comparable product quality. Biotechnol Bioeng. 2017;114:632–44. PMID:27666939.
  • Yamane-Ohnuki N, Kinoshita S, Inoue-Urakubo M, Kusunoki M, Iida S, Nakano R, Wakitani M, Niwa R, Sakurada M, Uchida K, et al. Establishment of FUT8 knockout Chinese hamster ovary cells: an ideal host cell line for producing completely defucosylated antibodies with enhanced antibody-dependent cellular cytotoxicity. Biotechnol Bioeng. 2004;87:614–22. PMID:15352059.
  • Malphettes L, Freyvert Y, Chang J, Liu PQ, Chan E, Miller JC, Zhou Z, Nguyen T, Tsai C, Snowden AW, et al. Highly efficient deletion of FUT8 in CHO cell lines using zinc-finger nucleases yields cells that produce completely nonfucosylated antibodies. Biotechnol Bioeng. 2010;106:774–83. PMID:20564614.
  • Mori K, Kuni-Kamochi R, Yamane-Ohnuki N, Wakitani M, Yamano K, Imai H, Kanda Y, Niwa R, Iida S, Uchida K, et al. Engineering Chinese hamster ovary cells to maximize effector function of produced antibodies using FUT8 siRNA. Biotechnol Bioeng. 2004;88:901–8. PMID:15515168.
  • Chan KF, Shahreel W, Wan C, Teo G, Hayati N, Tay SJ, Tong WH, Yang Y, Rudd PM, Zhang P, et al. Inactivation of GDP-fucose transporter gene (Slc35c1) in CHO cells by ZFNs, TALENs and CRISPR-Cas9 for production of fucose-free antibodies. Biotechnol J. 2016;11:399–414. PMID:26471004.
  • Zhang P, Haryadi R, Chan KF, Teo G, Goh J, Pereira NA, Feng H, Song Z. Identification of functional elements of the GDP-fucose transporter SLC35C1 using a novel Chinese hamster ovary mutant. Glycobiology. 2012;22:897–911. PMID:22492235.
  • Xu X, Nagarajan H, Lewis NE, Pan S, Cai Z, Liu X, Chen W, Xie M, Wang W, Hammond S, et al. The genomic sequence of the Chinese hamster ovary (CHO)-K1 cell line. Nat Biotechnol. 2011;29:735–41. PMID:21804562.
  • Howard DR, Fukuda M, Fukuda MN, Stanley P. The GDP-fucose:N-acetylglucosaminide 3-alpha-L-fucosyltransferases of LEC11 and LEC12 Chinese hamster ovary mutants exhibit novel specificities for glycolipid substrates. J Biol Chem. 1987;262:16830–7. PMID:2890642.
  • Bardhi A, Wu Y, Chen W, Li W, Zhu Z, Zheng JH, Wong H, Jeng E, Jones J, Ochsenbauer C, et al. Potent In Vivo NK Cell-Mediated Elimination of HIV-1-infected cells mobilized by a gp120-bispecific and hexavalent broadly neutralizing fusion protein. J Virol. 2017;91:00937–17.
  • Cua S, Tan HL, Fong WJ, Chin A, Lau A, Ding V, Song Z, Yang Y, Choo A. Targeting of embryonic annexin A2 expressed on ovarian and breast cancer by the novel monoclonal antibody 2448. Oncotarget. 2018;9:13206–21. PMID:29568351.
  • Ferrara C, Brunker P, Suter T, Moser S, Puntener U, Umana P. Modulation of therapeutic antibody effector functions by glycosylation engineering: influence of Golgi enzyme localization domain and co-expression of heterologous beta1, 4-N-acetylglucosaminyltransferase III and Golgi alpha-mannosidase II. Biotechnol Bioeng. 2006;93:851–61. PMID:16435400.
  • Kneidinger B, Graninger M, Adam G, Puchberger M, Kosma P, Zayni S, Messner P. Identification of two GDP-6-deoxy-D-lyxo-4-hexulose reductases synthesizing GDP-D-rhamnose in Aneurinibacillus thermoaerophilus L420-91T. J Biol Chem. 2001;276:5577–83. PMID:11096116.
  • von Horsten HH, Ogorek C, Blanchard V, Demmler C, Giese C, Winkler K, Kaup M, Berger M, Jordan I, Sandig V. Production of non-fucosylated antibodies by co-expression of heterologous GDP-6-deoxy-D-lyxo-4-hexulose reductase. Glycobiology. 2010;20:1607–18. PMID:20639190.
  • Okeley NM, Alley SC, Anderson ME, Boursalian TE, Burke PJ, Emmerton KM, Jeffrey SC, Klussman K, Law CL, Sussman D, et al. Development of orally active inhibitors of protein and cellular fucosylation. Proc Natl Acad Sci U S A. 2013;110:5404–9. PMID:23493549.
  • Loos A, Steinkellner H. IgG-Fc glycoengineering in non-mammalian expression hosts. Arch Biochem Biophys. 2012;526:167–73. PMID:22634260.
  • Lerouge P, Cabanes-Macheteau M, Rayon C, Fischette-Laine AC, Gomord V, Faye L. N-glycoprotein biosynthesis in plants: recent developments and future trends. Plant Mol Biol. 1998;38:31–48. PMID:9738959.
  • Bakker H, Bardor M, Molthoff JW, Gomord V, Elbers I, Stevens LH, Jordi W, Lommen A, Faye L, Lerouge P, et al. Galactose-extended glycans of antibodies produced by transgenic plants. Proc Nat Acad Sci U S A. 2001;98:2899–904.
  • Bardor M, Loutelier-Bourhis C, Paccalet T, Cosette P, Fitchette AC, Vezina LP, Trepanier S, Dargis M, Lemieux R, Lange C, et al. Monoclonal C5-1 antibody produced in transgenic alfalfa plants exhibits a N-glycosylation that is homogenous and suitable for glyco-engineering into human-compatible structures. Plant Biotechnol J. 2003;1:451–62. PMID:17134403.
  • Bardor M, Faveeuw C, Fitchette AC, Gilbert D, Galas L, Trottein F, Faye L, Lerouge P. Immunoreactivity in mammals of two typical plant glyco-epitopes, core alpha(1,3)-fucose and core xylose. Glycobiology. 2003;13:427–34. PMID:12626420.
  • Cox KM, Sterling JD, Regan JT, Gasdaska JR, Frantz KK, Peele CG, Black A, Passmore D, Moldovan-Loomis C, Srinivasan M, et al. Glycan optimization of a human monoclonal antibody in the aquatic plant Lemna minor. Nat Biotechnol. 2006;24:1591–7. PMID:17128273.
  • Strasser R, Stadlmann J, Schahs M, Stiegler G, Quendler H, Mach L, Glossl J, Weterings K, Pabst M, Steinkellner H. Generation of glyco-engineered Nicotiana benthamiana for the production of monoclonal antibodies with a homogeneous human-like N-glycan structure. Plant Biotechnol J. 2008;6:392–402. PMID:18346095.
  • Schahs 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. PMID:17678502.
  • Strasser R, Altmann F, Mach L, Glossl J, Steinkellner H. Generation of Arabidopsis thaliana plants with complex N-glycans lacking beta1,2-linked xylose and core alpha1,3-linked fucose. FEBS Lett. 2004;561:132–6. PMID:15013764.
  • Strasser R, Castilho A, Stadlmann J, Kunert R, Quendler H, Gattinger P, Jez J, Rademacher T, Altmann F, Mach L, et al. Improved virus neutralization by plant-produced anti-HIV antibodies with a homogeneous beta1,4-galactosylated N-glycan profile. J Biol Chem. 2009;284:20479–85. PMID:19478090.
  • Lin CW, Tsai MH, Li ST, Tsai TI, Chu KC, Liu YC, Lai MY, Wu CY, Tseng YC, Shivatare SS, et al. A common glycan structure on immunoglobulin G for enhancement of effector functions. Proc Natl Acad Sci U S A. 2015;112:10611–6. PMID:26253764.
  • Gasdaska JR, Sherwood S, Regan JT, Dickey LF. An afucosylated anti-CD20 monoclonal antibody with greater antibody-dependent cellular cytotoxicity and B-cell depletion and lower complement-dependent cytotoxicity than rituximab. Mol Immunol. 2012;50:134–41. PMID:22305040.
  • Mossner E, Brunker P, Moser S, Puntener U, Schmidt C, Herter S, Grau R, Gerdes C, Nopora A, van Puijenbroek E, et al. Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell-mediated B-cell cytotoxicity. Blood. 2010;115:4393–402. PMID:20194898.
  • Cardarelli PM, Rao-Naik C, Chen S, Huang H, Pham A, Moldovan-Loomis MC, Pan C, Preston B, Passmore D, Liu J, et al. A nonfucosylated human antibody to CD19 with potent B-cell depletive activity for therapy of B-cell malignancies. Cancer Immunol Immunother. 2010;59:257–65. PMID:19657637.
  • Ward E, Mittereder N, Kuta E, Sims GP, Bowen MA, Dall'Acqua W, Tedder T, Kiener P, Coyle AJ, Wu H, et al. A glycoengineered anti-CD19 antibody with potent antibody-dependent cellular cytotoxicity activity in vitro and lymphoma growth inhibition in vivo. Br J Haematol. 2011;155:426–37. PMID:21902688.
  • Herbst R, Wang Y, Gallagher S, Mittereder N, Kuta E, Damschroder M, Woods R, Rowe DC, Cheng L, Cook K, et al. B-cell depletion in vitro and in vivo with an afucosylated anti-CD19 antibody. J Pharmacol Exp Ther. 2010;335:213–22. PMID:20605905.
  • Matlawska-Wasowska K, Ward E, Stevens S, Wang Y, Herbst R, Winter SS, Wilson BS. Macrophage and NK-mediated killing of precursor-B acute lymphoblastic leukemia cells targeted with a-fucosylated anti-CD19 humanized antibodies. Leukemia. 2013;27:1263–74. PMID:23307031.
  • Breton CS, Nahimana A, Aubry D, Macoin J, Moretti P, Bertschinger M, Hou S, Duchosal MA, Back J. A novel anti-CD19 monoclonal antibody (GBR 401) with high killing activity against B cell malignancies. J Hematol Oncol. 2014;7:33. PMID:24731302.
  • Gerdes CA, Nicolini VG, Herter S, van Puijenbroek E, Lang S, Roemmele M, Moessner E, Freytag O, Friess T, Ries CH, et al. GA201 (RG7160): a novel, humanized, glycoengineered anti-EGFR antibody with enhanced ADCC and superior in vivo efficacy compared with cetuximab. Clin Cancer Res. 2013;19:1126–38. PMID:23209031.
  • Grugan KD, Dorn K, Jarantow SW, Bushey BS, Pardinas JR, Laquerre S, Moores SL, Chiu ML. Fc-mediated activity of EGFR x c-Met bispecific antibody JNJ-61186372 enhanced killing of lung cancer cells. MAbs. 2017;9:114–26. PMID:27786612.
  • Schanzer JM, Wartha K, Croasdale R, Moser S, Kunkele KP, Ries C, Scheuer W, Duerr H, Pompiati S, Pollman J, et al. A novel glycoengineered bispecific antibody format for targeted inhibition of epidermal growth factor receptor (EGFR) and insulin-like growth factor receptor type I (IGF-1R) demonstrating unique molecular properties. J Biol Chem. 2014;289:18693–706. PMID:24841203.
  • Feldmann H, Geisbert TW. Ebola haemorrhagic fever. Lancet. 2011;377:849–62. PMID:21084112.
  • Haque A, Hober D, Blondiaux J. Addressing therapeutic options for ebola virus infection in current and future outbreaks. Antimicrob Agents Chemother. 2015;59:5892–902. PMID:26248374.
  • Stephenson KE, Barouch DH. Broadly Neutralizing Antibodies for HIV Eradication. Current HIV/AIDS Reports. 2016;13:31–7. PMID:26841901.
  • Hiatt A, Bohorova N, Bohorov O, Goodman C, Kim D, Pauly MH, Velasco J, Whaley KJ, Piedra PA, Gilbert BE, et al. Glycan variants of a respiratory syncytial virus antibody with enhanced effector function and in vivo efficacy. Proc Natl Acad Sci U S A. 2014;111:5992–7. PMID:24711420.
  • Zeitlin L, Pettitt J, Scully C, Bohorova N, Kim D, Pauly M, Hiatt A, Ngo L, Steinkellner H, Whaley KJ, et al. Enhanced potency of a fucose-free monoclonal antibody being developed as an Ebola virus immunoprotectant. Proc Natl Acad Sci U S A. 2011;108:20690–4. PMID:22143789.
  • Cartron G, de Guibert S, Dilhuydy MS, Morschhauser F, Leblond V, Dupuis J, Mahe B, Bouabdallah R, Lei G, Wenger M, et al. Obinutuzumab (GA101) in relapsed/refractory chronic lymphocytic leukemia: final data from the phase 1/2 GAUGUIN study. Blood. 2014;124:2196–202. PMID:25143487.
  • Morschhauser FA, Cartron G, Thieblemont C, Solal-Celigny P, Haioun C, Bouabdallah R, Feugier P, Bouabdallah K, Asikanius E, Lei G, et al. Obinutuzumab (GA101) monotherapy in relapsed/refractory diffuse large b-cell lymphoma or mantle-cell lymphoma: results from the phase II GAUGUIN study. J Clin Oncol. 2013;31:2912–9. PMID:23835718.
  • Salles G, Morschhauser F, Lamy T, Milpied N, Thieblemont C, Tilly H, Bieska G, Asikanius E, Carlile D, Birkett J, et al. Phase 1 study results of the type II glycoengineered humanized anti-CD20 monoclonal antibody obinutuzumab (GA101) in B-cell lymphoma patients. Blood. 2012;119:5126–32. PMID:22431570.
  • Salles GA, Morschhauser F, Solal-Celigny P, Thieblemont C, Lamy T, Tilly H, Gyan E, Lei G, Wenger M, Wassner-Fritsch E, et al. Obinutuzumab (GA101) in patients with relapsed/refractory indolent non-Hodgkin lymphoma: results from the phase II GAUGUIN study. J Clin Oncol. 2013;31:2920–6. PMID:23835715.
  • Goede V, Fischer K, Busch R, Engelke A, Eichhorst B, Wendtner CM, Chagorova T, de la Serna J, Dilhuydy MS, Illmer T, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med. 2014;370:1101–10. PMID:24401022.
  • Goede V, Fischer K, Engelke A, Schlag R, Lepretre S, Montero LF, Montillo M, Fegan C, Asikanius E, Humphrey K, et al. Obinutuzumab as frontline treatment of chronic lymphocytic leukemia: updated results of the CLL11 study. Leukemia. 2015;29:1602–4. PMID:25634683.
  • Duvic M, Pinter-Brown LC, Foss FM, Sokol L, Jorgensen JL, Challagundla P, Dwyer KM, Zhang X, Kurman MR, Ballerini R, et al. Phase 1/2 study of mogamulizumab, a defucosylated anti-CCR4 antibody, in previously treated patients with cutaneous T-cell lymphoma. Blood. 2015;125:1883–9. PMID:25605368.
  • Kolbeck R, Kozhich A, Koike M, Peng L, Andersson CK, Damschroder MM, Reed JL, Woods R, Dall'acqua WW, Stephens GL, et al. MEDI-563, a humanized anti-IL-5 receptor alpha mAb with enhanced antibody-dependent cell-mediated cytotoxicity function. J Allergy Clin Immunol. 2010;125:1344-53 e2.
  • FitzGerald JM, Bleecker ER, Nair P, Korn S, Ohta K, Lommatzsch M, Ferguson GT, Busse WW, Barker P, Sproule S, et al. Benralizumab, an anti-interleukin-5 receptor alpha monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 2016;388:2128–41. PMID:27609406.
  • Bleecker ER, FitzGerald JM, Chanez P, Papi A, Weinstein SF, Barker P, Sproule S, Gilmartin G, Aurivillius M, Werkstrom V, et al. Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting beta2-agonists (SIROCCO): a randomised, multicentre, placebo-controlled phase 3 trial. Lancet. 2016;388:2115–27. PMID:27609408.
  • Beck A, Reichert JM. Marketing approval of mogamulizumab: a triumph for glyco-engineering. MAbs. 2012;4:419–25. PMID:22699226.
  • Sawas A, Farber CM, Schreeder MT, Khalil MY, Mahadevan D, Deng C, Amengual JE, Nikolinakos PG, Kolesar JM, Kuhn JG, et al. A phase 1/2 trial of ublituximab, a novel anti-CD20 monoclonal antibody, in patients with B-cell non-Hodgkin lymphoma or chronic lymphocytic leukaemia previously exposed to rituximab. Br J Haematol. 2017;177:243–53. PMID:28220479.
  • Eisner F, Pichler M, Goletz S, Stoeger H, Samonigg H. A glyco-engineered anti-HER2 monoclonal antibody (TrasGEX) induces a long-lasting remission in a patient with HER2 overexpressing metastatic colorectal cancer after failure of all available treatment options. J Clin Pathol. 2015;68:1044–6. PMID:26386048.
  • Gallagher S, Turman S, Yusuf I, Akhgar A, Wu Y, Roskos LK, Herbst R, Wang Y. Pharmacological profile of MEDI-551, a novel anti-CD19 antibody, in human CD19 transgenic mice. Int Immunopharmacol. 2016;36:205–12. PMID:27163209.
  • Schanzer JM, Wartha K, Moessner E, Hosse RJ, Moser S, Croasdale R, Trochanowska H, Shao C, Wang P, Shi L, et al. XGFR*, a novel affinity-matured bispecific antibody targeting IGF-1R and EGFR with combined signaling inhibition and enhanced immune activation for the treatment of pancreatic cancer. MAbs. 2016;8:811–27. PMID:26984378.
  • Dalle S, Reslan L, Besseyre de Horts T, Herveau S, Herting F, Plesa A, Friess T, Umana P, Klein C, Dumontet C. Preclinical studies on the mechanism of action and the anti-lymphoma activity of the novel anti-CD20 antibody GA101. Molecular cancer therapeutics. 2011;10:178–85.
  • Vail ME, Murone C, Tan A, Hii L, Abebe D, Janes PW, Lee FT, Baer M, Palath V, Bebbington C, et al. Targeting EphA3 inhibits cancer growth by disrupting the tumor stromal microenvironment. Cancer Res. 2014;74:4470–81. PMID:25125683.
  • Junttila TT, Parsons K, Olsson C, Lu Y, Xin Y, Theriault J, Crocker L, Pabonan O, Baginski T, Meng G, et al. Superior in vivo efficacy of afucosylated trastuzumab in the treatment of HER2-amplified breast cancer. Cancer Res. 2010;70:4481–9. PMID:20484044.
  • Mirschberger C, Schiller CB, Schraml M, Dimoudis N, Friess T, Gerdes CA, Reiff U, Lifke V, Hoelzlwimmer G, Kolm I, et al. RG7116, a therapeutic antibody that binds the inactive HER3 receptor and is optimized for immune effector activation. Cancer Res. 2013;73:5183–94. PMID:23780344.
  • Niwa R, Shoji-Hosaka E, Sakurada M, Shinkawa T, Uchida K, Nakamura K, Matsushima K, Ueda R, Hanai N, Shitara K. 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. PMID:15026353.
  • Yano H, Ishida T, Inagaki A, Ishii T, Ding J, Kusumoto S, Komatsu H, Iida S, Inagaki H, Ueda R. Defucosylated anti CC chemokine receptor 4 monoclonal antibody combined with immunomodulatory cytokines: a novel immunotherapy for aggressive/refractory Mycosis fungoides and Sezary syndrome. Clin Cancer Res. 2007;13:6494–500. PMID:17975162.
  • Ito A, Ishida T, Utsunomiya A, Sato F, Mori F, Yano H, Inagaki A, Suzuki S, Takino H, Ri M, et al. Defucosylated anti-CCR4 monoclonal antibody exerts potent ADCC against primary ATLL cells mediated by autologous human immune cells in NOD/Shi-scid, IL-2R gamma(null) mice in vivo. J Immunol. 2009;183:4782–91. PMID:19748990.
  • Ishii T, Ishida T, Utsunomiya A, Inagaki A, Yano H, Komatsu H, Iida S, Imada K, Uchiyama T, Akinaga S, et al. Defucosylated humanized anti-CCR4 monoclonal antibody KW-0761 as a novel immunotherapeutic agent for adult T-cell leukemia/lymphoma. Clin Cancer Res. 2010;16:1520–31. PMID:20160057.
  • Hsi ED, Steinle R, Balasa B, Szmania S, Draksharapu A, Shum BP, Huseni M, Powers D, Nanisetti A, Zhang Y, et al. CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin Cancer Res. 2008;14:2775–84. PMID:18451245.
  • Gomathinayagam S, Laface D, Houston-Cummings NR, Mangadu R, Moore R, Shandil I, Sharkey N, Li H, Stadheim TA, Zha D. In vivo anti-tumor efficacy of afucosylated anti-CS1 monoclonal antibody produced in glycoengineered Pichia pastoris. J Biotechnol. 2015;208:13–21. PMID:26015261.
  • Silence K, Dreier T, Moshir M, Ulrichts P, Gabriels SM, Saunders M, Wajant H, Brouckaert P, Huyghe L, Van Hauwermeiren T, et al. ARGX-110, a highly potent antibody targeting CD70, eliminates tumors via both enhanced ADCC and immune checkpoint blockade. MAbs. 2014;6:523–32. PMID:24492296.
  • Nagase-Zembutsu A, Hirotani K, Yamato M, Yamaguchi J, Takata T, Yoshida M, Fukuchi K, Yazawa M, Takahashi S, Agatsuma T. Development of DS-5573a: A novel afucosylated mAb directed at B7-H3 with potent antitumor activity. Cancer Sci. 2016;107:674–81. PMID:26914241.
  • Estupina P, Fontayne A, Barret JM, Kersual N, Dubreuil O, Le Blay M, Pichard A, Jarlier M, Pugniere M, Chauvin M, et al. The anti-tumor efficacy of 3C23K, a glyco-engineered humanized anti-MISRII antibody, in an ovarian cancer model is mainly mediated by engagement of immune effector cells. Oncotarget. 2017;8:37061–79. PMID:28427157.