Skip to main content
mAbs logoLink to mAbs
. 2018 May 7;10(5):693–711. doi: 10.1080/19420862.2018.1466767

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

Natasha A Pereira 1, Kah Fai Chan 1, Pao Chun Lin 1, Zhiwei Song 1,
PMCID: PMC6150623  PMID: 29733746

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.

KEYWORDS: ADCC, Fc fucosylation, Fc gamma receptors, in vivo efficacy, therapeutic monoclonal antibodies

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.1-4 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.5-7

The FcγRIII binds the Fc region of IgG1 antibodies by interacting with the hinge region and the CH2 domain. 8 , 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. 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. 11 Several approaches have been utilized to increase the affinity between antibody and the FcγRIII. These include engineering the Fc region through amino acid mutations 12 and glycoengineering the Fc N-glycan to reduce core fucose.13-15 It is now widely recognized that removal of the core fucose from Fc N-glycans represents the most effective approach to enhance ADCC activity. 14 , 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. 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. 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). 18 , 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. 20 Cells expressing the FcγRIIIa-V158 allele mediate ADCC more effectively. 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. 5 , 21-23

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. 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. 25 , 26 Lewis-related tri- or tetra-saccharides play critical roles in leukocyte adhesion during inflammatory response and lymphocyte homing. 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. 28 , 29 SLea or CA 19-9 (cancer antigen 19-9) is one of the commonly used tumor markers in clinics. 28 , 30 Lewis antigens may contribute to adhesion of cancer cells to vascular endothelium and promote hematogenous metastasis of cancer cells. 31 , 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.33-36 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.37-48 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. 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. 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. 50 , 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. 52 , 53 O-fucosylation of Notch protein is essential for Notch signaling which plays an important role in the regulation of embryonic development. 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). 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. 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). 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.57-60

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. 14 Antibodies produced in Lec13 cells carry a significant amount of afucosylated N-glycans due to the mutated GMD gene in these cells. 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. 15 Another study also suggested that the removal of core fucose from antibodies was sufficient to achieve maximal ADCC activity. 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. 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. 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). 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. 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. 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. 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. 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. 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. 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. 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. 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. 19 Furthermore, in all FcγRs, the regions that interact with the antibody are highly conserved, yet all other receptors lack this glycosylation site. 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. 65 , 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. 11 , 67-69 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. 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. 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. 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. 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. 12

With the use of computational structure-based design and high-throughput screening, a series of engineered Fc variants were generated. 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. 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. 72 In vivo, it showed enhanced anti-tumor effect in mouse lymphoma xenograft over the wild type analogue. 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. 73 An Fc variant 18 with several mutations (F243L/R292P/Y300L/V305I/P396L) was identified and demonstrated about 100-fold enhanced ADCC activity. 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. 74 MGAH22 showed enhanced affinity to both FcγRIIIa variants (F158 and V158), but decreased affinity to inhibitory FcγRIIb. 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. 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). 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. 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. 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. 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. 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). 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. 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. 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. 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. 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. 59 , 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. 80 , 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). 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. 80 The CHO-K1 transcriptome data have shown that among all Golgi fucosyltransferases, only FUT8 is expressed. 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. 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. 80 This strategy has been used to produce afucosylated antibodies in a few recent studies. 84 , 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. 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). 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. 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. 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. 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. 91 Antibody N-glycans produced in plants are predominantly GnGnXF3 structures containing the unwanted residues β1,2-xylose and core α1,3-fucose. 92 , 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. 94 Strategies to overcome this immunogenicity include use of RNAi knockdown of α1,3-fucosyltransferase (FucT) and β1,2-xylosyltransferase (XylT) in plants 95 , 96 and FucT/XylT-knockout lines. 97 , 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. 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. 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. 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. 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 Table 1. 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.

Name and format Target Fucosylation level Method of glycoengineering Result in in vivo model (murine/non-human primates) Reference
BLX-300 (Rituximab) CD20 afucosylated Lemna aquatic plant-based system with RNA silencing to eliminate the expression of plant specific xylosyl and fucosyl transferase genes Temporal enhancement of B-cell depletion in cynomolgus monkeys in comparison to fucosylated rituximab during the first 72hrs at low doses Gasdaska et al. 101
Chimeric IgG1          
Obinutuzumab/ GA101 CD20 Reduced (<30%) Coexpression with GnT III and α-ManII in CHO cells (GlycoMAb Technology) Enhanced tumor inhibition of GA101 compared with rituximab in human lymphoma xenograft mouse models Mossner et al. 102
Humanized IgG1       Enhanced B-cell depletion in spleen and lymph nodes of cynomolgus monkeys over rituximab Dalle et al. 131
        Enhanced tumor growth inhibition over rituximab in human-transformed follicular lymphoma RL model in SCID mice.  
Rituximab CD20 afucosylated Commercial rituximab treated with endoglycosidase/fucosidase to generate GlcNAc-rituximab Enhanced depletion of huCD20+ B cells in an FcγR-humanized mouse model over original rituximab Li et al. 70
Chimeric IgG1          
GBR 401 CD19 Reduced (∼50%) CHO cells with reduced fucosylation Enhanced B-cell depletion over rituximab in xenografted SCID mouse Breton et al. 107
Humanized IgG1          
Inebilizumab/MEDI-551 CD19 afucosylated FUT8−/− CHO cells (Potelligent® Technology) Enhanced B-cell depletion in huCD19/CD20 transgenic mouse over rituximab Herbst et al. 105
Humanized IgG1       A direct comparison between MEDI-551 and the fucosylated anti-CD19 in CD19+ Raji and Daudi cells lymphoma xenograft SCID mouse model only showed minor or insignificant improvement in tumor inhibition respectively Ward et al. 104
        Prolonged animal survival in SCID mice engrafted with human pre-B cells over afucosylated control IgG1 Matlawska-Wasowska et al. 106
        Enhanced B-cell depletion over wild type counterpart in human CD19 transgenic mouse at lower doses Gallagher et al. 129
MDX-1342 CD19 afucosylated FUT8−/− CHO cells (Potelligent® Technology) Dose-dependent enhancement of survival in murine B-cell lymphoma model with Ramos cells Cardarelli et al. 103
Human IgG1       Enhanced B-cell depletion over fucosylated counterpart in cynomolgus monkeys  
Imgatuzumab /GA201/ RG7160 EGFR Reduced (∼15%) Coexpression with GnT III and α-ManII in CHO cells (GlycoMAb Technology) Enhanced survival rate over fucosylated counterpart in mouse xenograft models displaying murine FcγRIV and over commercial Cetuximab in mouse xenograft models displaying murine FcγRIV and/or human FcγRIIIA. Gerdes et al. 108
Humanized IgG1          
ARGX-111 c-MET afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Potent inhibition of c-Met-amplified tumor growth in MKN-45 xenograft mice (a)
Human IgG1          
XGFR IGF-1R and EGFR Reduced Coexpression with GnT III and α-ManII in CHO cells (GlycoMAb Technology) Improved survival rate of mice treated with XGFR over its fucosylated counterpart in intrasplenic colon carcinoma model in SCID beige mice Schanzer et al. 110
Bispecific antibody with EGFR (GA201) and IGF-1R (R1507) specificities          
XGFR* IGF-1R and EGFR Reduced Coexpression with GnT III and α-ManII in CHO cells (GlycoMAb Technology) Enhanced tumor inhibition over fucosylated bispecific orthotopic Mia-PaCa2 pancreatic cancer xenograft model in SCID mice Schanzer et al. 130
Bispecific antibody with EGFR (GA201) and affinity-matured IGF-1R (R1507) – F13B5 specificities          
JNJ-61186372 EGFR and c-Met Reduced (<10%) CHO cells with low level of fucose Enhanced xenograft tumor inhibition in nude mouse over control and afucosylated IgG2 isotype Grugan et al. 109
Bispecific human IgG1          
Ifabotuzumab/KB004/IIIA4 EPHA3 afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Enhanced tumor growth inhibition over control antibody in DU145 or 22Rv1 xenograft mice Vail et al. 132
Humanized IgG1          
Bemarituzumab /FPA144 FGFR2b afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Enhanced tumour growth inhibition in FGFR2-overexpressing gastric cancer xenograft mouse model over isotype control (b)
Humanized IgG1          
Afucosylated Trastuzumab HER2 afucosylated FUT8−/− CHO cells (Potelligent® Technology) Enhanced tumor growth inhibition over fucosylated trastuzumab in KPL-4 xenografts in human FcγRIIIα mice Junttila et al. 133
Humanized IgG1          
TrasGEX/ GT-MAB7.3-GEX/Glycooptimized Trastuzumab-GEX HER2 Reduced Human glycoengineered production cell lines (GlycoExpress technology) Potent Her2+ BT474 tumor growth inhibition in nude mice but exhibited similar tumor volume and number of regression between fucosylated and reduced fucosylated antibodies (c)
Humanized IgG1          
Lumretuzumab /RG7116/RO5479599/ GE-HuMAb-HER3 HER3 Reduced Coexpression with GnT-III and α-ManII in CHO cells (GlycoMAb Technology) Enhanced survival of SCID-beige mice with A549-B34 NSCLC xenografts Mirschberger et al.134
Humanized IgG1       Enhanced tumor inhibition in A549 orthotopic mouse models over WT-huMab-HER3  
Palivizumab-N Respiratory syncytial virus (RSV) afucosylated Transgenic N. benthamiana with RNAi to eliminate the expression of plant specific xylosyl and fucosyl transferase genes Enhanced RSV protection in rat models over fucosylated counterpart (d)
Humanized IgG1          
h-13F6 Heavily glycosylated mucin-like domain of EBOV glycoprotein (GP) afucosylated N. benthamiana plants with RNAi to eliminate the expression of plant specific xylosyl and fucosyl transferase genes Enhanced anti-viral protection over CHO cells-produced fucosylated counterpart Hiatt et al., 114 Zeitlin et al., 115
Chimeric IgG1          
LSEVh-LS-F Hexavalent fusion protein with IgG1 Fc CD4 and HIV-1 gp120-binding sites afucosylated GDP-fucose transporter −/− CHO cells Potent inhibition of SHIV infection in macaque models over PBS control Bardhi et al. 84
        Potent in vivo neutralization of an HIV-1 strain resistant to the broadly neutralizing antibodies VRC01 and 3BNC117 in humanized mouse  
KM2760 CC chemokine receptor 4 (CCR4) Reduced YB2/0 cells Enhanced anti-tumor activity over fucosylated counterpart in human PBMC-engrafted mouse model Niwa et al. 135
Chimeric IgG1       Significant anti-tumor activity in disseminated and non-disseminated Sezary syndrome (SS) and Mycosis fungoides (MF) mouse models Yano et al. 136
        Potent anti-tumor effect of KM2760 in NOG mice bearing adult T cell leukemia/lymphoma (ATLL) cells Ito et al. 137
Mogamulizumab/ POTELIGEO/ KM0761 CC chemokine receptor 4 (CCR4) afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Induced potent tumor growth inhibition and enhanced survival in ATLL tumor-bearing mice over vehicle control Ishii et al. 138
Humanized IgG1          
Benralizumab /MEDI-563Humanized IgG1 IL-5Rα afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Efficient eosinophils depletion in non-human primates Kolbeck et al. 123
KHK2823          
Humanized IgG1 IL-3Rα (CD123) afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Tumor growth inhibition of human AML cell line MOLM-13 grafted into nude rats compared to vehicle control (e)
        Significant depletion of IL-3Rα-positive cells in the peripheral blood of cynomolgus monkeys  
Low fucose Elotuzumab/ HuLuc63-LF Signaling Lymphocyte Activation Molecule (SLAMF7, also called CS1) Reduced YB2/0 cells Enhanced anti-tumor activity in OPM2 xenograft SCID mice model Hsi et al. 139
Humanized IgG1          
Afucosylated anti-CS1 SLAMF7/ CS1 afucosylated Pichia pastoris, which normally cannot produce GDP-fucose, is glycoengineered to eliminate fungal type glycans and to produce complex biantennary N-linked glycans Enhanced anti-tumor efficacy in SCID mice xenograft tumor model over HEK293 produced fucosylated anti-CS1 antibody Gomathinayagam et al. 140
Humanized IgG1          
AK002 Sialic acid immunoglobulin-like lectins 8 (Siglec-8) afucosylated FUT8–/– CHO cells (Potelligent® Technology) Information not available  
Humanized IgG1          
BMS-986218 T-cell receptor cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Information not available  
Human IgG1(f)          
Cusatuzumab/ARGX-110Humanized IgG1 CD70, a member of TNF receptor ligand family afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Fucosylated version shown potent tumor growth inhibition in Raji xenograft mice (g) Silence et al. 141
           
DS-5573a B7-H3, a member of B7 family afucosylated FUT8 −/− CHO cells (Potelligent® Technology) DS-5573a showed dose dependent and significant tumor inhibition in MDA-MB-231-bearing SCID mice Nagase-Zembutsu et al. 142
Humanized IgG1          
Gatipotuzumab/ PankoMab-GEX/ GT-MAB 2.5 GEX Tumor specific glycoepitope of Muc1 (TA-Muc1) Reduced Human glycoengineered production cell lines (GlycoExpress technology) Information not available  
Humanized IgG1          
GM102/ 3C23KHumanized IgG1 anti-mullerian Hormone Receptor II (AMHR2) Reduced YB2/0 cells (EMABling® version) Inhibited tumor growth in COV434-MISRII tumor bearing mice while 2C23K-FcKO (could not bind FcRIIIα) did not reduce tumor growth Estupina et al.143
           
GSK2831781/ IMP731 Lymphocyte activation gene (LAG)-3 afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Enhanced depletion of human LAG-3+ T cells in SCID mice over fucosylated antibody (h)
Humanized IgG1          
KHK4083 OX40 afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Information not available  
Human IgG1          
OBT357/ MEN1112/ OX-001/OX-357 Bst1/CD157 afucosylated FUT8 −/− CHO cells (Potelligent® Technology) Information not available  
Humanized IgG1          
SEA-CD40Humanized IgG1 CD40 afucosylated CHO cells. Use of modified sugars (fucosylation inhibitor, 2-fluorofucose) in culture media to inhibit fucosylation Enhanced immune stimulating ability over parental dacetuzumab in xenograft tumor models (i)
SEA-BCMAHumanized IgG1 BCMA (B-cell maturation antigen) afucosylated CHO cells. Use of modified sugars (fucosylation inhibitor, 2-fluorofucose) in culture media to inhibit fucosylation Enhanced survival of SCID mice with tumor compared to control antibody (j)
           
TRX518Humanized IgG1 Glucocorticoid-induced TNF receptor (GITR) aglycosylated (k) Information not available Information not available  

(a) Aftimos P. et al., A Phase I, first-in-human study of ARGX-111, a monoclonal antibody targeting c-Met in patients with solid tumors, ASCO Poster 2015.

(b) Abigael T. et al., FPA144: A therapeutic antibody for treating patients with gastric cancers bearing FGFR2 gene amplification, Proceedings: AACR Annual Meeting 2014.

(c) Goletz S. et al., Patent Application US 2015/0166664 A1.

(d) Bossenmaier et al. GE-huMab-HER3, a novel humanized, glycoengineered HER3 antibody with enhanced ADCC and superior preclinical in vitro and in vivo efficacy, Proceedings: AACR 103rd Annual Meeting 2012.

(e) Akiyama T. et al., First Preclinical Report of the Efficacy and PD Results of KHK2823, a Non-Fucosylated Fully Human Monoclonal Antibody Against IL-3Rα, Blood, 2015 126:1349.

(f) BMS-986218 is a glycoengineered version of Ipilimumab, which is a human IgG1.

(g) ARGX-110 showed enhanced ADCC over fucosylated version in vitro. But only the fucosylated antibody was tested in the animal model.

(h) Written in Hamblin P.A. et al., Anti-LAG-3 binding proteins, Patent application number WO2014140180 (A1) 2014.

(i) Gardai S.J. et al., SEA-CD40, a sugar engineered non-fucosylated anti-CD40 antibody with improved immune activating capabilities, Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015.

(j) Sussman et al., BCMA antibodies and use of same to treat cancer and immunological disorder, Patent application publication number US 2017/0233484.

(k) This antibody is mutated such that it does not contain the conserved Fc N-glycosylation site.

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 monkeys 101 and in human FcγR- and CD20-transgenic mice 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. 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.103-107 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. 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. 105 , 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.108-110 In addition to the anti-EGFR antibody imgatuzumab (GA201 or RG7160), bi-specific glycoengineered formats against two different receptors have also been developed. 109 , 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. 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. 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. 113 Antibodies against RSV, EBOV and HIV have been glycoengineered to become afucosylated to further improve their anti-viral activity. 84 , 114 , 115 Enhanced binding to the FcγR by these afucosylated antibodies was correlated with enhanced efficacy in murine models. 114 , 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. 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 (Table 2, 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.

Antibody name and company Target and format Conditions Current updates
Obinutuzumab/ GA101/ Gazyva (a) CD20 Chronic lymphocytic leukemia ; Non-Hodgkin's lymphoma Marketed
Roche Humanized IgG1 with low fucose content Various forms of B-cell associated lymphomas Phase 1, 2, 3(b)
    Kidney Failure, Chronic Phase 1
    Lupus Nephritis Phase 2
Mogamulizumab/ CC chemokine receptor 4(CCR4) Humanized afucosylated IgG1 Relapsed or refractory CCR4-positive adult T-cell leukemia-lymphoma; Cutaneous T cell lymphoma ; Peripheral T-cell lymphoma Marketed
POTELIGEO/KM0761(a)   Solid Tumors Phase 1
Kyowa Hakko Kirin   Advanced Solid Tumors Phase 1
    Gastric Cancer, Esophageal Cancer, Lung Cancer, Renal Cancer Phase 1
    Solid Tumor, Cancer, Carcinoma Phase 1
    Diffuse Large B-Cell Lymphoma, Recurrent and/ Refractory Hodgkin Lymphoma, Recurrent and/ Refractory Non-Hodgkin Lymphoma Phase 1 and 2
    Solid Tumor Cancer, Carcinoma, Hepatocellular Carcinoma Phase 1 and 2
    Advanced Solid Tumors, Metastatic Solid Tumors Phase 1 and 2
    Adult T-cell Leukemia-Lymphoma Phase 2
    Cutaneous T-Cell Lymphoma Phase 3
    HTLV-1 Associated Myelopathy Phase 3
Benralizumab/MEDI-563/ Fasenra(a) IL-5Rα Asthma Marketed
AstraZeneca Humanized afucosylated IgG1 Eosinophilic Gastritis or Gastroenteritis Phase 1 and 2
    Hypereosinophilic Syndrome Phase 2
    Eosinophilic Chronic Rhinosinusitis Phase 2
    Asthma Phase 2, 3(c)
    Chronic Rhinosinusitis (Diagnosis), Nasal Polyps, Eosinophilia Phase 2
    Moderate to Very Severe Chronic Obstructive Pulmonary Disease Phase 3
    Nasal Polyposis Phase 3
    Severe Prednisone Dependent Eosinophilic Asthma Phase 3
    Chronic Idiopathic Urticaria Phase 4
Inebilizumab/ MEDI-551 CD19 Scleroderma Phase 1 (Completed )
MedImmune Humanized afucosylated IgG1 Diffuse large B cell lymphoma Phase 2 (Completed)
    Blood Cancer, Advanced B Cell Malignancies Phase 1 and 2 (Completed)
    Multiple Sclerosis, Relapsing Forms Phase 1 (Completed)
    Chronic lymphocytic leukemia Phase 2 (Completed)
    Relapsed/Refractory Aggressive B-cell Lymphomas Phase 1 and 2 (Completed)
    B-cell Malignancies Phase 1
    Neuromyelitis Optica and Neuromyelitis Optica Spectrum Disorders Phase 2 and 3
    Early Myeloma Phase 1
    Multiple Myeloma Phase 2
Ublituximab/ TG1101/ LFB-R603 CD20 Various forms of B-cell associated lymphomas Phase 1 and 2 (Completed)
TG Therapeutics Inc Chimeric IgG1, low fucose content Chronic Lymphocytic Leukemia Phase 1 (Completed)
    Chronic Lymphocytic Leukemia, Non-Hodgkin's Lymphoma Phase 1, 2(c)
    Non-Hodgkin Lymphoma, B-cell Lymphoma, Waldenstrom's Macroglobulinemia, Marginal Zone Lymphoma, Chronic Lymphocytic Leukemia, Small Lymphocytic Lymphoma, Primary Central Nervous System Lymphoma Phase 1
    Neuromyelitis Optica, Neuromyelitis Optica Spectrum Disorder Phase 1
    Chronic Lymphocytic Leukemia, Mantle Cell Lymphoma Phase 1 and 2
    Multiple Sclerosis Phase 2
    Diffuse Large B-Cell, Lymphoma Follicular Lymphoma, Marginal Zone Lymphoma, Small Lymphocytic Lymphoma Phase 2 and 3
    Chronic Lymphocytic Leukemia Phase1, 2, 3(c)
    Relapsing Multiple Sclerosis (RMS) Phase 2, 3(c)
Tomuzotuximab / CetuGEX™/ GT-MAB5.2 GEX EGFR Solid Tumors Phase 1
Glycotope Chimeric glyco-optimized(reduced fucosylated) IgG1 Carcinoma, Squamous Cell of Head and Neck Phase 2
Ifabotuzumab/KB004/IIIA4 EPHA3 Glioblastoma Phase 1
Humanigen Humanized afucosylated IgG1 Myelodysplastic Syndrome (MDS) Phase 1 (Suspended)
    Myelofibrosis (MF) Phase 2 (Suspended)
Bemarituzumab/ FPA144 FGFR2b Gastrointestinal Cancer, Metastatic Gastric Cancer Phase 1
Five Prime Therapeutics Humanized afucosylated IgG1 Transitional Cell Carcinoma of the Genitourinary Tract (Bladder Cancer) Phase 1
TrasGEX/ GT-MAB7.3-GEX/Glycooptimized Trastuzumab-GEX HER2 Solid Tumors Phase 1 (Completed)
Glycotope Humanized glyco-optimized (reduced fucosylated) IgG1    
Lumretuzumab /RG7116 /RO5479599/ GE-HuMAb-HER3 HER3 Neoplasms Phase 1 (Completed)
Roche Humanized reduced fucosylated IgG1 Squamous Non-Small Cell Lung Cancer Phase1 and 2 (Terminated)
    Breast Cancer Phase 1 (Completed)
GSK2849330 HER3 Cancer (Dose escalation study) Phase 1 (Completed)
GlaxoSmithKline Humanized afucosylated IgG1/3 Cancer (Immuno Positron Emission Tomography study) Phase 1 (Completed)
ARGX-111 c-MET Cancer Phase 1 (Completed)
Argenx Human afucosylated IgG1    
Roledumab/ LFB R593 Rhesus (Rh)D Rh disease Phase 2 and 3
LFB Biotechnologies Human IgG1 with low fucose content    
MEDI-570 ICOS Systemic lupus erythematosus Phase 1 (Discontinued)
MedImmune Human afucosylated IgG1 Various stages and grades of T cell lymphoma Phase 1
Cusatuzumab/ ARGX-110 CD70, a member of TNF receptor ligand family Acute Myeloid Leukemia, High Risk Myelodysplastic Syndrome Phase 1 and 2
Argenx Humanized afucosylated IgG1 Cancer Phase 1
    Advanced Cancer Phase 1 and 2
AK002 Sialic acid immunoglobulin-like lectins (Siglec)-8 Healthy Phase 1 (Completed)
Allakos Humanized afucosylated IgG1 Indolent Systemic Mastocytosis Phase 1
    Atopic Keratoconjunctivitis, Vernal Keratoconjunctivitis, Phase 1
    Perennial Allergic Conjunctivitis  
    Chronic Urticaria Actinic Keratosis Phase 2 Phase 2
BMS-986218 T-cell receptor cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) Advanced Cancer Phase 1 and 2
Bristol Myers Squibb Afucosylated IgG1    
GM102/ 3C23K anti-mullerian Hormone Receptor II (AMHR2) Neoplasm, Gynecologic Phase 1
GamaMabs Pharma Humanized reduced fucosylated IgG1    
GSK2831781/ IMP731 Lymphocyte activation gene (LAG) – 3 Psoriasis Phase 1
GlaxoSmithKline Humanized afucosylated IgG1    
Gatipotuzumab/PankoMab-GEX/ GT-MAB 2.5 GEX Tumor specific glycoepitope of Muc1 (TA-Muc1) Solid Tumors Phase 1
Glycotope Humanized glyco-optimized (reduced fucosylated) IgG1 Ovarian Epithelial Cancer, Recurrent Fallopian Tube Cancer, Primary Peritoneal Cancer Phase 2
       
OBT357/MEN1112/OX-001/OX-357 Bst1/CD157 Recurrent Adult Acute Myeloid Leukemia, Acute Myeloid Leukemia, in Relapse Phase 1
Oxford BioTherapeutics, in collaboration with Menarini Humanized afucosylated IgG1    
SEA-BCMA BCMA (B-cell maturation antigen) Multiple Myeloma, Multiple Myeloma in Relapse, Phase 1
Unum Therapeutics and Seattle Genetics Humanized afucosylated IgG1 Refractory Multiple Myeloma  
SEA-CD40 CD40 Cancer and carcinomas Phase 1
Seattle Genetics Humanized afucosylated IgG1    
KHK2823 IL-3Rα / CD123 Acute Myeloid Leukemia, Myelodysplastic Syndrome Phase 1
Kyowa Hakko Kirin Pharma Humanized afucosylated IgG1    
KHK4083 OX40 Dermatitis, Atopic Phase 1
Kyowa Hakko Kirin Pharma Humanized afucosylated IgG1 Healthy Men and Subjects With Ulcerative Colitis Phase 1
    Ulcerative Colitis, Digestive System Diseases, Colitis, Gastrointestinal Diseases, Inflammatory Bowel Diseases, Phase 2
     Intestinal Diseases, Colonic Diseases, Autoimmune Disease, Abdominal Pain  
TRX518 Glucocorticoid-induced TNF receptor (GITR) Unresectable Stage III or Stage IV Malignant Melanoma or Other Solid Tumor Malignancies Phase 1
Leap Therapeutics Humanized aglycosylated IgG1(d) Solid tumors Phase 1

(a) Only ongoing clinical trials are listed for antibodies that are already on the market

(b) GA101 is being tested in numerous clinical trials for various forms of B-cell associated cancers

(c) Different clinical trials identifiers

(d) This antibody is mutated such that it does not contain the conserved Fc N-glycosylation site

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. 102 Results from Phase1b/2 trials indicated that all patients with CLL experienced rapid and sustained removal of B cells in the peripheral blood.116-119 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. 120 In addition, this combination resulted in substantially increased time to next treatment. 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. 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. 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. 124 , 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. 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. 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. 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 (Table 1), 26 of them have been studied in clinical trials and 3 have been approved for use in clinical practice (Table 2). We expect that more afucosylated antibodies will enter clinical trials and subsequently be approved for clinical use.

Funding Statement

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

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.

References

  • 1.Ecker DM, Jones SD, Levine HL. The therapeutic monoclonal antibody market. MAbs. 2015;7:9–14. doi: 10.4161/19420862.2015.989042. PMID:25529996 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Reichert JM. Antibodies to watch in 2015. MAbs. 2015;7:1–8. doi: 10.4161/19420862.2015.988944. PMID:25484055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Reichert JM. Antibodies to watch in 2016. MAbs. 2016;8:197–204. doi: 10.1080/19420862.2015.1125583. PMID:26651519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Reichert JM. Antibodies to watch in 2017. MAbs. 2017;9:167–81. doi: 10.1080/19420862.2016.1269580. PMID:27960628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.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. [DOI] [PubMed] [Google Scholar]
  • 6.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. [DOI] [PubMed] [Google Scholar]
  • 7.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. [DOI] [PubMed] [Google Scholar]
  • 8.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. [DOI] [PubMed] [Google Scholar]
  • 9.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. [DOI] [PubMed] [Google Scholar]
  • 10.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. [DOI] [PubMed] [Google Scholar]
  • 11.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.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. [DOI] [PubMed] [Google Scholar]
  • 13.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. [DOI] [PubMed] [Google Scholar]
  • 14.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. [DOI] [PubMed] [Google Scholar]
  • 15.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. [DOI] [PubMed] [Google Scholar]
  • 16.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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. [PubMed] [Google Scholar]
  • 19.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.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. [DOI] [PubMed] [Google Scholar]
  • 21.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.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. [DOI] [PubMed] [Google Scholar]
  • 23.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.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Becker DJ, Lowe JB. Fucose: biosynthesis and biological function in mammals. Glycobiology. 2003;13:41R–53R. doi: 10.1093/glycob/cwg054. PMID:12651883. [DOI] [PubMed] [Google Scholar]
  • 25.Scharberg EA, Olsen C, Bugert P. The H blood group system. Immunohematology. 2016;32:112–118. PMID:27834485. [PubMed] [Google Scholar]
  • 26.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. [DOI] [PubMed] [Google Scholar]
  • 27.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. [DOI] [PubMed] [Google Scholar]
  • 28.Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinical implications. Nat Rev Cancer. 2015;15:540–55. doi: 10.1038/nrc3982. PMID:26289314. [DOI] [PubMed] [Google Scholar]
  • 29.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.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hauselmann I, Borsig L. Altered tumor-cell glycosylation promotes metastasis. Frontiers Oncol. 2014;4:28. doi: 10.3389/fonc.2014.00028.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.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.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.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. [PubMed] [Google Scholar]
  • 33.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. [DOI] [PubMed] [Google Scholar]
  • 34.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. [DOI] [PubMed] [Google Scholar]
  • 35.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. [DOI] [PubMed] [Google Scholar]
  • 36.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. [DOI] [PubMed] [Google Scholar]
  • 37.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. [DOI] [PubMed] [Google Scholar]
  • 38.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. [DOI] [PubMed] [Google Scholar]
  • 39.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. [DOI] [PubMed] [Google Scholar]
  • 40.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. [DOI] [PubMed] [Google Scholar]
  • 41.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. [DOI] [PubMed] [Google Scholar]
  • 42.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. [PubMed] [Google Scholar]
  • 43.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. [PubMed] [Google Scholar]
  • 44.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. [DOI] [PubMed] [Google Scholar]
  • 45.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. [PubMed] [Google Scholar]
  • 46.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.. [DOI] [PubMed] [Google Scholar]
  • 47.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.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. [DOI] [PubMed] [Google Scholar]
  • 50.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. [DOI] [PubMed] [Google Scholar]
  • 51.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. [DOI] [PubMed] [Google Scholar]
  • 52.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. [DOI] [PubMed] [Google Scholar]
  • 53.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. [DOI] [PubMed] [Google Scholar]
  • 54.Stanley P. Regulation of Notch signaling by glycosylation. Curr Opin Structural Biol. 2007;17:530–5. doi: 10.1016/j.sbi.2007.09.007.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.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. [DOI] [PubMed] [Google Scholar]
  • 56.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. [DOI] [PubMed] [Google Scholar]
  • 57.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. [DOI] [PubMed] [Google Scholar]
  • 58.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.. [DOI] [PubMed] [Google Scholar]
  • 59.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. [DOI] [PubMed] [Google Scholar]
  • 60.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. [DOI] [PubMed] [Google Scholar]
  • 61.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. [DOI] [PubMed] [Google Scholar]
  • 62.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. [DOI] [PubMed] [Google Scholar]
  • 63.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. [DOI] [PubMed] [Google Scholar]
  • 64.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.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. [DOI] [PubMed] [Google Scholar]
  • 67.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.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. [DOI] [PubMed] [Google Scholar]
  • 69.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.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. [DOI] [PubMed] [Google Scholar]
  • 73.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. [DOI] [PubMed] [Google Scholar]
  • 74.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.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. [DOI] [PubMed] [Google Scholar]
  • 76.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. [DOI] [PubMed] [Google Scholar]
  • 77.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. [DOI] [PubMed] [Google Scholar]
  • 78.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. [DOI] [PubMed] [Google Scholar]
  • 79.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. [DOI] [PubMed] [Google Scholar]
  • 80.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. [DOI] [PubMed] [Google Scholar]
  • 81.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. [DOI] [PubMed] [Google Scholar]
  • 82.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.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. [PubMed] [Google Scholar]
  • 84.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.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. [DOI] [PubMed] [Google Scholar]
  • 87.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. [DOI] [PubMed] [Google Scholar]
  • 88.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. [DOI] [PubMed] [Google Scholar]
  • 89.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Loos A, Steinkellner H. IgG-Fc glycoengineering in non-mammalian expression hosts. Arch Biochem Biophys. 2012;526:167–73. PMID:22634260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.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. [PubMed] [Google Scholar]
  • 92.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.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. [DOI] [PubMed] [Google Scholar]
  • 94.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. [DOI] [PubMed] [Google Scholar]
  • 95.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. [DOI] [PubMed] [Google Scholar]
  • 96.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. [DOI] [PubMed] [Google Scholar]
  • 97.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. [DOI] [PubMed] [Google Scholar]
  • 98.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. [DOI] [PubMed] [Google Scholar]
  • 99.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.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. [DOI] [PubMed] [Google Scholar]
  • 102.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.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. [DOI] [PubMed] [Google Scholar]
  • 105.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. [DOI] [PubMed] [Google Scholar]
  • 106.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.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. [DOI] [PubMed] [Google Scholar]
  • 109.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Feldmann H, Geisbert TW. Ebola haemorrhagic fever. Lancet. 2011;377:849–62. PMID:21084112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Stephenson KE, Barouch DH. Broadly Neutralizing Antibodies for HIV Eradication. Current HIV/AIDS Reports. 2016;13:31–7. PMID:26841901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.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. [DOI] [PubMed] [Google Scholar]
  • 117.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. [DOI] [PubMed] [Google Scholar]
  • 118.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. [DOI] [PubMed] [Google Scholar]
  • 119.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. [DOI] [PubMed] [Google Scholar]
  • 120.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. [DOI] [PubMed] [Google Scholar]
  • 121.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. [DOI] [PubMed] [Google Scholar]
  • 122.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.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. [DOI] [PubMed] [Google Scholar]
  • 124.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. [DOI] [PubMed] [Google Scholar]
  • 125.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. [DOI] [PubMed] [Google Scholar]
  • 126.Beck A, Reichert JM. Marketing approval of mogamulizumab: a triumph for glyco-engineering. MAbs. 2012;4:419–25. PMID:22699226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.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. [DOI] [PubMed] [Google Scholar]
  • 130.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.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. [DOI] [PubMed] [Google Scholar]
  • 132.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. [DOI] [PubMed] [Google Scholar]
  • 133.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. [DOI] [PubMed] [Google Scholar]
  • 134.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. [DOI] [PubMed] [Google Scholar]
  • 135.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. [DOI] [PubMed] [Google Scholar]
  • 136.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. [DOI] [PubMed] [Google Scholar]
  • 137.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. [DOI] [PubMed] [Google Scholar]
  • 138.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. [DOI] [PubMed] [Google Scholar]
  • 139.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.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. [DOI] [PubMed] [Google Scholar]
  • 141.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.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. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from mAbs are provided here courtesy of Taylor & Francis

RESOURCES