SlideShare a Scribd company logo
1 of 33
Download to read offline
Glycosylation for
biopharmaceutical drugs
Ricky Connolly
Biotechnology, Dublin City University; email: ricky.connolly2@mail.dcu.ie




Abstract
Since the mid 2000s, the patents for many blockbuster drugs have begun to expire. This
is the driving force behind the current great development in the biopharmaceutical
industry, the ‘second generation’ biopharmaceuticals. This new trend is focussed on
modifying existing protein therapeutics in order to enhance their pharmacological,
biological, and structural properties. These modifications include but are not limited to:
generation of fusion conjugates, incorporation of chemical modifications such as
pegylation, and, crucially for this paper, modification of glycosylation profiles. The goal
of this paper is to outline the chemical and biological basis of protein glycosylation, to
examine the pharmacological and structural implications of glycosylation, to compare
and evaluate the current production strategies, and to survey the current range of
analytical methods available to characterise glycoprotein therapeutics.
Table of Contents
Introduction ................................................................................................................................. 3
   Overview ...................................................................................................................................... 3
   Glycan synthesis ........................................................................................................................... 4
Stability .......................................................................................................................................... 6
   Aggregation .................................................................................................................................. 6
   Crosslinking ................................................................................................................................. 7
   Proteolysis .................................................................................................................................... 7
   Oxidation ..................................................................................................................................... 7
   pH ................................................................................................................................................ 7
   Kinetic Denaturation ................................................................................................................... 8
   Chemical Denaturation ............................................................................................................... 8
   Temperature ................................................................................................................................ 8
Pharmacology .............................................................................................................................. 9
   Receptor Binding ......................................................................................................................... 9
   Circulatory lifetime .................................................................................................................... 10
   Bioavailability ............................................................................................................................. 10
   Distribution................................................................................................................................ 11
   Clearance rates ........................................................................................................................... 12
   Antibody function ...................................................................................................................... 12
   Immunogenicity ......................................................................................................................... 12
Glycosylation and host cells .................................................................................................. 13
   Mammalian Cells ....................................................................................................................... 14
   Yeasts.......................................................................................................................................... 15
   Bacteria....................................................................................................................................... 16
Analytical methods ................................................................................................................... 18
   Mass spectrometric .................................................................................................................... 18
   Chromatographic ....................................................................................................................... 19
   Electrophoretic .......................................................................................................................... 20
   Bioaffinity methods .................................................................................................................... 20
Bioinformatics ........................................................................................................................... 22
   Complexity of glycan structures ................................................................................................ 22
   Prediction tools .......................................................................................................................... 22
   Glycobiology databases .............................................................................................................. 23
References................................................................................................................................... 24



 Abstract | DCU
Introduction
The human genome contains at least 30,000 protein-coding genes, and through
alternative mRNA splicing, these give rise to over 100,000 proteins (Venter, 2001). The
diversity of the human proteome is further
magnified by post-translational modification of
proteins. At least 50 percent of mammalian
proteins are glycosylated (Zafar, 2011). More
than two thirds of the 200 or so biopharma-
ceutical products licenced for sale in the
United States and European Union are
recombinant human proteins (Li, 2010). In
2010, five of the top ten selling pharmaceutical
products were recombinant human proteins
and this is projected to rise to eight of the top
ten by 2014 (Hirschler, 2010). Additionally,
over 70 percent of the therapeutics currently in
clinical trials are glycosylated human proteins (Sethuraman, 2006).




Overview
Glycosylation is defined as the covalent attachment of oligosaccharide moieties (glycans)
to the side chains of amino acid residues of proteins. There are at least five different
classes of glycosylation, each defined by
the amino acid the glycan in linked to and
the type of linkage used. By far the most
common of these are N-linked and O-
linked glycosylation (Apweilier, 1999).
This review will focus solely on N-linked
glycosylation because it is more common
type found in human therapeutic protein
therapeutics (Pandhal, 2010). N-linked
glycosylation involves the enzymatic attachment of the N-Acetylglucosamine residue at
the terminal end of the glycan to the amide group of an asparagine residue by means of a
β1 glycosidic linkage (Geyer, 2006).



                                                                       DCU | Introduction
The asparagine must be located within the three residue sequence Asparagine-X-
Serine/Threonine, where X represents any amino acid except proline, which inhibits
attachment of the N-glycan through stearic hindrance caused by its rotationally-locked
side group (Shyama, 2010). There are further structural requirements in addition to the
Asp-X-Ser/Thr sequon, mostly concerning the three-dimensional localisation and
accessibility of the sequon within the folded protein (Apweilier, 1999).

Glycan synthesis
N-glycosylation takes place in the endoplasmic reticulum, at the same sites as protein
translation. The first step in glycan synthesis involves the addition of two GlcNAc
residues and five mannose residues to the membrane-anchored dolichol-PP. The
dolichol-PP-Man5GlcNAc2 group is then flipped to the inner membrane side of the
endoplasmic reticulum by the enzyme RFT1 (Pandhal, 2010).

The core oligosaccharide (Glc3Man9GlcNAc2) is constructed on the GlcNAc residue by
a series of glycosyltransferases. After this, the glycan is transferred to a sequon-labelled
asparagine residue on the protein chain as it emerges from the ribosome (Pandhal, 2010).




 Introduction | DCU
The glycan is trimmed by glucosidase I and II to remove the α1,2-linked (Shailubhai,
1987) and α1,3-linked (Saxena, 1987) glucose residues, respectively. This leaves us with a
Man9GlcNAc2 structure. Next, α1,2-mannosidase removes one of the α1,2-linked
mannose residues. The Man8GlcNAc2 glycan is then exported to the Golgi apparatus.
Here,     more   α1,2-mannosidases       remove   several       mannose    residues,   producing
Man5GlcNAc2. After this, the glycosylation pathway diverges into different patterns of
residue    trimming    and   addition,    catalysed   by    a    series   of   glycosidases   and
glycosyltransferases (Pandhal, 2010).

This results in three distinct prototypic classes of glycan structure into which the
majority of glycans fall: complex, hybrid, and oligomannose (Kornfeld, 1985).
Oligomannose glycans contain only mannose residues in addition to their core structures.
Hybrid glycans can contain a diverse range of residues including galactose, glucoronic
acid, and xylose (in plants). Complex glycans resemble hybrid glycans but they come
with varying degrees of fucosylation and sialylation (Pandhal, 2010). Sialylation is often
vital for correct protein function, this will be discussed later.




Glycans are constructed from monosaccharide units linked together with glycosidic
bonds. Unlike peptides and nucleic acids, one monosaccharide unit can be linked to
multiple other units. This is called branching. Biantennary glycans are the most common,
but triantennary and tetrantennary glycans are not unheard of (Campion, 1989). Another
common structural feature is to have a single (β1,4) GlcNAc residue linked to the first
branching point mannose unit. This bisecting GlcNAc residue is often involved in
signalling (Yoshimura, 1998). The molecular structures, branching types and families of


                                                                               DCU | Introduction
glycans attached to a protein describe its microheterogeneity in terms of glycosylation.
This is distinct from the macroheterogeneity, which is described by the total number of
glycans attached to the protein and their positions.




Stability
A protein instability is defined as a physical or chemical vulnerability that is prone to
altering the structural conformation or activity in a detrimental way. Although they have
high therapeutic efficacy, the main drawback to pharmaceutical use of proteins is their
inherent structural and chemical instabilities. These impose limitations on the protein
production and purification stages, product formulation, storage and transport, and half-
life in the body. Glycosylation has been shown to ameliorate many of these instabilities.
In this section, the major protein instabilities will be outlined, and the efficacy of
glycosylation on these will be investigated.

Aggregation
Proteins are very complex molecules with hundreds if not thousands of exposed
functional groups. This makes non-native aggregation a very common problem for
protein biopharmaceuticals. Aggregation can be caused by relatively small changes in the
protein structure (Saluja, 2008). Aggregation can drastically reduce the biological
function of a protein (Philo, 2009) and in some cases can induce an immunogenic
reaction in patients (Rosenberg, 2006). Glycosylation has been shown to prevent the
formation of protein aggregates (Kayser, 2011). The likely explanation is that the bulky
spatial nature of glycan moieties provides a level of stearic repulsion between proteins
(Imperiali, 1999), reducing the likelihood of aggregation (Hoiberg-Nielsen, 2006).

Precipitation
Precipitation is the ultimate effect of protein aggregation. Eventually, the proteins form
aggregates so large that they become insoluble (Chi, 2003). Precipitation is a major
problem for biotherapeutic product formulation because it is desirable to store protein
solutions at relatively high concentrations, but at high enough concentrations most
proteins will form aggregates and precipitate out of solution (Wang, 2005).
Glycosylation decreases the propensity to form insoluble precipitates for many proteins
(Kayser, 2011). It has been shown that the decrease in insolubility is directly

 Stability | DCU
proportional to the size and number of glycans present (Tams, 1999). One possible
explanation for this phenomenon is that glycans are more soluble than peptides, and they
confer some of this solubility to the glycoprotein as a whole.

Crosslinking
Crosslinking, also known as polymerisation-induced inactivation, occurs primarily when
the methionine residues of two different proteins form a disulphide linkage. This leads
to the formation of protein oligomers which have diminished biological activity (Wang,
1999). Glycosylation has been shown to prevent chemical crosslinking between proteins
(Runkel, 1998). The mechanism is probably the same as that of aggregation, the mutual
stearic repulsion between glycosylated proteins.

Proteolysis
Proteolysis is the degradation of the peptide backbone through hydrolytic reactions.
These reactions are carried out by the ubiquitous protease enzymes found in all tissues.
They are a major problem from a drug administration perspective because protease-
sensitivity can drastically reduce the level of therapeutic that reaches the site of action
(Tang, 2004). Glycosylation has been shown to inhibit proteolytic degradation (Vegarud,
1975). The most likely mechanism is through stearic blockage of the protease cleavage
sites by the glycan (Russell, 2009).

Oxidation
Proteins are susceptible to oxidative degradation at the level of their primary structure.
This is of particular concern for proteins rich in the more reactive amino acids such as
His, Met, Cys, Tyr and Trp (Manning, 2010), which are prone to accepting free radicals.
Since free radicals are basically unavoidable at all stages of the pipeline from cell culture
to drug administration (even exposure to stray light can be harmful (Kerwin, 2007)),
oxidation poses a serious threat to the stability and functionality of therapeutic proteins.
Glycosylation has been shown to reduce the effects of oxidative damage in at least one
commercial therapeutic (Uchida, 1997). One proposed mechanism for this protection is
that the glycans are ‘soaking up’ the radicals, thus preventing them from reaching the
protein itself (Pristov, 2011).

pH
Proteins are only stable within a limited pH range. pH denaturation begins when the ion
balance disrupts the hydrogen- and ion-bonding capacity of the amino acids. This affects
the tertiary structure of the protein and leads to the formation of non-native chemical

                                                                              DCU | Stability
bonds as the peptide reconfigures into a more thermodynamically-stable state (Solá,
2009). Glycosylation has been shown to improve the pH stability of protein therapeutics
by up to a 13-fold increase (Masarova, 2001). This marked improvement is due to the
fact that attached glycans decrease the solvent accessible surface area of the protein,
acting as a molecular buffer between the electrostatic forces of the solvent and those of
the protein.

Kinetic Denaturation
Due to their complex nature, protein molecules have several three-dimensional states in
which they are thermodynamically stable, but usually have only state in which they are
functionally active. This means that proteins can undergo kinetic inactivation even at
low temperatures by ‘flipping’ to another stable but inactive state (Arakawa, 2001). It has
been demonstrated that glycosylation increases the kinetic stability of proteins.
Specifically, the level of stability conferred seems to be correlated to the number of
glycans present, their positioning on the protein, and their size (Solá, 2007). This is most
likely because a folded, glycosylated protein has a lower free-energy profile compared to
both the unfolded protein and to the folded-unglycosylated protein (Shental-Bechor,
2008).

Chemical Denaturation
Chemical denaturation can be defined as the loss of structural integrity of a protein in
response to exposure to a chemical agent. One of the primary reasons for chemical
destabilisation is that the protein often has high Van der Waals affinity to the denaturant.
This allows the chemical molecule to intrude into the tertiary structure of the protein,
disrupting the global conformation and reducing functionality (Hual, 2008).
Glycosylation has been shown to promote conformational stability in opposition to
chemical denaturants (Sytkowski, 1991). The probable explanation for this effect is that
glycosylation essentially ‘compacts’ the protein, increasing the strength of its internal
ionic, hydrogen and Van der Waals bonds and reducing the peptide’s affinity for outside
chemical molecules (Solá, 2007).

Temperature
All of the bonds within a protein are sensitive to thermal fluctuations. Outside of a small
temperature range, these bonds will break or form non-native bonds, destroying the
biological activity of the protein (Vogt, 1997). The effects of freezing are less well
studied compared to those of heating, but some comprehensive studies of the
phenomena exist (Bhatnagar, 2007). Glycosylation has been shown to improve the

 Stability | DCU
thermal stability range for several therapeutically important proteins including EPO,
alpha 1-antitrypsin, interferon-β, and follicle-stimulating hormone (Solá, 2009). It is
likely that these increases in stability are due to the constraint of peptide mobility caused
by glycan attachment (Wormald, 1999). The magnitude of thermal stabilisation is
proportional to the number and size of the glycan attachments (Wang, 1996).




Pharmacology
Pharmacology       is   divided      into    two    fields   of    study:   pharmacokinetics      and
pharmacodynamics. Pharmacokinetics examines the action of drugs within the body over
a     given   period    of   time;    profiling    distribution,    metabolism,       and   excretion.
Pharmacodynamics studies the mechanisms of action of drugs within the body, studying
the     drug-receptor    interactions       and    dose/response     profiles.   In    other   words,
pharmacokinetics studies what the body does to the drug, while pharmacodynamics
studies what the drug does to the body (Benet, 1984).

Proteins tend to have poor pharmacokinetic profiles because they are very quickly
cleared by proteolytic degradation pathways, hepatic and renal elimination, and
receptor-mediated endocytosis (Tang, 2004). They have sharp pharmacodynamics
profiles due to their exceptionally high binding affinities with receptors compared to
small molecule drugs and have high turnover rates of their substrates. Glycosylation
strongly affects the pharmacological properties of a protein. In this section, the major
intrinsic pharmacokinetic and pharmacodynamic limitations of proteins are reviewed.
For each, the impact of glycosylation on these limitations are examined.

Receptor Binding
One of the most characteristic traits of proteins is the extraordinarily high affinity with
which they bind to their receptors. This is both as blessing and a curse. A high receptor
association rate allows the design of therapeutics with strong biological efficacy, but it
often means the protein has a very blunt therapeutic response curve (Solá, 2010). This
leads to dosage schemes that require multiple injections per day, with widely fluctuating
levels of drug in the body over the course of the day. For this reason, it is often desirable
to ‘smooth out’ the response curve to a protein therapeutic. In practise, this means
reducing receptor affinity of the protein.



                                                                                 DCU | Pharmacology
Glycosylation has been used to reduce the receptor affinity for several commercially
important protein therapeutics. Darling et al showed that EPO-IRS (the standard form
of human erythropoietin recognized European Pharmacopoeia) showed a 20-fold lower
receptor association rate compared with artificially deglycosylated EPO (Darling, 2002).


Similarly, another study compared the relative receptor binding affinities of several
isoforms of erythropoietin. The EPO isoforms are defined by the total number of sialic
acid residues found on their glycocomponent. The experiment involved measuring the
quantity of each isoform needed to displace inactive EPO from receptors expressed on
the surface of human erythroleukemia cells. There was a direct inverse relationship
between sialic acid content and receptor binding affinity (Egrie, 2001).
The most likely mechanisms by which this reduction in binding affinity can be explained
is that electrostatic repulsion between the sialic acid residues of the glycans and the
receptor serve to decrease the liklihood of receptor binding (Elliott, 2004).

Circulatory lifetime
Glycosylation can have a dramatic effect on the circulatory lifetime of a therapeutic
protein. Comparing the serum half-life of the enzyme ceruloplasmin in it’s a natively-
glycosylated state with that of an artificially deglycosylated variant, the circulatory
lifetime of the deglycosylated variant was found to be an order of magnitude lower
(Morell, 1968). Similarly, the addition of extra glycans (hyperglycosylation) has been
shown to decrease the clearance rate of proteins.

Perlman et al (2003) observed a 4-fold increase in serum half-life of a variant of human
follicle stimulating hormone (FSH) with two addition N-linked glycosylation sites. The
sialic acid content of attached glycans has been shown to be of high importance to
extending the serum half-life of a protein. Sialic residues have a net negative charge at
biological pH. This electrostatic repulsion confers protection from both renal (Kanwar,
1984) and hepatic (Morell, 1971) clearence mechanisms.

Bioavailability
Today the vast majority of protein therapeutics are delivered by parenteral injection
(Soltero 2001). This contrasts with small molecule drugs, which are typically delivered
through oral routes (Nandita, 2003). From a clinical standpoint, it would be very
desirable to have oral-administered protein therapeutics. Unfortunately, there are several
barriers to making this technology a reality.



 Pharmacology | DCU
Unless injected directly into the bloodstream, drugs must pass through several
membrane barriers before they can begin systemic circulation, and if the target of the
drug is intracellular, the peptide must pass through the lipid membrane of the cell.
There are four mechanisms by which a protein therapeutic may pass through a
membrane: passive and facilitated diffusion, active transport, and receptor mediated
endocytosis (Kopacek, 2011). The rate at which a drug absorbs in the body is determined
by the rate at which the drug is transported across these barriers.

Glycosylation has been shown to improve protein absorption in several cases. Egleton et
al (2001) demonstrated that the addition of an O-linked glycan to an opioid peptide
icreased its blood brain barrier permeability from 1.0 μl/(min·g) to 2.2 μl/(min·g), with a
comparable increase in measured analgesic activity. Albert et al (1993) produced a
glycosylated, orally-active version of octreotide, a regulatory peptide that inhibits the
production of somatropin and other growth hormones, which had ten times greater oral
bioavailability compared to the parent molecule. Nomoto et al (1998) significantly
improved intestinal uptake of a peptide by the sodium ion-dependent D-glucose
transporter through the addition of a small glycan moiety.

Distribution
Drug distribution throughout the tissues of the body is controlled by blood perfusion,
plasma protein and tissue binding affinity, pH, and membrane permeability (Kopacek,
2011). Controlling the distribution of a drug is vital to ensuring that the drug reaches
the target tissue and that it does not end up in the wrong tissue, where it will have no (or
even adverse) biological effects.

Glycosylation has been shown to improve the tissue distribution of therapeutic drugs.
Sasayama et al (2000) chemically glycosylated human interleukin-1ɑ with an                 N-
acetylneuraminic acid moiety and monitored the in vivo tissue distribution in rats
following intraperitoneal (IP) injection. Up to five-fold greater levels of the glycosylated
variant were observed in the kidney, spleen, lung, and blood. Similarly, Ceaglio et al
(2008) created a mutant version of interferon-ɑ with four additional N-glycans which
displayed a ten-fold increase in distribution half-life (t1/2β) after ten hours post-injection,
and levels remained detectable 96 hours after injection. The most likely explanations for
the increase in tissue distribution are that the glycoproteins are protected from
proteolytic and immune-inactivation pathways or that the more soluble glycans reduce
the hydrophobicity of the peptide, increasing the rate of transport.


                                                                          DCU | Pharmacology
Clearance rates
Drug clearance studies measure the rate of removal of drug from circulation. This
mainly takes place in the liver and kidneys. The drugs are carried along by the flow of
blood until they reach the liver, where they are taken up by hepatocytes and are broken
down by nonspecific proteases, before the amino acids are recycled back into the body’s
metabolic cycle (Kahn, 2011). It is advantageous to attempt to reduce the clearance rates
of protein biopharmaceuticals to a minimum so that expensive drug is not wasted. Any
effective reduction of the level of clearance of a therapeutic will be valuable from a
clinical and economic point of view, and glycosylation has shown to be an efficient
strategy to achieve this reduction.

A novel hyperglycosylated variant of human EPO, darbepoetin alfa (DA), was
engineered to display two additional N-linked glycans. In a double-blind, randomized,
cross-over clinical trial in humans, DA showed a 2.5-fold lower rate of clearance (1.6
mL/h·kg versus 4.0 mL/h·kg) (Macdougall, 1999). The reduction in clearance rate
observed for hyperglycosylated proteins has been attributed to the increased sialic acid
content of the introduced glycans (Egrie, 1993).

Antibody function
IgG antibodies have two large biantennary N-linked carbohydrate moities attached to
the Fc effector region . The structure of these glycans has an effect on the chain
orientation, chain spacing and surface residue exposure (Kaneko, 2006). These
differences in structure can alter antibody effector function (Burton, 2006). It has been
shown that IgG antibodies engineered to remove fucose residues from their glycan
component have a much stronger antibody-dependent cell-mediated cytotoxicity profile
compared to wild-type IgGs (Yamane-Ohnuki, 2004). This is of clinical importance
because higher activation of immune system cells can stimulate the body to fight a wide
range of conditions, including cancer (Satoh, 2006).

Immunogenicity
Glycosylation has been shown to help prevent the generation of neutralizing antibodies
against therapeutic drugs. There are several theories which attempt to explain this effect.
One explanation is that, as outlined earlier, glycosylation inhibits the formation of
protein aggregates. Antibodies are often raised more efficiently against aggregates than
individual proteins, so it stands to reason that glycosylation prevents the development of
an immune reaction by inhibiting aggregation of the drug in circulation (Moore 1980).
Another possible explanation of the inhibition of immune response is that the bulky

 Pharmacology | DCU
nature of the carbohydrate moieties provide a degree of stearic hindrance, shielding the
peptide chain below from immune cells (Casadevall, 2002). A further theory is that the
terminal sialic acid residues of glycans provide a degree of electrostatic repulsion, again
preventing immune cell surface receptors from binding (Fernandes, 2002).




Glycosylation and host cells
It is estimated that well over 50 percent of human proteins are glycosylated (Apweiler,
1999), although this figure is disputed (Khoury, 2011). Nevertheless, almost 40 percent
of   all    approved       biopharmaceutical
                                                                 2%         55%                 a.
products     are    glycosylated        (Walsh,         2% 2%
2010). Therefore, for an organism to                                                  Mammalian
                                                  10%
become widely used as a producer
                                                                                      E. coli
strain, it must be able to perform
                                                                                      Yeast
glycosylation to some extent. Between
2006        and       2010,        58       new                                       Insect

biopharmaceuticals gained approval.                                                   Animals

32     of   these    were        produced    in   29%                                 Synthetic
mammalian cell lines and of these, 24
                                                                              CHO
were produced in Chinese hamster
                                                       3%   3%
                                                                   75%
                                                                                            b.
ovary (CHO) cells. It is clear that               6%                          NS0
                                                  3%
CHO cells are still the workhorse of                                          Sp2/0
                                                  3%
the biopharmaceutical industry.
                                                  7%                          rat-mouse hybrid
                                                                              hybridoma
Other common host cell systems                                                Murine hybridoma
include the various mouse myeloma
                                                                              Murine myeloma
strains, including NS0 and sp2/0, as
                                                                              Immortalized
well as yeast cells, mostly Pichia                                            human
pastoris and Saccharomyces cerevisiae.
Despite      its    lack    of     mammalian
glycosylation machinery, E. coli is still widely used for smaller proteins and antibody
fragments, and E. coli-produced biopharmaceuticals accounted for 30 percent of newly
approved products during the period from 2006 to 2010 (Walsh, 2010).

                                                                DCU | Glycosylation and host cells
Mammalian Cells
The obvious choice for producing recombinant human proteins are mammalian cells.
The major advantage of mammalian cells is that they possess the ability to produce
proteins with fully human-type glycosylation (Butler, 2005). Additionally, they are able
to produce large molecular weight proteins, and natively possess the machinery to
conduct correct folding, quality control, and secretion (Demain, 2009).

There are several major limitations to mammalian cells as a producer strain. Mammalian
cells have extraordinarily high cost of culture, mostly due their fastidious media
requirements. A fully validated production process using these cells can cost $2-4 million
per year in growth media alone (Demain, 2009). They also have a slower growth cycle
than microbial producer strains, with doubling times of 12-20 hours, approximately ten
times slower than that of E. coli strains (Sunstrom, 2000).

Mammalian cell lines are susceptible to infection by viruses which are potentially
pathogenic to humans. Infection of a production process can be disastrous and can cause
the entire production facility to be shut down by regulators (Berting, 2010). Whole
batches of product have been lost to viral contamination (Bethencourt, 2009). Their
media requirements are notoriously fastidious. Mammalian cells produce their own
glycosylated proteins, which makes downstream separation of the target glycoprotein
much more difficult.

Historically, mammalian cell culture processes have suffered from low product yields,
but through advances in media and production optimization, gene amplification and
plasmid engineering, this productivity has increased markedly, often reaching expression
levels of 10-15 g/L for some products in CHO cells (Huang, 2010).

In addition to these advances, new producer strains have been developed. The PER.C6
cell line is derived from human retinal tissue and looks to have a future as a major
producer cell line. PER.C6 cells can tolerate very high cell densities in culture; up to 150
x 106 cells/mL have been reported (Golden, 2009), they show robust scale up
characteristics (Xie, 2003), and can produce recombinant protein at very high titres of at
least to 25 g/L (Schirmer, 2010).




 Glycosylation and host cells | DCU
Yeasts
Like humans, yeasts belong to the eukaryotic domain. This being so, many of the
metabolic pathways and molecular machinery are conserved between both species,
including much of the glycosylation apparatus (Rich, 2009). The two most common
strains used for industrial production of recombinant proteins are Saccharomyces
cerevisiae and Pichia pastoris. Both strains have had their genomes thoroughly
characterized (Goffeau, 1996) (De Schutter, 2009).

Yeasts have properties that make them attractive as a biopharmaceutical production
system for several reasons. Yeast cells enjoy both the fast growth rates of microbes and
the complex enzymatic machinery of eukaryotes (Verma, 1998). They offer reasonable
yields of recombinant product, yields of up to 9 g/L have been reported (Valdivieso-
Ugarte, 2006). Their growth kinetics and nutritional requirements are well characterized,
so scale-up from lab bench to industrial fermentation is a relatively simple procedure
(Hamilton, 2007).

Yeasts can tolerate very high cell densities, as high as 130 g/L in some strains (Gellison,
1992), magnifying the product yield attainable per litre of culture volume (Cregg, 2009).
Additionally, yeasts can be induced to secrete expressed recombinant therapeutics into
the culture media, simplifying the downstream processing, which typically accounts for
50-80 percent of the total cost associated with a production process (Roque, 2004). Yeast
cells are capable of carrying out N-linked glycosylation at the Asn-X-Ser/Thr motif, but
there are differences between native yeast-produced N-glycans and those produced in
human cells.

Both yeast and human cells share the same initial steps in their N-linked glycosylation
pathways. The Glc3Man9GlcNAc2 glycan precursor is constructed on a membrane
bound dolichol-PP on the endoplasmic reticulum. This precursor is then transferred to
an Asn residue on the peptide as it emerges from the ribosome (Jigami, 2008). This is
carried out by an oligosaccharyltransferase complex consisting of eight subunits; which
differ structurally between humans and yeast, but which carry out the same function
(Kelleher, 2006). After this, the glycan is trimmed by three glucose units and one
mannose unit, leaving a Man8GlcNAc2 structure, which is then exported to the Golgi
apparatus (Jigami, 2008).


                                                           DCU | Glycosylation and host cells
It is after this step that human and yeast N-glycosylation diverge. In human cells, the
glycan is trimmed and extended with a series of glycosidases and glycosyltransferases to
yield the final glycan structure, which is often capped with sialic acid residues (Hamilton,
2007). In yeast cells, the core Man8GlcNAc2 is extended, adding numerous mannose
units, leaving the glycan comparatively hypermannosylated (Gemmill, 1999).

The major technical hurdle with using yeast cells as a host for producing recombinant
human therapeutics is the inactivation of the endogenous machinery that leads to
hypermannosylation and the introduction of glycosyltransferases and glycosidases that
lead to the production of human-like glycans. The first objective has been achieved in S.
cerevisiae by eliminating the Och1 gene, which codes for the first enzyme in the
mannose-extension pathway, thereby preventing hypermannosylation (Nakanishi-
Shindo, 1993).

On the second objective, progress has been made towards producing human-type
glycans. By introducing the Mns-II and Gnt-II genes, minimal human complex-type
glycans (GlcNAc2Man3GlcNAc2) have been produced in P. pastoris (Hamilton, 2003).
Combined, these advances pave the way towards the use of yeast cells to produce
functionl, homogeneous human therapeutic glycoproteins.




Bacteria
Although once dismissed as lacking the ability to perform complex glycosylation, it is
now known that many bacterial strains can produce even more complex and diverse
glycan structures than even mammalian cells (Abu-Qarn, 2008). This opens the door to
the possibility of using bacterial cells as production vectors. This is an extremely
desirable goal from an economic standpoint for several reasons. Bacteria have much
shorter generation times than the currently used CHO cells. Given optimal growth
conditions, an E. coli population can double in as little as 18-20 minutes (Irwin, 2010),
while a mammalian culture will take 12-20 hours (Nakahara, 2002) (Sunstrom, 2000).

Bacterial hosts tend to be quite easy to modify genetically, and there are strains available
to meet any number of specific culture and production conditions (Bachmann, 1972).
 Glycosylation and host cells | DCU
They can grow to quite high cell densities in culture (20 to 175 g/l dry biomass) (Lee,
1996), with recombinant target protein accounting for up to 30 percent of total cellular
protein by weight (Suzuki, 2006). Yields as high as 0.5 mg/mL have been achieved by
optimizing many conditions simultaneously (Sivashanmugam, 2009).

One of the main intrinsic limitations to bacterial expression systems is the tendency of
recombinant proteins to crash out into aggregates and inclusion bodies. This is partly
because bacteria lack the specific charaponins which aid in the folding of mammalian
proteins. This problem has been theoretically mitigated by coexpressing the chaperone
with the target protein (Lin, 2001).

There are large differences between bacterial and mammalian glycosylation patterns.
The mammalian core glycan is attached to the asparagine residues by an N-
acetylglucosamine-β(1-4) linkage, whereas C. jejuni, the model organism for studying
bacterial glycosylation, utilises an N-acetylgalactosamine-α(1-3)-bacillosamine linkage
(Stanley, 2009) (Young, 2002). Bacillosamine is a rare amino sugar found in bacteria.
Additionally, the recognition sequence differs between human and bacterial systems.

Human cells utilise the three residue Asn-X-Ser/Thr sequon, while bacteria use a longer
five residue Asp/Glu -X-Asp-X-Ser/Thr sequon (Rich, 2009), where x represents any
amino acid except proline, which inhibits attachment of the N-glycan through stearic
hindrance (Shyama, 2010). Additionally, human glycosylation occurs during protein
translation as the peptide emerges from the ribosome, while bacterial glycosylation
occurs after translation and folding as a true post-translational modification (Kowarik,
2006).

Taking into account these differences, a putative roadmap can be built. First, it will be
necessary to manipulating the bacterial host cells into utilising the mammalian GlcNAc
peptide linkage. Then it will be necessary to deal with the sequon problem. This can be
solved in two ways. The bacterial machinery can be modified to utilise the human-type
sequon or the target protein sequence can be modified to include the bacterial N-
glycosylation sequon. Ideally, the glycosylation apparatus will need to be transferred into
a more ideal production host such as E. coli or B. subtilis.




                                                               DCU | Glycosylation and host cells
All three of these goals have been achieved to an extent. By transferring pglB, an
oligosaccharyltransferase with relaxed substrate specificity, into bacterial expression
systems, human-type glycans with GlcNAc at their non-reducing can be linked to
proteins (Wacker, 2006).

Considering the problem of consensus sequences, Kowarik (2006) et al were able to
engineer additional Asp/Glu-X-Asp-X-Ser/Thr sequons into AcrA, a component of a
multi-drug efflux complex, from C. jejuni, and achieved functional glycosylation at these
sites. Thus there is no theoretical reason why human therapeutic proteins could not be
engineered to replace their N-gylcosylation sequons with bacterial versions, or to add
additional sequons to noncatalytic loops, where they are less likely to interfere with
protein function.

Considering the host strain issue, once the mechanics of C. jejuni glycosylation were
elucidated in detail, it was relatively easy to transfer this system into other bacterial
species, including E. coli (Wacker, 2002). Furthermore, this system has been used to
produce functional, correctly folded, glycosylated murine single-chain antibody
fragments, proteins with high therapeutic potential (Lizak, 2011).




Analytical methods
To analyse a pool glycan structures extracted from a cell or protein sample, two steps are
needed. Firstly, the glycans must be separated and secondly, the individual glycans must
be identified. To date, three major classes of glycan analysis have emerged;
electrophoresis, chromatography and mass spectrometry (MS) (Pabst, 2011). Note than
in practise, many of these techniques are used in tandem to achieve higher-resolution
separation or more comprehensive analysis.

Mass spectrometric
Within mass spectrometry, there are two methods which have dominated. Matrix-
assisted laser desorption/ionization – time of flight (MALDI-TOF) is suited to the
analysis of biological structures because the method of ionization is less hard, meaning

 Analytical methods | DCU
fragile biological structures are less likely to fragment. The glycans are embedded in a
matrix composed of crystallised low molecular weight molecules. 2,5-dihydroxy benzoic
acid (DHB) and its derivatives are the most widely-used matrices (Harvey, 2005). The
matrix is irradiated with a high-powered UV laser, causing some of the matrix molecules
to vaporise into a hot cloud of gas. Through a series of reactions, the glycans become
bound to charged species, usually sodium cations (Morelle, 2007). The charged glycans
are then sent to a mass analyser.

The mass analyser generates a defined electric filed which accelerates the glycans
towards a detector. This ‘time of flight’ is determined by the mass-to-charge ratio (m/z)
of the glycan. The analyser outputs a mass spectrum, where each peak represents a
charged fragment of the original glycan (Morelle, 2009). By comparing the output
spectrum to a database of such spectra, the glycan can be identified.

The other major MS technique is electrospray ionization (ESI).Instead of embedding the
sample in a molecular matrix; the ions are formed directly from solution (Chait, 2011).
The solution containing the glycans is forced through a thin capillary tube. At the tip of
the tube, a strong electric field is applied. As the glycans leave the tube, they are exposed
to this field and ionized. This creates a spray of charged particles. The ions are then
detected by a TOF analyser, or other equivalent apparatus (Loo, 2000).

MS techniques can be coupled into tandem arrays. These can be used, for example, to
first separate a pool of glycans by their mass charge ratio, then selecting a set of glycans
within a specific range of (m/z) vales to go on to a second stage where they are
fragmented    by collision-induced dissociation (CID) and analysed again, providing
further structural information (Harvey, 2000).

Chromatographic
In chromatographic methods, the glycan solution is pumped through a densely-packed
stationary phase. The glycans are slowed down on their passage through the column by
the stationary phase particles. The samples elute from the column at different times, this
is the retention time. The retention time is directly dependant on the glycan mass, so
with a properly calibrated column the mass of each glycan can be determined from the
retention time.



                                                                    DCU | Analytical methods
Within the glycobiology field, several chromatographic techniques have dominated.
High pH anion exchange chromatography (HPAEC) has been one of the most widely-
used techniques to date (Townsend, 1991). HPAEC is sensitive to branching patterns
and oligosaccharide compositions. The glycans bind to the stationary phase which is
loaded with negatively-charged functional groups. The glycans are then displaced and
eluted by the introduction of a positively charged eluent (Dionex, 1997).

Another method used for smaller proteins is reverse-phase high-performance liquid
chromatography (RP-HPLC).          RP-HPLC uses a non-polar stationary phase. The
glycans are derivatised (tagged) with a highly hydrophobic molecule that allows the
glycan to be selectively eluted (Wuhrer, 2005). This method can provide detailed
information about glycan structure and isomeric configuration (Gillmeister, 2009).
Another advantage of HPLC-based systems is that there exist large databases of peak
positions for glycans, making identification much easier (Campbell, 2008).

Electrophoretic
The major electrophoretic method used in the characterization of glycans is capillary
electrophoresis (CE). Like all electrophoretic methods, CE separation is based on the
size-to-charge ratio of a sample. Protein glyco-isoforms tend to be structurally very
similar. The differences may be difficult to detect using chromatographic or
spectrometric methods (Zamfir, 2008). The major advantage of CE is that it has
unrivalled separation power (Mechref, 2009). The sample is introduced into a chamber
where it is taken up by a capillary tube. An electric field is applied over the tube. As the
glycans travel through the tube, they are separated out by the slight differences in their
mass-to-charge ratios (Landers, 1995).




Bioaffinity methods
Bioaffinity methods are the newest breakthrough in the field of glycobiology applications.
It is defined as the separation of molecules based on their reversible interaction with
biological macromolecules. These separations involve highly-specific interactions
between the glycoligand and a carbohydrate-binding protein (Tetala, 2010). The
advantage of bioaffinity chromatography is that it allows highly-selective one step



 Analytical methods | DCU
purification of glycoproteins, meaning less sample needs to be used. It can be used for
analytical, preparatory and diagnostic applications.

Lectins are proteins involved in carbohydrate-based recognition. Given their high-
specificity for distinct oligosaccharide epitopes, lectins are the logical solution for the
stationary-phase ligand. They are able to select for not only overall glycan structure, but
also for the configuration of the linkages between the monosaccharide units (Mechref,
2002). This allows them to isolate a target from a pool of glycans or glycoproteins.

Several lectin-affinity based techniques have been developed. The first is the standard
chromatographic column approach, where the lectins are immobilised onto a stationary
phase and the pool of glycoproteins are washed through the column (Tetala, 2010). The
target glycoproteins bind to the lectins and can be eluted and fractionated. This
approach can be improved by adding multiple lectins to the same column, allowing
simultaneous selection of multiple targets at one time (Yang, 2004). Another approach is
to immobilize the lectins onto the surface of a microtiter plate. This allows simultaneous
analysis of an even greater number of glycoproteins in a rapids and high-throughput
fashion (Kuno, 2005).

A modification of this approach is the enzyme-linked lectin assay (ELLA) (Wu, 2009).
Similar to the standard ELISA assay, the target glycoprotein is bound to the surface of a
microtiter plate, before a blocking solution is added to prevent nonspecific binding.
Then lectins are added to the wells, and bind to the target glycoprotein, if present. The
lectins are detected by the addition of labelled antibodies raised to bind to antigens on
the lectin. The label is detected and the quantity of label is proportional to the quantity
of target glycoprotein.

A similar technique is the carbohydrate array, in which glycans themselves are
immobilized to the plate and their interaction with a target protein is assayed (Oyelaran,
2007).




                                                                  DCU | Analytical methods
Bioinformatics
The primary goal of glycomics is to profile the expression and activity of all of the
glycosyltransferases, glycosidases, and other glycosylation apparatus, as well as the entire
glycan component within a cell under specific conditions (Aoki-Kinoshita, 2008). The
glycoprofiles of cells under different conditions can be compared. From this, we will
begin to deduce to conditions which lead to specific glycosylation events and, ultimately,
have the ability to rationally design the glycosylation machinery of producer cell lines.
Bioinformatic methods will be the key to achieving this goal.

Complexity of glycan structures
Computationally speaking, glycomics is a much more daunting field than proteomics and
genomics. Unlike genes and proteins, the glycoprofile of a cell is not encoded directly
by the genome but indirectly through the compliment of glycosylation enzymes active in
the cell. This means that to predict the glycosylation state of a newly-translated protein,
one would need to have full knowledge of the entire spectrum of glycosylation enzymes
expressed at the time as well as their substrate specificities, kinetic rates, their cofactors
and inhibitors, and a plethora of other variables.

Additionally, glycans themselves are structurally highly complex molecules. Unlike the
linear sequence of nucleic acids or amino acids which describe genes and proteins,
glycans are composed of sugar monosaccharaides. These can be linked together by
different types of bonds, and a residue can be linked to more than one other residue
(branching), and each branch can be linked in a number of different ways. Other
structural variables include anomeric configuration, epimeric configuration, and
reducing terminal attachments (Laine, 1994).

These structural traits magnify enormously the number of possible unique structures
that can be built from a given set of residues. To put this into perspective, the four DNA
bases can give rise to 256 possible four-unit combinations, and the twenty amino acids
can give rise to 160,000 possible four-unit arrangements, while a four-unit glycan can
potentially be assembled in 15 million different combinations (Von der Leith, 2004).

Prediction tools
N-glycosylation only occurs at sites which carry the specific Asn-XSer/Thr motif. If this
motif occurs in a given peptide sequence, it represents a potential glycosylation site.
 Bioinformatics | DCU
Bioinformaticians have taken advantage of this knowledge to uncover information about
the ubiquity of glycoproteins in nature. Zafar et al (2011) used a computer algorithm to
scan all of the sequence data available on the ExPASy protein database and flag any
sequences which contained the signature motif. They found that more than 50 percent
of all proteins (prokaryotic and eukaryotic) contain at least one copy of the motif. This
overturns previously held assumptions about the exclusivity of glycosylation machinery
to eukaryotes (Nothaft, 2010).

A similar experiment conducted by Thanka et al applied statistical analysis to the
sequences of 992 experimentally-confirmed O-linked glycoproteins in an effort to
discover a signature O-linked motif analogous to the Asn-XSer/Thr motif of N-
glycosylation (Christlet, 2001). They found that the presence of a proline residue at
either the + or -1 position relative to the serine/threonine site strongly promotes
           3
glycosylation and that aromatic amino acids near the site strongly inhibit glycosylation.

Glycobiology databases
To aid to experiments like these, several online tools and databases have been developed
to identify signature motifs for different types of glycosylation within an uploaded
sequence (Kamath, 2011). NetNGlyc is an artificial neural network trained not only to
find N-glycosylation sequence motifs, but to look at them in the context of the
surrounding amino acids whose influence on the local topology and physiochemical
properties may affect the glycosylation state of the biding site (Gupta, 2004).

Similarly, NetOGlyc parses the local sequence surrounding serine/threonine sites to find
probable O-linked glycosylation sites (Julenius, 2005). Several databases have emerged
which attempt to catalogue and document glycoproteins. Each has a different specialty.
GlycoBase records the HPLC elution data for N-linked glycans (Campbell, 2008), while
GlycosuiteDB contains over 3200 unique entries from 245 different species,
documenting the glycan structure, peptide linkage type and host protein (Cooper, 2003).
O-GlycBase contains detailed information on O- and C- linked glycans and was the
dataset used to train the NetOGlyc neural net mentioned above.

An enormous amount of data is being produced by glycobiology labs around the world
and the bottleneck has now shifted to the computational analysis and interpretation of
this data. To fully take advantage of the possibilities that this field offers, it will be
necessary to build and utilise new bioinformatics tools, algorithms and databases.


                                                                       DCU | Bioinformatics
References
Abu-Qarn M (2008). Not just for Eukarya anymore: protein glycosylation in Bacteria and
Archaea. Curr Opin Struct Biol. 18(5):544-50.

Albert R (1993). SDZ CO 611: a highly potent glycated analog of somatostatin with improved
oral activity. Life Sci. 53(6), 517-25.

Aoki-Kinoshita KF (2008). An Introduction to Bioinformatics for Glycomics Research. PLoS
Comput Biol. 4(5), e1000075.

Apweiler R (1999). On the frequency of protein glycosylation, as deduced from analysis of the
SWISS-PROT database. Biochim. Biophys. Acta. 1473, 4–8.

Arakawa T (2001). Factors affecting short-term and long-term stabilities of proteins. Adv Drug
Deliv Rev. 46(1−3), 307–326.

Bachmann BJ (1972). Pedigrees of some mutant strains of Escherichia coli K-12. Bacteriol Rev.
36(4), 525–557.

Benet L (1984). Pharmacokinetics: Basic Principles and Its Use as a Tool in Drug Metabolism.
In: Drug Metabolism and Drug Toxicity (Mitchell JR and Horning MG) Raven Press, New
York, 199.

Berting A (2010). Virus susceptibility of Chinese hamster ovary (CHO) cells and detection of
viral contaminations by adventitious agent testing. Biotechnol Bioeng. 106(4), 598-607.

Bethencourt V (2009). Virus stalls Genzyme plant. Nature Biotechnology. 27, 681.

Bhatnagar BS (2007). Protein stability during freezing: separation of stresses and mechanisms of
protein stabilization. Pharm Dev Technol. 12(5), 505-23.

build and utilise new bioinformatics tools, algorithms and databases.

Burton D (2006). Sugar Determines Antibody Activity. 313(5787), 627-628.

Butler M (2005). Animal cell cultures: recent achievements and perspectives in the production of
biopharmaceuticals. Appl Microbiol Biotechnol. 68, 283–91.

Campbell M (2008). GlycoBase and autoGU: tools for HPLC-based glycan analysis.
Bioinformatics. 24(9), 1214-1216.

Campbell MP (2008). GlycoBase and autoGU: tools for HPLC-based glycan analysis.
Bioinformatics. 24 (9), 1214-1216

Campion B (1989). Presence of fucosylated triantennary, tetraantennary and pentaantennary
glycans in transferrin synthesized by the human hepatocarcinoma cell line Hep G2. Eur J
Biochem. 184(2), 405-13.

 References | DCU
Casadevall N (2002). Pure red-cell aplasia and antierythropoietin antibodies in patients treated
with recombinant erythropoietin. N. Engl. J. Med. 346(7), 469-75.

Ceaglio N (2008). Novel long-lasting interferon alpha derivatives designed by glycoengineering.
Biochimie. 90(3), 437–49

Chait BT (2011). Mass Spectrometry in the Postgenomic Era. Annu Rev Biochem. 80, 239-46.

Chi EY (2003). Physical stability of proteins in aqueous solution: mechanism and driving forces
in non-native protein aggregation. Pharm. Res. 20, 1325–1336.

Christlet HT (2001). Database Analysis of O-Glycosylation Sites in Proteins. Biophysical journal.
80(2), 952-960

Cooper C (2003). GlycoSuiteDB: a curated relational database of glycoprotein glycan structures
and their biological sources. Nucl. Acids Res. 31(1), 511-513.

Cregg JM (2009). Expression in the yeast Pichia pastoris. Methods Enzymol. 463, 169-89.

Darling RJ (2002). Glycosylation of erythropoietin affects receptor binding kinetics: role of
electrostatic interactions. Biochemistry. 41, 14524-14531.

De Schutter K (2009). Genome sequence of the recombinant protein production host Pichia
pastoris. Nat Biotechnol. 27(6), 561-6.

Demain AL (2009). Production of recombinant proteins by microbes and higher organisms.
Biotechnol Adv. 27(3), 297-306.

Dionex Corporation (1997) Glycoprotein Oligosaccharide Analysis Using High-performance
Anion-Exchange Chromatograph. Dionex Technical Note 42.

Egleton RD (2001). Improved blood-brain barrier penetration and enhanced analgesia of an
opioid peptide by glycosylation. J. Pharmacol. Exp. Ther. 299(3), 967-72.

Egrie JC (1993). The role of carbohydrate on the biological activity of erythropoietin.
Glycoconjugate J. 10, 263.

Egrie JC (2001). Development and characterization of novel erythropoiesis stimulating protein
(NESP). Br. J. Cancer. 1, 3-10.

Elliott S (2004). Control of rHuEPO biological activity: the role of carbohydrate. Exp. Hematol.
32, 1146-1155.

Fernandes AI (2001). The effect of polysialylation on the immunogenicity and antigenicity of
asparaginase: implication in its pharmacokinetics. Int. J. Pharm. 217(1-2), 215-24.

Gellison G (1992). High-level expression of foreign genes in Hansenula polymorpha. Biotech
Adv. 10, 179–89

                                                                                DCU | References
Gemmill TR (1999). Overview of N- and O-linked oligosaccharide structures found in various
yeast species. Biochim. Biophys. Acta. 1426, 227-237.

Geyer H (2006). Strategies for analysis of glycoprotein glycosylation. Biochim Biophys Acta.
1764(12), 1853-69.

Gillmeister MP (2009). An HPLC-MALDI MS method for N-glycan analyses using smaller size
samples: application to monitor glycan modulation by medium conditions. Glycoconj J. 26(9),
1135-49.

Goffeau A (1996). Life with 6000 genes. Science. 274(5287), 563-7.

Golden K (2009). The XD Process: Development of a High-Titer Process for PER.C6 Cells.
21st Meeting of the European Society of Animal Cell Technology, Dublin, Ireland, 7–10 June
2009.

Gupta R (2004). Prediction of N-glycosylation sites in human proteins. [In preparation].

Hamilton SR (2003). Production of complex human glycoproteins in yeast. Science. 301(5637),
1244-6.

Hamilton SR (2007). Glycosylation engineering in yeast: the advent of fully humanized yeast.
Curr Opin Biotechnol. 18(5), 387-92.

Harvey DJ (2000). Collision‐induced fragmentation of underivatized N‐linked carbohydrates
ionized by electrospray. J Mass Spectrom. 35(10), 1178-90.

Harvey DJ (2005). Structural determination of N-linked glycans by matrix-assisted laser
desorption/ionization and electrospray ionization mass spectrometry. Proteomics. 5(7), 1774-86.

Hirschler B (2010). World's top-selling drugs in 2014 vs 2010. Thomson Reuters research
[Online]. Available from: http://www.reuters.com/article/2010/04/13/roche-avastin-drugs-
idUSLDE63C0BC20100413 [Accessed 22 November 2011].

Hoiberg-Nielsen R (2006). Interrelationships of glycosylation and aggregation kinetics for
Peniophora lycii phytase. Biochemistry. 45(15), 5057–5066.

Hual L (2008). Urea denaturation by stronger dispersion interactions with proteins than water
implies a 2-stage unfolding. Proc Natl Acad Sci U S A. 105(44), 16928-33.

Huang YM (2010). Maximizing productivity of CHO cell-based fed-batch culture using
chemically defined media conditions and typical manufacturing equipment. Biotechnol Prog. 26,
1400–10.

Imperiali B (1999). Effect of N-linked glycosylation on glycopeptide and glycoprotein structure.
Curr Opin. Chem. Biol. 3(6), 643-9.

 References | DCU
Irwin P (2010). Evidence for a bimodal distribution of Escherichia coli doubling times below a
threshold initial cell concentration. BMC Microbiol. 10, 207.

Jigami Y (2008). Yeast glycobiology and its application. Biosci Biotechnol Biochem. 72(3), 637-
48.

Julenius K (2005). Prediction, conservation analysis and structural characterization of
mammalian mucin-type O-glycosylation sites. Glycobiology. 15, 153-164.

Kahn CM (2011). Drug Distribution. The Merck Veterinary Manual, 9th Edition. [Online].
Available from: http://www.merckvetmanual.com/mvm/index.jsp?cfile=
                                                                 htm/bc/190110.htm
[Accessed 16 November 2011].

Kamath KS (2011). Proteomic databases and tools to decipher post-translational modifications. J
Proteomics. [Epub ahead of print].

Kaneko Y (2006). Anti-Inflammatory Activity of Immunoglobulin G Resulting from Fc
Sialylation. Science. 313(5787), 670-673.

Kanwar YS (1984). Biophysiology of glomerular filtration and proteinuria. Lab. Invest. 51, 7–21.

Kayser V (2011). Glycosylation influences on the aggregation propensity of therapeutic
monoclonal antibodies. Biotechnology Journal. 6, 38–44.

Kelleher DJ (2006). An evolving view of the eukaryotic oligosaccharyltransferase. Glycobiology.
16(4), 47-62.

Kerwin BA (2007). Protect from light: photodegradation and protein biologics. J Pharm Sci. 96,
1468–79.

Khoury GA (2011). Proteome-wide post-translational modification statistics: frequency analysis
and curation of the swiss-prot database. Sci Rep. 13, 1.

Kopacek KB (2011). Drug Adsorption. The Merck Manual of Diagnosis and Therapy, 19th
Edition [Online]. Available from:
http://www.merckmanuals.com/professional/clinical_
                                                 pharmacology/pharmacokinetics/drug_
                                                                                   abso
rption.html#
           v1108980 [Accessed 16 November 2011].

Kopacek KB (2011). Drug Distribution. The Merck Manual of Diagnosis and Therapy, 19th
Edition [Online]. Available from:
http://www.merckmanuals.com/professional/clinical_
                                                 pharmacology/pharmacokinetics/drug_
                                                                                   abso
rption.html#
           v1108980 [Accessed 16 November 2011].

Kornfeld R (1985). Assembly of asparagine-linked oligosaccharides. Annu Rev Biochem. 54, 631-
664.


                                                                                DCU | References
Kowarik M (2006). Definition of the bacterial N-glycosylation site consensus sequence. EMBO J.
25(9), 1957–1966.

Kowarik M (2006). N-Linked glycosylation of folded proteins by the bacterial
oligosaccharyltransferase. Science. 314, 1148–1150

Kuno A (2005). Evanescent-field fluorescence-assisted lectin microarray: a new strategy for
glycan profiling. Nat. Methods. 2, 851–856.

Laine RA. (1994). A calculation of all possible oligosaccharide isomers both branched and linear
yields 1.05 3 10(12) structures for a reducing hexasaccharide. Glycobiology. 4: 759–767.

Landers JP (1995). Clinical capillary electrophoresis. Clinical Chemistry. 41, 495-509.

Lee SY (1996). High cell density culture of Escherichia coli. Trends Biotechnol. 14, 98-105

Li H (2010). Pharmacological significance of glycosylation in therapeutic proteins. Curr Opin
Biotechnol. 20(6), 678-84.

Lin WJ (2001). DegP-coexpression minimizes inclusion-body formation upon overproduction of
recombinant penicillin acylase in Escherichia coli. Biotechnol Bioeng. 73(6), 484-92.

Lizak C (2011). N-Linked glycosylation of antibody fragments in Escherichia coli. Bioconjug
Chem. 22(3), 488-96.

Loo, J. (2000). Electrospray ionization mass spectrometry: a technology for studying
noncovalent macromolecular complexes. International Journal of Mass Spectrometry. 200(1-3),
175-186.

Macdougall I (1999). Pharmacokinetics of Novel Erythropoiesis Stimulating Protein Compared
with Epoetin Alfa in Dialysis Patients. J. Am. Soc. Nephrol. 10, 2392–2395.

Manning MC (2010). Stability of protein pharmaceuticals: an update. Pharm Res. 27(4), 544-75.

Masarova J (2001). Stability enhancement of Escherichia coli penicillin G acylase by
glycosylation with yeast mannan. Biotechnol Appl Biochem. 34, 127–133.

Mechref T (2002). Structural Investigations of Glycoconjugates at High Sensitivity. Chem. Rev.
102(2), 321–370

Mechref Y (2009) Glycomic analysis by capillary electrophoresis-mass spectrometry. Mass
Spectrom Rev. 28(2):207-22.

Moore WV (1980). Role of aggregated human growth hormone (hGH) in development of
antibodies to hGH. J. Clin. Endocrinol. Metab. 51(4), 691-7.

Morell AG (1968). Physical and chemical studies on ceruloplasmin. V. Metabolic studies on
sialic acid-free ceruloplasmin in vivo. J. Biol. Chem. 243(1), 155–9.

 References | DCU
Morell AG (1971). The role of sialic acid in determining the survival of glycoproteins in the
circulation. J. Biol. Chem. 246, 1461–1467.

Morelle W (2007). Analysis of protein glycosylation by mass spectrometry. Nat. Protoc. 2(7),
1585-602.

Morelle W (2009). Analysis of N- and O-linked glycans from glycoproteins using MALDI-TOF
mass spectrometry. Methods Mol Biol. 534, 5-21.

Nakahara T (2002). Effects of exposure of CHO-K1 cells to a 10-T static magnetic field.
Radiology. 224(3), 817-22.

Nandita GD (2003). Controlled-release of oral dosage forms. Formulation, Fill and Finish. 10-
16.

Nomoto M (1998). Improvement of intestinal absorption of peptide drugs by glycosylation:
transport of tetrapeptide by the sodium ion-dependent D-glucose transporter. J. Pharm. Sci.
87(3), 326-32.

Nothaft H (2010). Protein glycosylation in bacteria: sweeter than ever. Nature Reviews
Microbiology. 8, 765-778.

Oyelaran O (2007). Application of carbohydrate array technology to antigen discovery and
vaccine development. Expert Rev Vaccines. 6(6), 957-69.

Pabst M (2011). Glycan analysis by modern instrumental methods. Proteomics. 11, 631–643

Pandhal J (2010). N-Linked glycoengineering for human therapeutic proteins in bacteria.
Biotechnol Lett. 32(9), 1189-98.

Perlman S (2003). Glycosylation of an N-terminal extension prolongs the half-life and increases
the in vivo activity of follicle stimulating hormone. J. Clin. Endocrinol. Metab. 88(7), 3227-35.

Philo JS (2009). Mechanisms of protein aggregation. Curr Pharm Biotechnol. 10, 348–51.

Pristov JB (2011). A comparative study of antioxidative activities of cell-wall polysaccharides.
Carbohydr Res. 346(14), 2255-9.

Rich JR (2009). Emerging methods for the production of homogeneous human glycoproteins.
Nat Chem Biol. 5(4), 206-15.

Roque AC (2004). Antibodies and genetically engineered related molecules: production and
purification. Biotechnol Prog. 20(3), 639-54.

Rosenberg AS (2006). Effects of protein aggregates: an immunologic perspective. AAPS J. 8,
501–7.



                                                                                 DCU | References
Runkel L (1998). Structural and functional differences between glycosylated and non-
glycosylated forms of human interferon-beta (IFN-beta). Pharm Res. 15(4), 641–649.

Russell D (2009). Site-selective chemical protein glycosylation protects from autolysis and
proteolytic degradation. Carbohydr Res. 344(12), 1508-14

Saluja A (2008). Nature and consequences of protein-protein interactions in high protein
concentration solutions. International Journal of Pharmaceutics. 358, 1-15

Sasayama S (2000). Glycosylated human interleukin-1alpha, neoglyco IL-1alpha, coupled with
N-acetylneuraminic acid exhibits selective activities in vivo and altered tissue distribution.
Glycoconj. J. 17(6), 353-9.

Satoh M (2006). Non-fucosylated therapeutic antibodies as next-generation therapeutic
antibodies. Expert Opin. Biol. Ther. 6(11), 1161-73.

Saxena S (1987). Purification and characterization of glucosidase II involved in N-linked
glycoprotein processing in bovine mammary gland. Biochem J. 247(3), 563–570.

Schirmer EB (2010). Primary clarification of very high-density cell culture harvests by enhanced
cell settling. BioProcess Int. 8, 32–9.

Sethuraman N (2006). Challenges in therapeutic glycoprotein production. Curr Opin
Biotechnol.17, 341–346

Shailubhai K (1987). Purification and characterization of glucosidase I involved in N-linked
glycoprotein processing in bovine mammary gland. Biochem J. 247(3), 555–562.

Shental-Bechor D (2008). Effect of glycosylation on protein folding: A close look at
thermodynamic stabilization. PNAS. 105(24), 8256-8261.

Shyama R (2010). Evolutionary Pattern of N-Glycosylation Sequon Numbers in Eukaryotic
ABC Protein Superfamilies. Bioinform. Biol. Insights. 4, 9–17.

Sivashanmugam A (2009). Practical protocols for production of very high yields of recombinant
proteins using Escherichia coli. Protein Sci. 18(5), 936–948.

Solá RJ (2007). Modulation of protein biophysical properties by chemical glycosylation:
biochemical insights and biomedical implications. Cell Mol Life Sci. 64(16), 2133–2152.

Solá RJ (2009). Effects of glycosylation on the stability of protein pharmaceuticals. J Pharm Sci.
98(4), 1223-45.

Solá RJ et al (2010). Glycosylation of therapeutic proteins: an effective strategy to optimize
efficacy. BioDrugs. 24(1), 9-21.



 References | DCU
Soltero R (2001). The oral delivery of protein and peptide drugs. Innov. Pharmacol. Technol. 1,
106–110

Stanley P (2009). N-Glycans. In: Source Essentials of Glycobiology. (Varki A, Cummings RD,
Esko JD, Freeze HH, Stanley P, Bertozzi CR, Hart GW, Etzler ME, eds.). 2nd edition. Cold
Spring Harbor (NY): Cold Spring Harbor Laboratory Press, Chapter 8.

Sunstrom NA (2000). Regulated autocrine growth of CHO cells. Cytotechnology. 34(1-2), 39-
46.

Suzuki M (2006). Bacterial bioreactors for high yield production of recombinant protein. J Biol
Chem. 281(49), 37559-65.

Sytkowski AJ (1991). Biological activity and structural stability of N-deglycosylated recombinant
human erythropoietin. Biochem Biophys Res Commun. 176(2), 698-704.

Tams JW (1999). Adapting protein solubility by glycosylation. N-glycosylation mutants of
Coprinus cinereus peroxidase in salt and organic solutions. Biochim Biophys Acta. 1432(2), 214–
221.

Tang L (2004). Pharmacokinetic aspects of biotechnology products, J. Pharm. Sci. 93, 2184–
2204.

Tetala KK (2010). Bioaffinity chromatography on monolithic supports. J Sep Sci. 33(3), 422-38.

Townsend RR (1991). Analysis of glycoprotein oligosaccharides using high-pH anion exchange
chromatography. Glycobiology. 1(2), 139-47.

Uchida E (1997). Effect of active oxygen radicals on protein and carbohydrate moieties of
recombinant human erythropoietin. Free Radic Res. 27(3), 311–323.

Valdivieso-Ugarte M (2006). Expression of an Aspergillus niger glucose oxidase in
saccharomyces cerevisiae and its use to optimize fructo-oligosaccharides synthesis. Biotechnol
Prog. 22(4), 1096-101.

Vegarud G (1975). The resistance of glycoproteins to proteolytic inactivation. Acta Chem Scand
B. 29(8), 887–888.

Venter JC (2001). The sequence of the human genome. Science. 291, 1304–1351.

Verma R (1998). Antibody engineering: comparison of bacterial, yeast, insect and mammalian
expression systems. J Immunol Methods. 216(1-2), 165-81.

Vogt G (1997). Protein thermal stability, hydrogen bonds, and ion pairs. J Mol Biol. 269(4), 631-
43.



                                                                               DCU | References
Von der Lieth CW (2004). Bioinformatics for glycomics: status, methods, requirements and
perspectives. Brief Bioinform. 5(2), 164-78.

Wacker M (2002). N-Linked glycosylation in Campylobacter jejuni and its functional transfer
into E. coli. Science 298, 1790–1793.

Wacker M (2006). Substrate specificity of bacterial oligosaccharyltransferase suggests a common
transfer mechanism for the bacterial and eukaryotic systems. Proc. Natl. Acad. Sci. USA. 103,
7088–7093.

Walsh G (2010). Biopharmaceutical benchmarks 2010. Nat Biotechnol. 28(9), 917-24.

Wang C (1996). Influence of the carbohydrate moiety on the stability of glycoproteins.
Biochemistry. 35(23), 7299–7307.

Wang W (1999). Instability, stabilization, and formulation of liquid protein pharmaceuticals. Int
J Pharm. 185(2), 129–188.

Wang W (2005). Protein aggregation and its inhibition in biopharmaceutics. Int J Pharm. 289(1-
2), 1-30.

Wormald MR (1999). Glycoproteins: glycan presentation and protein-fold stability. Structure
Fold Des. 7(7), 155–160.

Wu AM (2009). Lectins as tools in glycoconjugate research. Glycoconj J. 26(8), 899-913.

Wuhrer M (2005). Protein glycosylation analysis by liquid chromatography–mass spectrometry.
J Chromatogr B Analyt Technol Biomed Life Sci. 825(2), 124-33.

Xie L (2003). Large-scale propagation of a replication-defective adenovirus vector in stirred-tank
bioreactor PER.C6 cell culture under sparging conditions. Biotechnol Bioeng. 83(1), 45-52.

Yamane-Ohnuki N (2004). Establishment of FUT8 knockout Chinese hamster ovary cells: an
ideal host cell line for producing completely defucosylated antibodies with enhanced
antibodydependent cellular cytotoxicity. Biotechnol. Bioeng. 87, 614–622.

Yang Z (2004). Approach to the comprehensive analysis of glycoproteins isolated from human
serum using a multi-lectin affinity column. J. Chromatogr. A. 1053, 79–88

Yoshimura M (1998). Bisecting GlcNAc structure is implicated in suppression of stroma-
dependent haemopoiesis in transgenic mice expressing N-acetylglucosaminyltransferase III.
Biochem J. 331(3), 733–742.

Young NM (2002). Structure of the N-linked glycan present on multiple glycoproteins in the
Gram-negative bacterium, Campylobacter jejuni. J Biol Chem. 277(45), 42530-9




 References | DCU
Zafar S (2011). Computational analysis reveals abundance of potential glycoproteins in Archaea,
Bacteria and Eukarya. Bioinformation. 6(9), 352-5.

Zamfir AD (2008). Glycosylation analysis of glycoproteins and proteoglycans using capillary
electrophoresis-mass spectrometry strategies. ELECTROPHORESIS. 29, 2485–2507.




                                                                              DCU | References

More Related Content

What's hot

What's hot (19)

Biosynthesis of steroids
Biosynthesis of steroidsBiosynthesis of steroids
Biosynthesis of steroids
 
NA- 01: Chemistry of Nucleotides & It's clinical importance
NA- 01: Chemistry of Nucleotides & It's clinical importanceNA- 01: Chemistry of Nucleotides & It's clinical importance
NA- 01: Chemistry of Nucleotides & It's clinical importance
 
Structural elucidation of sucrose
Structural elucidation of sucroseStructural elucidation of sucrose
Structural elucidation of sucrose
 
12 glycolysis
12 glycolysis12 glycolysis
12 glycolysis
 
Glycine and Serine Amino acid
Glycine and Serine Amino acid Glycine and Serine Amino acid
Glycine and Serine Amino acid
 
Cholesterol for classs
Cholesterol for classsCholesterol for classs
Cholesterol for classs
 
CARBOHYDRATE METABOLISM : GLYCOLYSIS
CARBOHYDRATE METABOLISM : GLYCOLYSISCARBOHYDRATE METABOLISM : GLYCOLYSIS
CARBOHYDRATE METABOLISM : GLYCOLYSIS
 
Carbohydrate Metabolism
Carbohydrate Metabolism Carbohydrate Metabolism
Carbohydrate Metabolism
 
CHO metabolism
CHO metabolismCHO metabolism
CHO metabolism
 
Digestion glycolysis
Digestion glycolysisDigestion glycolysis
Digestion glycolysis
 
Cholesterol synthesis steps and regulation
Cholesterol synthesis   steps and regulationCholesterol synthesis   steps and regulation
Cholesterol synthesis steps and regulation
 
Biosynthesis of Phospholipids
Biosynthesis of PhospholipidsBiosynthesis of Phospholipids
Biosynthesis of Phospholipids
 
Lipids
LipidsLipids
Lipids
 
Pentose phosphate pathway,hmp shunt
Pentose phosphate pathway,hmp shuntPentose phosphate pathway,hmp shunt
Pentose phosphate pathway,hmp shunt
 
Glycolysis (with animated pathway)
Glycolysis (with animated pathway)Glycolysis (with animated pathway)
Glycolysis (with animated pathway)
 
C0532011019
C0532011019C0532011019
C0532011019
 
CARBOHYDRATES
CARBOHYDRATESCARBOHYDRATES
CARBOHYDRATES
 
Metabolism of Carbohydrates
Metabolism of CarbohydratesMetabolism of Carbohydrates
Metabolism of Carbohydrates
 
Cholesterolsynthesis stepsandregulation-17-18
Cholesterolsynthesis stepsandregulation-17-18Cholesterolsynthesis stepsandregulation-17-18
Cholesterolsynthesis stepsandregulation-17-18
 

Similar to Glycosylation literature survey ricky connolly 091211

Glycogen Biochemistry
Glycogen BiochemistryGlycogen Biochemistry
Glycogen BiochemistryRasol Sindy
 
利用分子動力學電腦模擬研究聚穀氨醯胺及胰
利用分子動力學電腦模擬研究聚穀氨醯胺及胰利用分子動力學電腦模擬研究聚穀氨醯胺及胰
利用分子動力學電腦模擬研究聚穀氨醯胺及胰Hsin-Lin Chiang
 
Cdg alg9 tj marrie research day poster:
Cdg alg9 tj marrie research day poster:  Cdg alg9 tj marrie research day poster:
Cdg alg9 tj marrie research day poster: the-fog
 
sodium Glucose co transporter 2 inhibitors GLIFLOZINS .pptx
sodium Glucose co transporter 2 inhibitors GLIFLOZINS .pptxsodium Glucose co transporter 2 inhibitors GLIFLOZINS .pptx
sodium Glucose co transporter 2 inhibitors GLIFLOZINS .pptxLawanYarimaMasaba
 
Ch14-glycosaminoglycans-glycoproteins4.pptx
Ch14-glycosaminoglycans-glycoproteins4.pptxCh14-glycosaminoglycans-glycoproteins4.pptx
Ch14-glycosaminoglycans-glycoproteins4.pptxSudhaKavitha1
 
Classification of polysaccharides, gluconeogenesis and glucogenolysis.
Classification of polysaccharides, gluconeogenesis and glucogenolysis.Classification of polysaccharides, gluconeogenesis and glucogenolysis.
Classification of polysaccharides, gluconeogenesis and glucogenolysis.Arunima Sur
 
Protein Glycosylation and its Role in Muscular Dystrophies
Protein Glycosylation and its Role in Muscular DystrophiesProtein Glycosylation and its Role in Muscular Dystrophies
Protein Glycosylation and its Role in Muscular DystrophiesJames Franklin
 
Lec 6 level 3-nu(carbohydrate metabolism ii)
Lec 6  level 3-nu(carbohydrate metabolism ii)Lec 6  level 3-nu(carbohydrate metabolism ii)
Lec 6 level 3-nu(carbohydrate metabolism ii)dream10f
 
glycogenesis and glycogenolysis
glycogenesis and glycogenolysis glycogenesis and glycogenolysis
glycogenesis and glycogenolysis NANDITA42
 
Glycan Structural Analysis Throughout Biotherapeutic Development
Glycan Structural Analysis Throughout Biotherapeutic Development Glycan Structural Analysis Throughout Biotherapeutic Development
Glycan Structural Analysis Throughout Biotherapeutic Development SGS
 
Biochemistry exam 3 enzyme sugars nov2016 S Jamal
Biochemistry exam 3 enzyme sugars nov2016 S JamalBiochemistry exam 3 enzyme sugars nov2016 S Jamal
Biochemistry exam 3 enzyme sugars nov2016 S JamalSayed Jamal
 

Similar to Glycosylation literature survey ricky connolly 091211 (20)

Therapeutic Glycoprotein
Therapeutic GlycoproteinTherapeutic Glycoprotein
Therapeutic Glycoprotein
 
Glycogen Biochemistry
Glycogen BiochemistryGlycogen Biochemistry
Glycogen Biochemistry
 
Kaempferol increases levels of oenzyme Q
Kaempferol increases levels of oenzyme QKaempferol increases levels of oenzyme Q
Kaempferol increases levels of oenzyme Q
 
Gluconeogenesis
GluconeogenesisGluconeogenesis
Gluconeogenesis
 
利用分子動力學電腦模擬研究聚穀氨醯胺及胰
利用分子動力學電腦模擬研究聚穀氨醯胺及胰利用分子動力學電腦模擬研究聚穀氨醯胺及胰
利用分子動力學電腦模擬研究聚穀氨醯胺及胰
 
Cdg alg9 tj marrie research day poster:
Cdg alg9 tj marrie research day poster:  Cdg alg9 tj marrie research day poster:
Cdg alg9 tj marrie research day poster:
 
in silico
in silico in silico
in silico
 
glycoprotein
glycoproteinglycoprotein
glycoprotein
 
biosynthesis of polysaccharides
 biosynthesis of polysaccharides biosynthesis of polysaccharides
biosynthesis of polysaccharides
 
sodium Glucose co transporter 2 inhibitors GLIFLOZINS .pptx
sodium Glucose co transporter 2 inhibitors GLIFLOZINS .pptxsodium Glucose co transporter 2 inhibitors GLIFLOZINS .pptx
sodium Glucose co transporter 2 inhibitors GLIFLOZINS .pptx
 
Ch14-glycosaminoglycans-glycoproteins4.pptx
Ch14-glycosaminoglycans-glycoproteins4.pptxCh14-glycosaminoglycans-glycoproteins4.pptx
Ch14-glycosaminoglycans-glycoproteins4.pptx
 
Classification of polysaccharides, gluconeogenesis and glucogenolysis.
Classification of polysaccharides, gluconeogenesis and glucogenolysis.Classification of polysaccharides, gluconeogenesis and glucogenolysis.
Classification of polysaccharides, gluconeogenesis and glucogenolysis.
 
Protein Glycosylation and its Role in Muscular Dystrophies
Protein Glycosylation and its Role in Muscular DystrophiesProtein Glycosylation and its Role in Muscular Dystrophies
Protein Glycosylation and its Role in Muscular Dystrophies
 
Lec 6 level 3-nu(carbohydrate metabolism ii)
Lec 6  level 3-nu(carbohydrate metabolism ii)Lec 6  level 3-nu(carbohydrate metabolism ii)
Lec 6 level 3-nu(carbohydrate metabolism ii)
 
Final Paper
Final PaperFinal Paper
Final Paper
 
glycogenesis and glycogenolysis
glycogenesis and glycogenolysis glycogenesis and glycogenolysis
glycogenesis and glycogenolysis
 
Glycan Structural Analysis Throughout Biotherapeutic Development
Glycan Structural Analysis Throughout Biotherapeutic Development Glycan Structural Analysis Throughout Biotherapeutic Development
Glycan Structural Analysis Throughout Biotherapeutic Development
 
Biochemistry exam 3 enzyme sugars nov2016 S Jamal
Biochemistry exam 3 enzyme sugars nov2016 S JamalBiochemistry exam 3 enzyme sugars nov2016 S Jamal
Biochemistry exam 3 enzyme sugars nov2016 S Jamal
 
Rational Drug design
Rational Drug designRational Drug design
Rational Drug design
 
Glycogen Metabolism.pptx
Glycogen Metabolism.pptxGlycogen Metabolism.pptx
Glycogen Metabolism.pptx
 

Recently uploaded

[2024]Digital Global Overview Report 2024 Meltwater.pdf
[2024]Digital Global Overview Report 2024 Meltwater.pdf[2024]Digital Global Overview Report 2024 Meltwater.pdf
[2024]Digital Global Overview Report 2024 Meltwater.pdfhans926745
 
08448380779 Call Girls In Greater Kailash - I Women Seeking Men
08448380779 Call Girls In Greater Kailash - I Women Seeking Men08448380779 Call Girls In Greater Kailash - I Women Seeking Men
08448380779 Call Girls In Greater Kailash - I Women Seeking MenDelhi Call girls
 
How to Troubleshoot Apps for the Modern Connected Worker
How to Troubleshoot Apps for the Modern Connected WorkerHow to Troubleshoot Apps for the Modern Connected Worker
How to Troubleshoot Apps for the Modern Connected WorkerThousandEyes
 
Boost Fertility New Invention Ups Success Rates.pdf
Boost Fertility New Invention Ups Success Rates.pdfBoost Fertility New Invention Ups Success Rates.pdf
Boost Fertility New Invention Ups Success Rates.pdfsudhanshuwaghmare1
 
Strategies for Landing an Oracle DBA Job as a Fresher
Strategies for Landing an Oracle DBA Job as a FresherStrategies for Landing an Oracle DBA Job as a Fresher
Strategies for Landing an Oracle DBA Job as a FresherRemote DBA Services
 
Exploring the Future Potential of AI-Enabled Smartphone Processors
Exploring the Future Potential of AI-Enabled Smartphone ProcessorsExploring the Future Potential of AI-Enabled Smartphone Processors
Exploring the Future Potential of AI-Enabled Smartphone Processorsdebabhi2
 
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...Miguel Araújo
 
Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024The Digital Insurer
 
Tech Trends Report 2024 Future Today Institute.pdf
Tech Trends Report 2024 Future Today Institute.pdfTech Trends Report 2024 Future Today Institute.pdf
Tech Trends Report 2024 Future Today Institute.pdfhans926745
 
CNv6 Instructor Chapter 6 Quality of Service
CNv6 Instructor Chapter 6 Quality of ServiceCNv6 Instructor Chapter 6 Quality of Service
CNv6 Instructor Chapter 6 Quality of Servicegiselly40
 
08448380779 Call Girls In Diplomatic Enclave Women Seeking Men
08448380779 Call Girls In Diplomatic Enclave Women Seeking Men08448380779 Call Girls In Diplomatic Enclave Women Seeking Men
08448380779 Call Girls In Diplomatic Enclave Women Seeking MenDelhi Call girls
 
How to convert PDF to text with Nanonets
How to convert PDF to text with NanonetsHow to convert PDF to text with Nanonets
How to convert PDF to text with Nanonetsnaman860154
 
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...Neo4j
 
Presentation on how to chat with PDF using ChatGPT code interpreter
Presentation on how to chat with PDF using ChatGPT code interpreterPresentation on how to chat with PDF using ChatGPT code interpreter
Presentation on how to chat with PDF using ChatGPT code interpreternaman860154
 
From Event to Action: Accelerate Your Decision Making with Real-Time Automation
From Event to Action: Accelerate Your Decision Making with Real-Time AutomationFrom Event to Action: Accelerate Your Decision Making with Real-Time Automation
From Event to Action: Accelerate Your Decision Making with Real-Time AutomationSafe Software
 
🐬 The future of MySQL is Postgres 🐘
🐬  The future of MySQL is Postgres   🐘🐬  The future of MySQL is Postgres   🐘
🐬 The future of MySQL is Postgres 🐘RTylerCroy
 
Scaling API-first – The story of a global engineering organization
Scaling API-first – The story of a global engineering organizationScaling API-first – The story of a global engineering organization
Scaling API-first – The story of a global engineering organizationRadu Cotescu
 
04-2024-HHUG-Sales-and-Marketing-Alignment.pptx
04-2024-HHUG-Sales-and-Marketing-Alignment.pptx04-2024-HHUG-Sales-and-Marketing-Alignment.pptx
04-2024-HHUG-Sales-and-Marketing-Alignment.pptxHampshireHUG
 
2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...Martijn de Jong
 
GenCyber Cyber Security Day Presentation
GenCyber Cyber Security Day PresentationGenCyber Cyber Security Day Presentation
GenCyber Cyber Security Day PresentationMichael W. Hawkins
 

Recently uploaded (20)

[2024]Digital Global Overview Report 2024 Meltwater.pdf
[2024]Digital Global Overview Report 2024 Meltwater.pdf[2024]Digital Global Overview Report 2024 Meltwater.pdf
[2024]Digital Global Overview Report 2024 Meltwater.pdf
 
08448380779 Call Girls In Greater Kailash - I Women Seeking Men
08448380779 Call Girls In Greater Kailash - I Women Seeking Men08448380779 Call Girls In Greater Kailash - I Women Seeking Men
08448380779 Call Girls In Greater Kailash - I Women Seeking Men
 
How to Troubleshoot Apps for the Modern Connected Worker
How to Troubleshoot Apps for the Modern Connected WorkerHow to Troubleshoot Apps for the Modern Connected Worker
How to Troubleshoot Apps for the Modern Connected Worker
 
Boost Fertility New Invention Ups Success Rates.pdf
Boost Fertility New Invention Ups Success Rates.pdfBoost Fertility New Invention Ups Success Rates.pdf
Boost Fertility New Invention Ups Success Rates.pdf
 
Strategies for Landing an Oracle DBA Job as a Fresher
Strategies for Landing an Oracle DBA Job as a FresherStrategies for Landing an Oracle DBA Job as a Fresher
Strategies for Landing an Oracle DBA Job as a Fresher
 
Exploring the Future Potential of AI-Enabled Smartphone Processors
Exploring the Future Potential of AI-Enabled Smartphone ProcessorsExploring the Future Potential of AI-Enabled Smartphone Processors
Exploring the Future Potential of AI-Enabled Smartphone Processors
 
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...
Mastering MySQL Database Architecture: Deep Dive into MySQL Shell and MySQL R...
 
Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024
 
Tech Trends Report 2024 Future Today Institute.pdf
Tech Trends Report 2024 Future Today Institute.pdfTech Trends Report 2024 Future Today Institute.pdf
Tech Trends Report 2024 Future Today Institute.pdf
 
CNv6 Instructor Chapter 6 Quality of Service
CNv6 Instructor Chapter 6 Quality of ServiceCNv6 Instructor Chapter 6 Quality of Service
CNv6 Instructor Chapter 6 Quality of Service
 
08448380779 Call Girls In Diplomatic Enclave Women Seeking Men
08448380779 Call Girls In Diplomatic Enclave Women Seeking Men08448380779 Call Girls In Diplomatic Enclave Women Seeking Men
08448380779 Call Girls In Diplomatic Enclave Women Seeking Men
 
How to convert PDF to text with Nanonets
How to convert PDF to text with NanonetsHow to convert PDF to text with Nanonets
How to convert PDF to text with Nanonets
 
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
 
Presentation on how to chat with PDF using ChatGPT code interpreter
Presentation on how to chat with PDF using ChatGPT code interpreterPresentation on how to chat with PDF using ChatGPT code interpreter
Presentation on how to chat with PDF using ChatGPT code interpreter
 
From Event to Action: Accelerate Your Decision Making with Real-Time Automation
From Event to Action: Accelerate Your Decision Making with Real-Time AutomationFrom Event to Action: Accelerate Your Decision Making with Real-Time Automation
From Event to Action: Accelerate Your Decision Making with Real-Time Automation
 
🐬 The future of MySQL is Postgres 🐘
🐬  The future of MySQL is Postgres   🐘🐬  The future of MySQL is Postgres   🐘
🐬 The future of MySQL is Postgres 🐘
 
Scaling API-first – The story of a global engineering organization
Scaling API-first – The story of a global engineering organizationScaling API-first – The story of a global engineering organization
Scaling API-first – The story of a global engineering organization
 
04-2024-HHUG-Sales-and-Marketing-Alignment.pptx
04-2024-HHUG-Sales-and-Marketing-Alignment.pptx04-2024-HHUG-Sales-and-Marketing-Alignment.pptx
04-2024-HHUG-Sales-and-Marketing-Alignment.pptx
 
2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...
 
GenCyber Cyber Security Day Presentation
GenCyber Cyber Security Day PresentationGenCyber Cyber Security Day Presentation
GenCyber Cyber Security Day Presentation
 

Glycosylation literature survey ricky connolly 091211

  • 1. Glycosylation for biopharmaceutical drugs Ricky Connolly Biotechnology, Dublin City University; email: ricky.connolly2@mail.dcu.ie Abstract Since the mid 2000s, the patents for many blockbuster drugs have begun to expire. This is the driving force behind the current great development in the biopharmaceutical industry, the ‘second generation’ biopharmaceuticals. This new trend is focussed on modifying existing protein therapeutics in order to enhance their pharmacological, biological, and structural properties. These modifications include but are not limited to: generation of fusion conjugates, incorporation of chemical modifications such as pegylation, and, crucially for this paper, modification of glycosylation profiles. The goal of this paper is to outline the chemical and biological basis of protein glycosylation, to examine the pharmacological and structural implications of glycosylation, to compare and evaluate the current production strategies, and to survey the current range of analytical methods available to characterise glycoprotein therapeutics.
  • 2. Table of Contents Introduction ................................................................................................................................. 3 Overview ...................................................................................................................................... 3 Glycan synthesis ........................................................................................................................... 4 Stability .......................................................................................................................................... 6 Aggregation .................................................................................................................................. 6 Crosslinking ................................................................................................................................. 7 Proteolysis .................................................................................................................................... 7 Oxidation ..................................................................................................................................... 7 pH ................................................................................................................................................ 7 Kinetic Denaturation ................................................................................................................... 8 Chemical Denaturation ............................................................................................................... 8 Temperature ................................................................................................................................ 8 Pharmacology .............................................................................................................................. 9 Receptor Binding ......................................................................................................................... 9 Circulatory lifetime .................................................................................................................... 10 Bioavailability ............................................................................................................................. 10 Distribution................................................................................................................................ 11 Clearance rates ........................................................................................................................... 12 Antibody function ...................................................................................................................... 12 Immunogenicity ......................................................................................................................... 12 Glycosylation and host cells .................................................................................................. 13 Mammalian Cells ....................................................................................................................... 14 Yeasts.......................................................................................................................................... 15 Bacteria....................................................................................................................................... 16 Analytical methods ................................................................................................................... 18 Mass spectrometric .................................................................................................................... 18 Chromatographic ....................................................................................................................... 19 Electrophoretic .......................................................................................................................... 20 Bioaffinity methods .................................................................................................................... 20 Bioinformatics ........................................................................................................................... 22 Complexity of glycan structures ................................................................................................ 22 Prediction tools .......................................................................................................................... 22 Glycobiology databases .............................................................................................................. 23 References................................................................................................................................... 24 Abstract | DCU
  • 3. Introduction The human genome contains at least 30,000 protein-coding genes, and through alternative mRNA splicing, these give rise to over 100,000 proteins (Venter, 2001). The diversity of the human proteome is further magnified by post-translational modification of proteins. At least 50 percent of mammalian proteins are glycosylated (Zafar, 2011). More than two thirds of the 200 or so biopharma- ceutical products licenced for sale in the United States and European Union are recombinant human proteins (Li, 2010). In 2010, five of the top ten selling pharmaceutical products were recombinant human proteins and this is projected to rise to eight of the top ten by 2014 (Hirschler, 2010). Additionally, over 70 percent of the therapeutics currently in clinical trials are glycosylated human proteins (Sethuraman, 2006). Overview Glycosylation is defined as the covalent attachment of oligosaccharide moieties (glycans) to the side chains of amino acid residues of proteins. There are at least five different classes of glycosylation, each defined by the amino acid the glycan in linked to and the type of linkage used. By far the most common of these are N-linked and O- linked glycosylation (Apweilier, 1999). This review will focus solely on N-linked glycosylation because it is more common type found in human therapeutic protein therapeutics (Pandhal, 2010). N-linked glycosylation involves the enzymatic attachment of the N-Acetylglucosamine residue at the terminal end of the glycan to the amide group of an asparagine residue by means of a β1 glycosidic linkage (Geyer, 2006). DCU | Introduction
  • 4. The asparagine must be located within the three residue sequence Asparagine-X- Serine/Threonine, where X represents any amino acid except proline, which inhibits attachment of the N-glycan through stearic hindrance caused by its rotationally-locked side group (Shyama, 2010). There are further structural requirements in addition to the Asp-X-Ser/Thr sequon, mostly concerning the three-dimensional localisation and accessibility of the sequon within the folded protein (Apweilier, 1999). Glycan synthesis N-glycosylation takes place in the endoplasmic reticulum, at the same sites as protein translation. The first step in glycan synthesis involves the addition of two GlcNAc residues and five mannose residues to the membrane-anchored dolichol-PP. The dolichol-PP-Man5GlcNAc2 group is then flipped to the inner membrane side of the endoplasmic reticulum by the enzyme RFT1 (Pandhal, 2010). The core oligosaccharide (Glc3Man9GlcNAc2) is constructed on the GlcNAc residue by a series of glycosyltransferases. After this, the glycan is transferred to a sequon-labelled asparagine residue on the protein chain as it emerges from the ribosome (Pandhal, 2010). Introduction | DCU
  • 5. The glycan is trimmed by glucosidase I and II to remove the α1,2-linked (Shailubhai, 1987) and α1,3-linked (Saxena, 1987) glucose residues, respectively. This leaves us with a Man9GlcNAc2 structure. Next, α1,2-mannosidase removes one of the α1,2-linked mannose residues. The Man8GlcNAc2 glycan is then exported to the Golgi apparatus. Here, more α1,2-mannosidases remove several mannose residues, producing Man5GlcNAc2. After this, the glycosylation pathway diverges into different patterns of residue trimming and addition, catalysed by a series of glycosidases and glycosyltransferases (Pandhal, 2010). This results in three distinct prototypic classes of glycan structure into which the majority of glycans fall: complex, hybrid, and oligomannose (Kornfeld, 1985). Oligomannose glycans contain only mannose residues in addition to their core structures. Hybrid glycans can contain a diverse range of residues including galactose, glucoronic acid, and xylose (in plants). Complex glycans resemble hybrid glycans but they come with varying degrees of fucosylation and sialylation (Pandhal, 2010). Sialylation is often vital for correct protein function, this will be discussed later. Glycans are constructed from monosaccharide units linked together with glycosidic bonds. Unlike peptides and nucleic acids, one monosaccharide unit can be linked to multiple other units. This is called branching. Biantennary glycans are the most common, but triantennary and tetrantennary glycans are not unheard of (Campion, 1989). Another common structural feature is to have a single (β1,4) GlcNAc residue linked to the first branching point mannose unit. This bisecting GlcNAc residue is often involved in signalling (Yoshimura, 1998). The molecular structures, branching types and families of DCU | Introduction
  • 6. glycans attached to a protein describe its microheterogeneity in terms of glycosylation. This is distinct from the macroheterogeneity, which is described by the total number of glycans attached to the protein and their positions. Stability A protein instability is defined as a physical or chemical vulnerability that is prone to altering the structural conformation or activity in a detrimental way. Although they have high therapeutic efficacy, the main drawback to pharmaceutical use of proteins is their inherent structural and chemical instabilities. These impose limitations on the protein production and purification stages, product formulation, storage and transport, and half- life in the body. Glycosylation has been shown to ameliorate many of these instabilities. In this section, the major protein instabilities will be outlined, and the efficacy of glycosylation on these will be investigated. Aggregation Proteins are very complex molecules with hundreds if not thousands of exposed functional groups. This makes non-native aggregation a very common problem for protein biopharmaceuticals. Aggregation can be caused by relatively small changes in the protein structure (Saluja, 2008). Aggregation can drastically reduce the biological function of a protein (Philo, 2009) and in some cases can induce an immunogenic reaction in patients (Rosenberg, 2006). Glycosylation has been shown to prevent the formation of protein aggregates (Kayser, 2011). The likely explanation is that the bulky spatial nature of glycan moieties provides a level of stearic repulsion between proteins (Imperiali, 1999), reducing the likelihood of aggregation (Hoiberg-Nielsen, 2006). Precipitation Precipitation is the ultimate effect of protein aggregation. Eventually, the proteins form aggregates so large that they become insoluble (Chi, 2003). Precipitation is a major problem for biotherapeutic product formulation because it is desirable to store protein solutions at relatively high concentrations, but at high enough concentrations most proteins will form aggregates and precipitate out of solution (Wang, 2005). Glycosylation decreases the propensity to form insoluble precipitates for many proteins (Kayser, 2011). It has been shown that the decrease in insolubility is directly Stability | DCU
  • 7. proportional to the size and number of glycans present (Tams, 1999). One possible explanation for this phenomenon is that glycans are more soluble than peptides, and they confer some of this solubility to the glycoprotein as a whole. Crosslinking Crosslinking, also known as polymerisation-induced inactivation, occurs primarily when the methionine residues of two different proteins form a disulphide linkage. This leads to the formation of protein oligomers which have diminished biological activity (Wang, 1999). Glycosylation has been shown to prevent chemical crosslinking between proteins (Runkel, 1998). The mechanism is probably the same as that of aggregation, the mutual stearic repulsion between glycosylated proteins. Proteolysis Proteolysis is the degradation of the peptide backbone through hydrolytic reactions. These reactions are carried out by the ubiquitous protease enzymes found in all tissues. They are a major problem from a drug administration perspective because protease- sensitivity can drastically reduce the level of therapeutic that reaches the site of action (Tang, 2004). Glycosylation has been shown to inhibit proteolytic degradation (Vegarud, 1975). The most likely mechanism is through stearic blockage of the protease cleavage sites by the glycan (Russell, 2009). Oxidation Proteins are susceptible to oxidative degradation at the level of their primary structure. This is of particular concern for proteins rich in the more reactive amino acids such as His, Met, Cys, Tyr and Trp (Manning, 2010), which are prone to accepting free radicals. Since free radicals are basically unavoidable at all stages of the pipeline from cell culture to drug administration (even exposure to stray light can be harmful (Kerwin, 2007)), oxidation poses a serious threat to the stability and functionality of therapeutic proteins. Glycosylation has been shown to reduce the effects of oxidative damage in at least one commercial therapeutic (Uchida, 1997). One proposed mechanism for this protection is that the glycans are ‘soaking up’ the radicals, thus preventing them from reaching the protein itself (Pristov, 2011). pH Proteins are only stable within a limited pH range. pH denaturation begins when the ion balance disrupts the hydrogen- and ion-bonding capacity of the amino acids. This affects the tertiary structure of the protein and leads to the formation of non-native chemical DCU | Stability
  • 8. bonds as the peptide reconfigures into a more thermodynamically-stable state (Solá, 2009). Glycosylation has been shown to improve the pH stability of protein therapeutics by up to a 13-fold increase (Masarova, 2001). This marked improvement is due to the fact that attached glycans decrease the solvent accessible surface area of the protein, acting as a molecular buffer between the electrostatic forces of the solvent and those of the protein. Kinetic Denaturation Due to their complex nature, protein molecules have several three-dimensional states in which they are thermodynamically stable, but usually have only state in which they are functionally active. This means that proteins can undergo kinetic inactivation even at low temperatures by ‘flipping’ to another stable but inactive state (Arakawa, 2001). It has been demonstrated that glycosylation increases the kinetic stability of proteins. Specifically, the level of stability conferred seems to be correlated to the number of glycans present, their positioning on the protein, and their size (Solá, 2007). This is most likely because a folded, glycosylated protein has a lower free-energy profile compared to both the unfolded protein and to the folded-unglycosylated protein (Shental-Bechor, 2008). Chemical Denaturation Chemical denaturation can be defined as the loss of structural integrity of a protein in response to exposure to a chemical agent. One of the primary reasons for chemical destabilisation is that the protein often has high Van der Waals affinity to the denaturant. This allows the chemical molecule to intrude into the tertiary structure of the protein, disrupting the global conformation and reducing functionality (Hual, 2008). Glycosylation has been shown to promote conformational stability in opposition to chemical denaturants (Sytkowski, 1991). The probable explanation for this effect is that glycosylation essentially ‘compacts’ the protein, increasing the strength of its internal ionic, hydrogen and Van der Waals bonds and reducing the peptide’s affinity for outside chemical molecules (Solá, 2007). Temperature All of the bonds within a protein are sensitive to thermal fluctuations. Outside of a small temperature range, these bonds will break or form non-native bonds, destroying the biological activity of the protein (Vogt, 1997). The effects of freezing are less well studied compared to those of heating, but some comprehensive studies of the phenomena exist (Bhatnagar, 2007). Glycosylation has been shown to improve the Stability | DCU
  • 9. thermal stability range for several therapeutically important proteins including EPO, alpha 1-antitrypsin, interferon-β, and follicle-stimulating hormone (Solá, 2009). It is likely that these increases in stability are due to the constraint of peptide mobility caused by glycan attachment (Wormald, 1999). The magnitude of thermal stabilisation is proportional to the number and size of the glycan attachments (Wang, 1996). Pharmacology Pharmacology is divided into two fields of study: pharmacokinetics and pharmacodynamics. Pharmacokinetics examines the action of drugs within the body over a given period of time; profiling distribution, metabolism, and excretion. Pharmacodynamics studies the mechanisms of action of drugs within the body, studying the drug-receptor interactions and dose/response profiles. In other words, pharmacokinetics studies what the body does to the drug, while pharmacodynamics studies what the drug does to the body (Benet, 1984). Proteins tend to have poor pharmacokinetic profiles because they are very quickly cleared by proteolytic degradation pathways, hepatic and renal elimination, and receptor-mediated endocytosis (Tang, 2004). They have sharp pharmacodynamics profiles due to their exceptionally high binding affinities with receptors compared to small molecule drugs and have high turnover rates of their substrates. Glycosylation strongly affects the pharmacological properties of a protein. In this section, the major intrinsic pharmacokinetic and pharmacodynamic limitations of proteins are reviewed. For each, the impact of glycosylation on these limitations are examined. Receptor Binding One of the most characteristic traits of proteins is the extraordinarily high affinity with which they bind to their receptors. This is both as blessing and a curse. A high receptor association rate allows the design of therapeutics with strong biological efficacy, but it often means the protein has a very blunt therapeutic response curve (Solá, 2010). This leads to dosage schemes that require multiple injections per day, with widely fluctuating levels of drug in the body over the course of the day. For this reason, it is often desirable to ‘smooth out’ the response curve to a protein therapeutic. In practise, this means reducing receptor affinity of the protein. DCU | Pharmacology
  • 10. Glycosylation has been used to reduce the receptor affinity for several commercially important protein therapeutics. Darling et al showed that EPO-IRS (the standard form of human erythropoietin recognized European Pharmacopoeia) showed a 20-fold lower receptor association rate compared with artificially deglycosylated EPO (Darling, 2002). Similarly, another study compared the relative receptor binding affinities of several isoforms of erythropoietin. The EPO isoforms are defined by the total number of sialic acid residues found on their glycocomponent. The experiment involved measuring the quantity of each isoform needed to displace inactive EPO from receptors expressed on the surface of human erythroleukemia cells. There was a direct inverse relationship between sialic acid content and receptor binding affinity (Egrie, 2001). The most likely mechanisms by which this reduction in binding affinity can be explained is that electrostatic repulsion between the sialic acid residues of the glycans and the receptor serve to decrease the liklihood of receptor binding (Elliott, 2004). Circulatory lifetime Glycosylation can have a dramatic effect on the circulatory lifetime of a therapeutic protein. Comparing the serum half-life of the enzyme ceruloplasmin in it’s a natively- glycosylated state with that of an artificially deglycosylated variant, the circulatory lifetime of the deglycosylated variant was found to be an order of magnitude lower (Morell, 1968). Similarly, the addition of extra glycans (hyperglycosylation) has been shown to decrease the clearance rate of proteins. Perlman et al (2003) observed a 4-fold increase in serum half-life of a variant of human follicle stimulating hormone (FSH) with two addition N-linked glycosylation sites. The sialic acid content of attached glycans has been shown to be of high importance to extending the serum half-life of a protein. Sialic residues have a net negative charge at biological pH. This electrostatic repulsion confers protection from both renal (Kanwar, 1984) and hepatic (Morell, 1971) clearence mechanisms. Bioavailability Today the vast majority of protein therapeutics are delivered by parenteral injection (Soltero 2001). This contrasts with small molecule drugs, which are typically delivered through oral routes (Nandita, 2003). From a clinical standpoint, it would be very desirable to have oral-administered protein therapeutics. Unfortunately, there are several barriers to making this technology a reality. Pharmacology | DCU
  • 11. Unless injected directly into the bloodstream, drugs must pass through several membrane barriers before they can begin systemic circulation, and if the target of the drug is intracellular, the peptide must pass through the lipid membrane of the cell. There are four mechanisms by which a protein therapeutic may pass through a membrane: passive and facilitated diffusion, active transport, and receptor mediated endocytosis (Kopacek, 2011). The rate at which a drug absorbs in the body is determined by the rate at which the drug is transported across these barriers. Glycosylation has been shown to improve protein absorption in several cases. Egleton et al (2001) demonstrated that the addition of an O-linked glycan to an opioid peptide icreased its blood brain barrier permeability from 1.0 μl/(min·g) to 2.2 μl/(min·g), with a comparable increase in measured analgesic activity. Albert et al (1993) produced a glycosylated, orally-active version of octreotide, a regulatory peptide that inhibits the production of somatropin and other growth hormones, which had ten times greater oral bioavailability compared to the parent molecule. Nomoto et al (1998) significantly improved intestinal uptake of a peptide by the sodium ion-dependent D-glucose transporter through the addition of a small glycan moiety. Distribution Drug distribution throughout the tissues of the body is controlled by blood perfusion, plasma protein and tissue binding affinity, pH, and membrane permeability (Kopacek, 2011). Controlling the distribution of a drug is vital to ensuring that the drug reaches the target tissue and that it does not end up in the wrong tissue, where it will have no (or even adverse) biological effects. Glycosylation has been shown to improve the tissue distribution of therapeutic drugs. Sasayama et al (2000) chemically glycosylated human interleukin-1ɑ with an N- acetylneuraminic acid moiety and monitored the in vivo tissue distribution in rats following intraperitoneal (IP) injection. Up to five-fold greater levels of the glycosylated variant were observed in the kidney, spleen, lung, and blood. Similarly, Ceaglio et al (2008) created a mutant version of interferon-ɑ with four additional N-glycans which displayed a ten-fold increase in distribution half-life (t1/2β) after ten hours post-injection, and levels remained detectable 96 hours after injection. The most likely explanations for the increase in tissue distribution are that the glycoproteins are protected from proteolytic and immune-inactivation pathways or that the more soluble glycans reduce the hydrophobicity of the peptide, increasing the rate of transport. DCU | Pharmacology
  • 12. Clearance rates Drug clearance studies measure the rate of removal of drug from circulation. This mainly takes place in the liver and kidneys. The drugs are carried along by the flow of blood until they reach the liver, where they are taken up by hepatocytes and are broken down by nonspecific proteases, before the amino acids are recycled back into the body’s metabolic cycle (Kahn, 2011). It is advantageous to attempt to reduce the clearance rates of protein biopharmaceuticals to a minimum so that expensive drug is not wasted. Any effective reduction of the level of clearance of a therapeutic will be valuable from a clinical and economic point of view, and glycosylation has shown to be an efficient strategy to achieve this reduction. A novel hyperglycosylated variant of human EPO, darbepoetin alfa (DA), was engineered to display two additional N-linked glycans. In a double-blind, randomized, cross-over clinical trial in humans, DA showed a 2.5-fold lower rate of clearance (1.6 mL/h·kg versus 4.0 mL/h·kg) (Macdougall, 1999). The reduction in clearance rate observed for hyperglycosylated proteins has been attributed to the increased sialic acid content of the introduced glycans (Egrie, 1993). Antibody function IgG antibodies have two large biantennary N-linked carbohydrate moities attached to the Fc effector region . The structure of these glycans has an effect on the chain orientation, chain spacing and surface residue exposure (Kaneko, 2006). These differences in structure can alter antibody effector function (Burton, 2006). It has been shown that IgG antibodies engineered to remove fucose residues from their glycan component have a much stronger antibody-dependent cell-mediated cytotoxicity profile compared to wild-type IgGs (Yamane-Ohnuki, 2004). This is of clinical importance because higher activation of immune system cells can stimulate the body to fight a wide range of conditions, including cancer (Satoh, 2006). Immunogenicity Glycosylation has been shown to help prevent the generation of neutralizing antibodies against therapeutic drugs. There are several theories which attempt to explain this effect. One explanation is that, as outlined earlier, glycosylation inhibits the formation of protein aggregates. Antibodies are often raised more efficiently against aggregates than individual proteins, so it stands to reason that glycosylation prevents the development of an immune reaction by inhibiting aggregation of the drug in circulation (Moore 1980). Another possible explanation of the inhibition of immune response is that the bulky Pharmacology | DCU
  • 13. nature of the carbohydrate moieties provide a degree of stearic hindrance, shielding the peptide chain below from immune cells (Casadevall, 2002). A further theory is that the terminal sialic acid residues of glycans provide a degree of electrostatic repulsion, again preventing immune cell surface receptors from binding (Fernandes, 2002). Glycosylation and host cells It is estimated that well over 50 percent of human proteins are glycosylated (Apweiler, 1999), although this figure is disputed (Khoury, 2011). Nevertheless, almost 40 percent of all approved biopharmaceutical 2% 55% a. products are glycosylated (Walsh, 2% 2% 2010). Therefore, for an organism to Mammalian 10% become widely used as a producer E. coli strain, it must be able to perform Yeast glycosylation to some extent. Between 2006 and 2010, 58 new Insect biopharmaceuticals gained approval. Animals 32 of these were produced in 29% Synthetic mammalian cell lines and of these, 24 CHO were produced in Chinese hamster 3% 3% 75% b. ovary (CHO) cells. It is clear that 6% NS0 3% CHO cells are still the workhorse of Sp2/0 3% the biopharmaceutical industry. 7% rat-mouse hybrid hybridoma Other common host cell systems Murine hybridoma include the various mouse myeloma Murine myeloma strains, including NS0 and sp2/0, as Immortalized well as yeast cells, mostly Pichia human pastoris and Saccharomyces cerevisiae. Despite its lack of mammalian glycosylation machinery, E. coli is still widely used for smaller proteins and antibody fragments, and E. coli-produced biopharmaceuticals accounted for 30 percent of newly approved products during the period from 2006 to 2010 (Walsh, 2010). DCU | Glycosylation and host cells
  • 14. Mammalian Cells The obvious choice for producing recombinant human proteins are mammalian cells. The major advantage of mammalian cells is that they possess the ability to produce proteins with fully human-type glycosylation (Butler, 2005). Additionally, they are able to produce large molecular weight proteins, and natively possess the machinery to conduct correct folding, quality control, and secretion (Demain, 2009). There are several major limitations to mammalian cells as a producer strain. Mammalian cells have extraordinarily high cost of culture, mostly due their fastidious media requirements. A fully validated production process using these cells can cost $2-4 million per year in growth media alone (Demain, 2009). They also have a slower growth cycle than microbial producer strains, with doubling times of 12-20 hours, approximately ten times slower than that of E. coli strains (Sunstrom, 2000). Mammalian cell lines are susceptible to infection by viruses which are potentially pathogenic to humans. Infection of a production process can be disastrous and can cause the entire production facility to be shut down by regulators (Berting, 2010). Whole batches of product have been lost to viral contamination (Bethencourt, 2009). Their media requirements are notoriously fastidious. Mammalian cells produce their own glycosylated proteins, which makes downstream separation of the target glycoprotein much more difficult. Historically, mammalian cell culture processes have suffered from low product yields, but through advances in media and production optimization, gene amplification and plasmid engineering, this productivity has increased markedly, often reaching expression levels of 10-15 g/L for some products in CHO cells (Huang, 2010). In addition to these advances, new producer strains have been developed. The PER.C6 cell line is derived from human retinal tissue and looks to have a future as a major producer cell line. PER.C6 cells can tolerate very high cell densities in culture; up to 150 x 106 cells/mL have been reported (Golden, 2009), they show robust scale up characteristics (Xie, 2003), and can produce recombinant protein at very high titres of at least to 25 g/L (Schirmer, 2010). Glycosylation and host cells | DCU
  • 15. Yeasts Like humans, yeasts belong to the eukaryotic domain. This being so, many of the metabolic pathways and molecular machinery are conserved between both species, including much of the glycosylation apparatus (Rich, 2009). The two most common strains used for industrial production of recombinant proteins are Saccharomyces cerevisiae and Pichia pastoris. Both strains have had their genomes thoroughly characterized (Goffeau, 1996) (De Schutter, 2009). Yeasts have properties that make them attractive as a biopharmaceutical production system for several reasons. Yeast cells enjoy both the fast growth rates of microbes and the complex enzymatic machinery of eukaryotes (Verma, 1998). They offer reasonable yields of recombinant product, yields of up to 9 g/L have been reported (Valdivieso- Ugarte, 2006). Their growth kinetics and nutritional requirements are well characterized, so scale-up from lab bench to industrial fermentation is a relatively simple procedure (Hamilton, 2007). Yeasts can tolerate very high cell densities, as high as 130 g/L in some strains (Gellison, 1992), magnifying the product yield attainable per litre of culture volume (Cregg, 2009). Additionally, yeasts can be induced to secrete expressed recombinant therapeutics into the culture media, simplifying the downstream processing, which typically accounts for 50-80 percent of the total cost associated with a production process (Roque, 2004). Yeast cells are capable of carrying out N-linked glycosylation at the Asn-X-Ser/Thr motif, but there are differences between native yeast-produced N-glycans and those produced in human cells. Both yeast and human cells share the same initial steps in their N-linked glycosylation pathways. The Glc3Man9GlcNAc2 glycan precursor is constructed on a membrane bound dolichol-PP on the endoplasmic reticulum. This precursor is then transferred to an Asn residue on the peptide as it emerges from the ribosome (Jigami, 2008). This is carried out by an oligosaccharyltransferase complex consisting of eight subunits; which differ structurally between humans and yeast, but which carry out the same function (Kelleher, 2006). After this, the glycan is trimmed by three glucose units and one mannose unit, leaving a Man8GlcNAc2 structure, which is then exported to the Golgi apparatus (Jigami, 2008). DCU | Glycosylation and host cells
  • 16. It is after this step that human and yeast N-glycosylation diverge. In human cells, the glycan is trimmed and extended with a series of glycosidases and glycosyltransferases to yield the final glycan structure, which is often capped with sialic acid residues (Hamilton, 2007). In yeast cells, the core Man8GlcNAc2 is extended, adding numerous mannose units, leaving the glycan comparatively hypermannosylated (Gemmill, 1999). The major technical hurdle with using yeast cells as a host for producing recombinant human therapeutics is the inactivation of the endogenous machinery that leads to hypermannosylation and the introduction of glycosyltransferases and glycosidases that lead to the production of human-like glycans. The first objective has been achieved in S. cerevisiae by eliminating the Och1 gene, which codes for the first enzyme in the mannose-extension pathway, thereby preventing hypermannosylation (Nakanishi- Shindo, 1993). On the second objective, progress has been made towards producing human-type glycans. By introducing the Mns-II and Gnt-II genes, minimal human complex-type glycans (GlcNAc2Man3GlcNAc2) have been produced in P. pastoris (Hamilton, 2003). Combined, these advances pave the way towards the use of yeast cells to produce functionl, homogeneous human therapeutic glycoproteins. Bacteria Although once dismissed as lacking the ability to perform complex glycosylation, it is now known that many bacterial strains can produce even more complex and diverse glycan structures than even mammalian cells (Abu-Qarn, 2008). This opens the door to the possibility of using bacterial cells as production vectors. This is an extremely desirable goal from an economic standpoint for several reasons. Bacteria have much shorter generation times than the currently used CHO cells. Given optimal growth conditions, an E. coli population can double in as little as 18-20 minutes (Irwin, 2010), while a mammalian culture will take 12-20 hours (Nakahara, 2002) (Sunstrom, 2000). Bacterial hosts tend to be quite easy to modify genetically, and there are strains available to meet any number of specific culture and production conditions (Bachmann, 1972). Glycosylation and host cells | DCU
  • 17. They can grow to quite high cell densities in culture (20 to 175 g/l dry biomass) (Lee, 1996), with recombinant target protein accounting for up to 30 percent of total cellular protein by weight (Suzuki, 2006). Yields as high as 0.5 mg/mL have been achieved by optimizing many conditions simultaneously (Sivashanmugam, 2009). One of the main intrinsic limitations to bacterial expression systems is the tendency of recombinant proteins to crash out into aggregates and inclusion bodies. This is partly because bacteria lack the specific charaponins which aid in the folding of mammalian proteins. This problem has been theoretically mitigated by coexpressing the chaperone with the target protein (Lin, 2001). There are large differences between bacterial and mammalian glycosylation patterns. The mammalian core glycan is attached to the asparagine residues by an N- acetylglucosamine-β(1-4) linkage, whereas C. jejuni, the model organism for studying bacterial glycosylation, utilises an N-acetylgalactosamine-α(1-3)-bacillosamine linkage (Stanley, 2009) (Young, 2002). Bacillosamine is a rare amino sugar found in bacteria. Additionally, the recognition sequence differs between human and bacterial systems. Human cells utilise the three residue Asn-X-Ser/Thr sequon, while bacteria use a longer five residue Asp/Glu -X-Asp-X-Ser/Thr sequon (Rich, 2009), where x represents any amino acid except proline, which inhibits attachment of the N-glycan through stearic hindrance (Shyama, 2010). Additionally, human glycosylation occurs during protein translation as the peptide emerges from the ribosome, while bacterial glycosylation occurs after translation and folding as a true post-translational modification (Kowarik, 2006). Taking into account these differences, a putative roadmap can be built. First, it will be necessary to manipulating the bacterial host cells into utilising the mammalian GlcNAc peptide linkage. Then it will be necessary to deal with the sequon problem. This can be solved in two ways. The bacterial machinery can be modified to utilise the human-type sequon or the target protein sequence can be modified to include the bacterial N- glycosylation sequon. Ideally, the glycosylation apparatus will need to be transferred into a more ideal production host such as E. coli or B. subtilis. DCU | Glycosylation and host cells
  • 18. All three of these goals have been achieved to an extent. By transferring pglB, an oligosaccharyltransferase with relaxed substrate specificity, into bacterial expression systems, human-type glycans with GlcNAc at their non-reducing can be linked to proteins (Wacker, 2006). Considering the problem of consensus sequences, Kowarik (2006) et al were able to engineer additional Asp/Glu-X-Asp-X-Ser/Thr sequons into AcrA, a component of a multi-drug efflux complex, from C. jejuni, and achieved functional glycosylation at these sites. Thus there is no theoretical reason why human therapeutic proteins could not be engineered to replace their N-gylcosylation sequons with bacterial versions, or to add additional sequons to noncatalytic loops, where they are less likely to interfere with protein function. Considering the host strain issue, once the mechanics of C. jejuni glycosylation were elucidated in detail, it was relatively easy to transfer this system into other bacterial species, including E. coli (Wacker, 2002). Furthermore, this system has been used to produce functional, correctly folded, glycosylated murine single-chain antibody fragments, proteins with high therapeutic potential (Lizak, 2011). Analytical methods To analyse a pool glycan structures extracted from a cell or protein sample, two steps are needed. Firstly, the glycans must be separated and secondly, the individual glycans must be identified. To date, three major classes of glycan analysis have emerged; electrophoresis, chromatography and mass spectrometry (MS) (Pabst, 2011). Note than in practise, many of these techniques are used in tandem to achieve higher-resolution separation or more comprehensive analysis. Mass spectrometric Within mass spectrometry, there are two methods which have dominated. Matrix- assisted laser desorption/ionization – time of flight (MALDI-TOF) is suited to the analysis of biological structures because the method of ionization is less hard, meaning Analytical methods | DCU
  • 19. fragile biological structures are less likely to fragment. The glycans are embedded in a matrix composed of crystallised low molecular weight molecules. 2,5-dihydroxy benzoic acid (DHB) and its derivatives are the most widely-used matrices (Harvey, 2005). The matrix is irradiated with a high-powered UV laser, causing some of the matrix molecules to vaporise into a hot cloud of gas. Through a series of reactions, the glycans become bound to charged species, usually sodium cations (Morelle, 2007). The charged glycans are then sent to a mass analyser. The mass analyser generates a defined electric filed which accelerates the glycans towards a detector. This ‘time of flight’ is determined by the mass-to-charge ratio (m/z) of the glycan. The analyser outputs a mass spectrum, where each peak represents a charged fragment of the original glycan (Morelle, 2009). By comparing the output spectrum to a database of such spectra, the glycan can be identified. The other major MS technique is electrospray ionization (ESI).Instead of embedding the sample in a molecular matrix; the ions are formed directly from solution (Chait, 2011). The solution containing the glycans is forced through a thin capillary tube. At the tip of the tube, a strong electric field is applied. As the glycans leave the tube, they are exposed to this field and ionized. This creates a spray of charged particles. The ions are then detected by a TOF analyser, or other equivalent apparatus (Loo, 2000). MS techniques can be coupled into tandem arrays. These can be used, for example, to first separate a pool of glycans by their mass charge ratio, then selecting a set of glycans within a specific range of (m/z) vales to go on to a second stage where they are fragmented by collision-induced dissociation (CID) and analysed again, providing further structural information (Harvey, 2000). Chromatographic In chromatographic methods, the glycan solution is pumped through a densely-packed stationary phase. The glycans are slowed down on their passage through the column by the stationary phase particles. The samples elute from the column at different times, this is the retention time. The retention time is directly dependant on the glycan mass, so with a properly calibrated column the mass of each glycan can be determined from the retention time. DCU | Analytical methods
  • 20. Within the glycobiology field, several chromatographic techniques have dominated. High pH anion exchange chromatography (HPAEC) has been one of the most widely- used techniques to date (Townsend, 1991). HPAEC is sensitive to branching patterns and oligosaccharide compositions. The glycans bind to the stationary phase which is loaded with negatively-charged functional groups. The glycans are then displaced and eluted by the introduction of a positively charged eluent (Dionex, 1997). Another method used for smaller proteins is reverse-phase high-performance liquid chromatography (RP-HPLC). RP-HPLC uses a non-polar stationary phase. The glycans are derivatised (tagged) with a highly hydrophobic molecule that allows the glycan to be selectively eluted (Wuhrer, 2005). This method can provide detailed information about glycan structure and isomeric configuration (Gillmeister, 2009). Another advantage of HPLC-based systems is that there exist large databases of peak positions for glycans, making identification much easier (Campbell, 2008). Electrophoretic The major electrophoretic method used in the characterization of glycans is capillary electrophoresis (CE). Like all electrophoretic methods, CE separation is based on the size-to-charge ratio of a sample. Protein glyco-isoforms tend to be structurally very similar. The differences may be difficult to detect using chromatographic or spectrometric methods (Zamfir, 2008). The major advantage of CE is that it has unrivalled separation power (Mechref, 2009). The sample is introduced into a chamber where it is taken up by a capillary tube. An electric field is applied over the tube. As the glycans travel through the tube, they are separated out by the slight differences in their mass-to-charge ratios (Landers, 1995). Bioaffinity methods Bioaffinity methods are the newest breakthrough in the field of glycobiology applications. It is defined as the separation of molecules based on their reversible interaction with biological macromolecules. These separations involve highly-specific interactions between the glycoligand and a carbohydrate-binding protein (Tetala, 2010). The advantage of bioaffinity chromatography is that it allows highly-selective one step Analytical methods | DCU
  • 21. purification of glycoproteins, meaning less sample needs to be used. It can be used for analytical, preparatory and diagnostic applications. Lectins are proteins involved in carbohydrate-based recognition. Given their high- specificity for distinct oligosaccharide epitopes, lectins are the logical solution for the stationary-phase ligand. They are able to select for not only overall glycan structure, but also for the configuration of the linkages between the monosaccharide units (Mechref, 2002). This allows them to isolate a target from a pool of glycans or glycoproteins. Several lectin-affinity based techniques have been developed. The first is the standard chromatographic column approach, where the lectins are immobilised onto a stationary phase and the pool of glycoproteins are washed through the column (Tetala, 2010). The target glycoproteins bind to the lectins and can be eluted and fractionated. This approach can be improved by adding multiple lectins to the same column, allowing simultaneous selection of multiple targets at one time (Yang, 2004). Another approach is to immobilize the lectins onto the surface of a microtiter plate. This allows simultaneous analysis of an even greater number of glycoproteins in a rapids and high-throughput fashion (Kuno, 2005). A modification of this approach is the enzyme-linked lectin assay (ELLA) (Wu, 2009). Similar to the standard ELISA assay, the target glycoprotein is bound to the surface of a microtiter plate, before a blocking solution is added to prevent nonspecific binding. Then lectins are added to the wells, and bind to the target glycoprotein, if present. The lectins are detected by the addition of labelled antibodies raised to bind to antigens on the lectin. The label is detected and the quantity of label is proportional to the quantity of target glycoprotein. A similar technique is the carbohydrate array, in which glycans themselves are immobilized to the plate and their interaction with a target protein is assayed (Oyelaran, 2007). DCU | Analytical methods
  • 22. Bioinformatics The primary goal of glycomics is to profile the expression and activity of all of the glycosyltransferases, glycosidases, and other glycosylation apparatus, as well as the entire glycan component within a cell under specific conditions (Aoki-Kinoshita, 2008). The glycoprofiles of cells under different conditions can be compared. From this, we will begin to deduce to conditions which lead to specific glycosylation events and, ultimately, have the ability to rationally design the glycosylation machinery of producer cell lines. Bioinformatic methods will be the key to achieving this goal. Complexity of glycan structures Computationally speaking, glycomics is a much more daunting field than proteomics and genomics. Unlike genes and proteins, the glycoprofile of a cell is not encoded directly by the genome but indirectly through the compliment of glycosylation enzymes active in the cell. This means that to predict the glycosylation state of a newly-translated protein, one would need to have full knowledge of the entire spectrum of glycosylation enzymes expressed at the time as well as their substrate specificities, kinetic rates, their cofactors and inhibitors, and a plethora of other variables. Additionally, glycans themselves are structurally highly complex molecules. Unlike the linear sequence of nucleic acids or amino acids which describe genes and proteins, glycans are composed of sugar monosaccharaides. These can be linked together by different types of bonds, and a residue can be linked to more than one other residue (branching), and each branch can be linked in a number of different ways. Other structural variables include anomeric configuration, epimeric configuration, and reducing terminal attachments (Laine, 1994). These structural traits magnify enormously the number of possible unique structures that can be built from a given set of residues. To put this into perspective, the four DNA bases can give rise to 256 possible four-unit combinations, and the twenty amino acids can give rise to 160,000 possible four-unit arrangements, while a four-unit glycan can potentially be assembled in 15 million different combinations (Von der Leith, 2004). Prediction tools N-glycosylation only occurs at sites which carry the specific Asn-XSer/Thr motif. If this motif occurs in a given peptide sequence, it represents a potential glycosylation site. Bioinformatics | DCU
  • 23. Bioinformaticians have taken advantage of this knowledge to uncover information about the ubiquity of glycoproteins in nature. Zafar et al (2011) used a computer algorithm to scan all of the sequence data available on the ExPASy protein database and flag any sequences which contained the signature motif. They found that more than 50 percent of all proteins (prokaryotic and eukaryotic) contain at least one copy of the motif. This overturns previously held assumptions about the exclusivity of glycosylation machinery to eukaryotes (Nothaft, 2010). A similar experiment conducted by Thanka et al applied statistical analysis to the sequences of 992 experimentally-confirmed O-linked glycoproteins in an effort to discover a signature O-linked motif analogous to the Asn-XSer/Thr motif of N- glycosylation (Christlet, 2001). They found that the presence of a proline residue at either the + or -1 position relative to the serine/threonine site strongly promotes 3 glycosylation and that aromatic amino acids near the site strongly inhibit glycosylation. Glycobiology databases To aid to experiments like these, several online tools and databases have been developed to identify signature motifs for different types of glycosylation within an uploaded sequence (Kamath, 2011). NetNGlyc is an artificial neural network trained not only to find N-glycosylation sequence motifs, but to look at them in the context of the surrounding amino acids whose influence on the local topology and physiochemical properties may affect the glycosylation state of the biding site (Gupta, 2004). Similarly, NetOGlyc parses the local sequence surrounding serine/threonine sites to find probable O-linked glycosylation sites (Julenius, 2005). Several databases have emerged which attempt to catalogue and document glycoproteins. Each has a different specialty. GlycoBase records the HPLC elution data for N-linked glycans (Campbell, 2008), while GlycosuiteDB contains over 3200 unique entries from 245 different species, documenting the glycan structure, peptide linkage type and host protein (Cooper, 2003). O-GlycBase contains detailed information on O- and C- linked glycans and was the dataset used to train the NetOGlyc neural net mentioned above. An enormous amount of data is being produced by glycobiology labs around the world and the bottleneck has now shifted to the computational analysis and interpretation of this data. To fully take advantage of the possibilities that this field offers, it will be necessary to build and utilise new bioinformatics tools, algorithms and databases. DCU | Bioinformatics
  • 24. References Abu-Qarn M (2008). Not just for Eukarya anymore: protein glycosylation in Bacteria and Archaea. Curr Opin Struct Biol. 18(5):544-50. Albert R (1993). SDZ CO 611: a highly potent glycated analog of somatostatin with improved oral activity. Life Sci. 53(6), 517-25. Aoki-Kinoshita KF (2008). An Introduction to Bioinformatics for Glycomics Research. PLoS Comput Biol. 4(5), e1000075. Apweiler R (1999). On the frequency of protein glycosylation, as deduced from analysis of the SWISS-PROT database. Biochim. Biophys. Acta. 1473, 4–8. Arakawa T (2001). Factors affecting short-term and long-term stabilities of proteins. Adv Drug Deliv Rev. 46(1−3), 307–326. Bachmann BJ (1972). Pedigrees of some mutant strains of Escherichia coli K-12. Bacteriol Rev. 36(4), 525–557. Benet L (1984). Pharmacokinetics: Basic Principles and Its Use as a Tool in Drug Metabolism. In: Drug Metabolism and Drug Toxicity (Mitchell JR and Horning MG) Raven Press, New York, 199. Berting A (2010). Virus susceptibility of Chinese hamster ovary (CHO) cells and detection of viral contaminations by adventitious agent testing. Biotechnol Bioeng. 106(4), 598-607. Bethencourt V (2009). Virus stalls Genzyme plant. Nature Biotechnology. 27, 681. Bhatnagar BS (2007). Protein stability during freezing: separation of stresses and mechanisms of protein stabilization. Pharm Dev Technol. 12(5), 505-23. build and utilise new bioinformatics tools, algorithms and databases. Burton D (2006). Sugar Determines Antibody Activity. 313(5787), 627-628. Butler M (2005). Animal cell cultures: recent achievements and perspectives in the production of biopharmaceuticals. Appl Microbiol Biotechnol. 68, 283–91. Campbell M (2008). GlycoBase and autoGU: tools for HPLC-based glycan analysis. Bioinformatics. 24(9), 1214-1216. Campbell MP (2008). GlycoBase and autoGU: tools for HPLC-based glycan analysis. Bioinformatics. 24 (9), 1214-1216 Campion B (1989). Presence of fucosylated triantennary, tetraantennary and pentaantennary glycans in transferrin synthesized by the human hepatocarcinoma cell line Hep G2. Eur J Biochem. 184(2), 405-13. References | DCU
  • 25. Casadevall N (2002). Pure red-cell aplasia and antierythropoietin antibodies in patients treated with recombinant erythropoietin. N. Engl. J. Med. 346(7), 469-75. Ceaglio N (2008). Novel long-lasting interferon alpha derivatives designed by glycoengineering. Biochimie. 90(3), 437–49 Chait BT (2011). Mass Spectrometry in the Postgenomic Era. Annu Rev Biochem. 80, 239-46. Chi EY (2003). Physical stability of proteins in aqueous solution: mechanism and driving forces in non-native protein aggregation. Pharm. Res. 20, 1325–1336. Christlet HT (2001). Database Analysis of O-Glycosylation Sites in Proteins. Biophysical journal. 80(2), 952-960 Cooper C (2003). GlycoSuiteDB: a curated relational database of glycoprotein glycan structures and their biological sources. Nucl. Acids Res. 31(1), 511-513. Cregg JM (2009). Expression in the yeast Pichia pastoris. Methods Enzymol. 463, 169-89. Darling RJ (2002). Glycosylation of erythropoietin affects receptor binding kinetics: role of electrostatic interactions. Biochemistry. 41, 14524-14531. De Schutter K (2009). Genome sequence of the recombinant protein production host Pichia pastoris. Nat Biotechnol. 27(6), 561-6. Demain AL (2009). Production of recombinant proteins by microbes and higher organisms. Biotechnol Adv. 27(3), 297-306. Dionex Corporation (1997) Glycoprotein Oligosaccharide Analysis Using High-performance Anion-Exchange Chromatograph. Dionex Technical Note 42. Egleton RD (2001). Improved blood-brain barrier penetration and enhanced analgesia of an opioid peptide by glycosylation. J. Pharmacol. Exp. Ther. 299(3), 967-72. Egrie JC (1993). The role of carbohydrate on the biological activity of erythropoietin. Glycoconjugate J. 10, 263. Egrie JC (2001). Development and characterization of novel erythropoiesis stimulating protein (NESP). Br. J. Cancer. 1, 3-10. Elliott S (2004). Control of rHuEPO biological activity: the role of carbohydrate. Exp. Hematol. 32, 1146-1155. Fernandes AI (2001). The effect of polysialylation on the immunogenicity and antigenicity of asparaginase: implication in its pharmacokinetics. Int. J. Pharm. 217(1-2), 215-24. Gellison G (1992). High-level expression of foreign genes in Hansenula polymorpha. Biotech Adv. 10, 179–89 DCU | References
  • 26. Gemmill TR (1999). Overview of N- and O-linked oligosaccharide structures found in various yeast species. Biochim. Biophys. Acta. 1426, 227-237. Geyer H (2006). Strategies for analysis of glycoprotein glycosylation. Biochim Biophys Acta. 1764(12), 1853-69. Gillmeister MP (2009). An HPLC-MALDI MS method for N-glycan analyses using smaller size samples: application to monitor glycan modulation by medium conditions. Glycoconj J. 26(9), 1135-49. Goffeau A (1996). Life with 6000 genes. Science. 274(5287), 563-7. Golden K (2009). The XD Process: Development of a High-Titer Process for PER.C6 Cells. 21st Meeting of the European Society of Animal Cell Technology, Dublin, Ireland, 7–10 June 2009. Gupta R (2004). Prediction of N-glycosylation sites in human proteins. [In preparation]. Hamilton SR (2003). Production of complex human glycoproteins in yeast. Science. 301(5637), 1244-6. Hamilton SR (2007). Glycosylation engineering in yeast: the advent of fully humanized yeast. Curr Opin Biotechnol. 18(5), 387-92. Harvey DJ (2000). Collision‐induced fragmentation of underivatized N‐linked carbohydrates ionized by electrospray. J Mass Spectrom. 35(10), 1178-90. Harvey DJ (2005). Structural determination of N-linked glycans by matrix-assisted laser desorption/ionization and electrospray ionization mass spectrometry. Proteomics. 5(7), 1774-86. Hirschler B (2010). World's top-selling drugs in 2014 vs 2010. Thomson Reuters research [Online]. Available from: http://www.reuters.com/article/2010/04/13/roche-avastin-drugs- idUSLDE63C0BC20100413 [Accessed 22 November 2011]. Hoiberg-Nielsen R (2006). Interrelationships of glycosylation and aggregation kinetics for Peniophora lycii phytase. Biochemistry. 45(15), 5057–5066. Hual L (2008). Urea denaturation by stronger dispersion interactions with proteins than water implies a 2-stage unfolding. Proc Natl Acad Sci U S A. 105(44), 16928-33. Huang YM (2010). Maximizing productivity of CHO cell-based fed-batch culture using chemically defined media conditions and typical manufacturing equipment. Biotechnol Prog. 26, 1400–10. Imperiali B (1999). Effect of N-linked glycosylation on glycopeptide and glycoprotein structure. Curr Opin. Chem. Biol. 3(6), 643-9. References | DCU
  • 27. Irwin P (2010). Evidence for a bimodal distribution of Escherichia coli doubling times below a threshold initial cell concentration. BMC Microbiol. 10, 207. Jigami Y (2008). Yeast glycobiology and its application. Biosci Biotechnol Biochem. 72(3), 637- 48. Julenius K (2005). Prediction, conservation analysis and structural characterization of mammalian mucin-type O-glycosylation sites. Glycobiology. 15, 153-164. Kahn CM (2011). Drug Distribution. The Merck Veterinary Manual, 9th Edition. [Online]. Available from: http://www.merckvetmanual.com/mvm/index.jsp?cfile= htm/bc/190110.htm [Accessed 16 November 2011]. Kamath KS (2011). Proteomic databases and tools to decipher post-translational modifications. J Proteomics. [Epub ahead of print]. Kaneko Y (2006). Anti-Inflammatory Activity of Immunoglobulin G Resulting from Fc Sialylation. Science. 313(5787), 670-673. Kanwar YS (1984). Biophysiology of glomerular filtration and proteinuria. Lab. Invest. 51, 7–21. Kayser V (2011). Glycosylation influences on the aggregation propensity of therapeutic monoclonal antibodies. Biotechnology Journal. 6, 38–44. Kelleher DJ (2006). An evolving view of the eukaryotic oligosaccharyltransferase. Glycobiology. 16(4), 47-62. Kerwin BA (2007). Protect from light: photodegradation and protein biologics. J Pharm Sci. 96, 1468–79. Khoury GA (2011). Proteome-wide post-translational modification statistics: frequency analysis and curation of the swiss-prot database. Sci Rep. 13, 1. Kopacek KB (2011). Drug Adsorption. The Merck Manual of Diagnosis and Therapy, 19th Edition [Online]. Available from: http://www.merckmanuals.com/professional/clinical_ pharmacology/pharmacokinetics/drug_ abso rption.html# v1108980 [Accessed 16 November 2011]. Kopacek KB (2011). Drug Distribution. The Merck Manual of Diagnosis and Therapy, 19th Edition [Online]. Available from: http://www.merckmanuals.com/professional/clinical_ pharmacology/pharmacokinetics/drug_ abso rption.html# v1108980 [Accessed 16 November 2011]. Kornfeld R (1985). Assembly of asparagine-linked oligosaccharides. Annu Rev Biochem. 54, 631- 664. DCU | References
  • 28. Kowarik M (2006). Definition of the bacterial N-glycosylation site consensus sequence. EMBO J. 25(9), 1957–1966. Kowarik M (2006). N-Linked glycosylation of folded proteins by the bacterial oligosaccharyltransferase. Science. 314, 1148–1150 Kuno A (2005). Evanescent-field fluorescence-assisted lectin microarray: a new strategy for glycan profiling. Nat. Methods. 2, 851–856. Laine RA. (1994). A calculation of all possible oligosaccharide isomers both branched and linear yields 1.05 3 10(12) structures for a reducing hexasaccharide. Glycobiology. 4: 759–767. Landers JP (1995). Clinical capillary electrophoresis. Clinical Chemistry. 41, 495-509. Lee SY (1996). High cell density culture of Escherichia coli. Trends Biotechnol. 14, 98-105 Li H (2010). Pharmacological significance of glycosylation in therapeutic proteins. Curr Opin Biotechnol. 20(6), 678-84. Lin WJ (2001). DegP-coexpression minimizes inclusion-body formation upon overproduction of recombinant penicillin acylase in Escherichia coli. Biotechnol Bioeng. 73(6), 484-92. Lizak C (2011). N-Linked glycosylation of antibody fragments in Escherichia coli. Bioconjug Chem. 22(3), 488-96. Loo, J. (2000). Electrospray ionization mass spectrometry: a technology for studying noncovalent macromolecular complexes. International Journal of Mass Spectrometry. 200(1-3), 175-186. Macdougall I (1999). Pharmacokinetics of Novel Erythropoiesis Stimulating Protein Compared with Epoetin Alfa in Dialysis Patients. J. Am. Soc. Nephrol. 10, 2392–2395. Manning MC (2010). Stability of protein pharmaceuticals: an update. Pharm Res. 27(4), 544-75. Masarova J (2001). Stability enhancement of Escherichia coli penicillin G acylase by glycosylation with yeast mannan. Biotechnol Appl Biochem. 34, 127–133. Mechref T (2002). Structural Investigations of Glycoconjugates at High Sensitivity. Chem. Rev. 102(2), 321–370 Mechref Y (2009) Glycomic analysis by capillary electrophoresis-mass spectrometry. Mass Spectrom Rev. 28(2):207-22. Moore WV (1980). Role of aggregated human growth hormone (hGH) in development of antibodies to hGH. J. Clin. Endocrinol. Metab. 51(4), 691-7. Morell AG (1968). Physical and chemical studies on ceruloplasmin. V. Metabolic studies on sialic acid-free ceruloplasmin in vivo. J. Biol. Chem. 243(1), 155–9. References | DCU
  • 29. Morell AG (1971). The role of sialic acid in determining the survival of glycoproteins in the circulation. J. Biol. Chem. 246, 1461–1467. Morelle W (2007). Analysis of protein glycosylation by mass spectrometry. Nat. Protoc. 2(7), 1585-602. Morelle W (2009). Analysis of N- and O-linked glycans from glycoproteins using MALDI-TOF mass spectrometry. Methods Mol Biol. 534, 5-21. Nakahara T (2002). Effects of exposure of CHO-K1 cells to a 10-T static magnetic field. Radiology. 224(3), 817-22. Nandita GD (2003). Controlled-release of oral dosage forms. Formulation, Fill and Finish. 10- 16. Nomoto M (1998). Improvement of intestinal absorption of peptide drugs by glycosylation: transport of tetrapeptide by the sodium ion-dependent D-glucose transporter. J. Pharm. Sci. 87(3), 326-32. Nothaft H (2010). Protein glycosylation in bacteria: sweeter than ever. Nature Reviews Microbiology. 8, 765-778. Oyelaran O (2007). Application of carbohydrate array technology to antigen discovery and vaccine development. Expert Rev Vaccines. 6(6), 957-69. Pabst M (2011). Glycan analysis by modern instrumental methods. Proteomics. 11, 631–643 Pandhal J (2010). N-Linked glycoengineering for human therapeutic proteins in bacteria. Biotechnol Lett. 32(9), 1189-98. Perlman S (2003). Glycosylation of an N-terminal extension prolongs the half-life and increases the in vivo activity of follicle stimulating hormone. J. Clin. Endocrinol. Metab. 88(7), 3227-35. Philo JS (2009). Mechanisms of protein aggregation. Curr Pharm Biotechnol. 10, 348–51. Pristov JB (2011). A comparative study of antioxidative activities of cell-wall polysaccharides. Carbohydr Res. 346(14), 2255-9. Rich JR (2009). Emerging methods for the production of homogeneous human glycoproteins. Nat Chem Biol. 5(4), 206-15. Roque AC (2004). Antibodies and genetically engineered related molecules: production and purification. Biotechnol Prog. 20(3), 639-54. Rosenberg AS (2006). Effects of protein aggregates: an immunologic perspective. AAPS J. 8, 501–7. DCU | References
  • 30. Runkel L (1998). Structural and functional differences between glycosylated and non- glycosylated forms of human interferon-beta (IFN-beta). Pharm Res. 15(4), 641–649. Russell D (2009). Site-selective chemical protein glycosylation protects from autolysis and proteolytic degradation. Carbohydr Res. 344(12), 1508-14 Saluja A (2008). Nature and consequences of protein-protein interactions in high protein concentration solutions. International Journal of Pharmaceutics. 358, 1-15 Sasayama S (2000). Glycosylated human interleukin-1alpha, neoglyco IL-1alpha, coupled with N-acetylneuraminic acid exhibits selective activities in vivo and altered tissue distribution. Glycoconj. J. 17(6), 353-9. Satoh M (2006). Non-fucosylated therapeutic antibodies as next-generation therapeutic antibodies. Expert Opin. Biol. Ther. 6(11), 1161-73. Saxena S (1987). Purification and characterization of glucosidase II involved in N-linked glycoprotein processing in bovine mammary gland. Biochem J. 247(3), 563–570. Schirmer EB (2010). Primary clarification of very high-density cell culture harvests by enhanced cell settling. BioProcess Int. 8, 32–9. Sethuraman N (2006). Challenges in therapeutic glycoprotein production. Curr Opin Biotechnol.17, 341–346 Shailubhai K (1987). Purification and characterization of glucosidase I involved in N-linked glycoprotein processing in bovine mammary gland. Biochem J. 247(3), 555–562. Shental-Bechor D (2008). Effect of glycosylation on protein folding: A close look at thermodynamic stabilization. PNAS. 105(24), 8256-8261. Shyama R (2010). Evolutionary Pattern of N-Glycosylation Sequon Numbers in Eukaryotic ABC Protein Superfamilies. Bioinform. Biol. Insights. 4, 9–17. Sivashanmugam A (2009). Practical protocols for production of very high yields of recombinant proteins using Escherichia coli. Protein Sci. 18(5), 936–948. Solá RJ (2007). Modulation of protein biophysical properties by chemical glycosylation: biochemical insights and biomedical implications. Cell Mol Life Sci. 64(16), 2133–2152. Solá RJ (2009). Effects of glycosylation on the stability of protein pharmaceuticals. J Pharm Sci. 98(4), 1223-45. Solá RJ et al (2010). Glycosylation of therapeutic proteins: an effective strategy to optimize efficacy. BioDrugs. 24(1), 9-21. References | DCU
  • 31. Soltero R (2001). The oral delivery of protein and peptide drugs. Innov. Pharmacol. Technol. 1, 106–110 Stanley P (2009). N-Glycans. In: Source Essentials of Glycobiology. (Varki A, Cummings RD, Esko JD, Freeze HH, Stanley P, Bertozzi CR, Hart GW, Etzler ME, eds.). 2nd edition. Cold Spring Harbor (NY): Cold Spring Harbor Laboratory Press, Chapter 8. Sunstrom NA (2000). Regulated autocrine growth of CHO cells. Cytotechnology. 34(1-2), 39- 46. Suzuki M (2006). Bacterial bioreactors for high yield production of recombinant protein. J Biol Chem. 281(49), 37559-65. Sytkowski AJ (1991). Biological activity and structural stability of N-deglycosylated recombinant human erythropoietin. Biochem Biophys Res Commun. 176(2), 698-704. Tams JW (1999). Adapting protein solubility by glycosylation. N-glycosylation mutants of Coprinus cinereus peroxidase in salt and organic solutions. Biochim Biophys Acta. 1432(2), 214– 221. Tang L (2004). Pharmacokinetic aspects of biotechnology products, J. Pharm. Sci. 93, 2184– 2204. Tetala KK (2010). Bioaffinity chromatography on monolithic supports. J Sep Sci. 33(3), 422-38. Townsend RR (1991). Analysis of glycoprotein oligosaccharides using high-pH anion exchange chromatography. Glycobiology. 1(2), 139-47. Uchida E (1997). Effect of active oxygen radicals on protein and carbohydrate moieties of recombinant human erythropoietin. Free Radic Res. 27(3), 311–323. Valdivieso-Ugarte M (2006). Expression of an Aspergillus niger glucose oxidase in saccharomyces cerevisiae and its use to optimize fructo-oligosaccharides synthesis. Biotechnol Prog. 22(4), 1096-101. Vegarud G (1975). The resistance of glycoproteins to proteolytic inactivation. Acta Chem Scand B. 29(8), 887–888. Venter JC (2001). The sequence of the human genome. Science. 291, 1304–1351. Verma R (1998). Antibody engineering: comparison of bacterial, yeast, insect and mammalian expression systems. J Immunol Methods. 216(1-2), 165-81. Vogt G (1997). Protein thermal stability, hydrogen bonds, and ion pairs. J Mol Biol. 269(4), 631- 43. DCU | References
  • 32. Von der Lieth CW (2004). Bioinformatics for glycomics: status, methods, requirements and perspectives. Brief Bioinform. 5(2), 164-78. Wacker M (2002). N-Linked glycosylation in Campylobacter jejuni and its functional transfer into E. coli. Science 298, 1790–1793. Wacker M (2006). Substrate specificity of bacterial oligosaccharyltransferase suggests a common transfer mechanism for the bacterial and eukaryotic systems. Proc. Natl. Acad. Sci. USA. 103, 7088–7093. Walsh G (2010). Biopharmaceutical benchmarks 2010. Nat Biotechnol. 28(9), 917-24. Wang C (1996). Influence of the carbohydrate moiety on the stability of glycoproteins. Biochemistry. 35(23), 7299–7307. Wang W (1999). Instability, stabilization, and formulation of liquid protein pharmaceuticals. Int J Pharm. 185(2), 129–188. Wang W (2005). Protein aggregation and its inhibition in biopharmaceutics. Int J Pharm. 289(1- 2), 1-30. Wormald MR (1999). Glycoproteins: glycan presentation and protein-fold stability. Structure Fold Des. 7(7), 155–160. Wu AM (2009). Lectins as tools in glycoconjugate research. Glycoconj J. 26(8), 899-913. Wuhrer M (2005). Protein glycosylation analysis by liquid chromatography–mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 825(2), 124-33. Xie L (2003). Large-scale propagation of a replication-defective adenovirus vector in stirred-tank bioreactor PER.C6 cell culture under sparging conditions. Biotechnol Bioeng. 83(1), 45-52. Yamane-Ohnuki N (2004). Establishment of FUT8 knockout Chinese hamster ovary cells: an ideal host cell line for producing completely defucosylated antibodies with enhanced antibodydependent cellular cytotoxicity. Biotechnol. Bioeng. 87, 614–622. Yang Z (2004). Approach to the comprehensive analysis of glycoproteins isolated from human serum using a multi-lectin affinity column. J. Chromatogr. A. 1053, 79–88 Yoshimura M (1998). Bisecting GlcNAc structure is implicated in suppression of stroma- dependent haemopoiesis in transgenic mice expressing N-acetylglucosaminyltransferase III. Biochem J. 331(3), 733–742. Young NM (2002). Structure of the N-linked glycan present on multiple glycoproteins in the Gram-negative bacterium, Campylobacter jejuni. J Biol Chem. 277(45), 42530-9 References | DCU
  • 33. Zafar S (2011). Computational analysis reveals abundance of potential glycoproteins in Archaea, Bacteria and Eukarya. Bioinformation. 6(9), 352-5. Zamfir AD (2008). Glycosylation analysis of glycoproteins and proteoglycans using capillary electrophoresis-mass spectrometry strategies. ELECTROPHORESIS. 29, 2485–2507. DCU | References