You are on page 1of 11

Methods 116 (2017) 84–94

Contents lists available at ScienceDirect

Methods
journal homepage: www.elsevier.com/locate/ymeth

Affinity chromatography: A versatile technique for antibody purification


Sushrut Arora a, Vikas Saxena b, B. Vijayalakshmi Ayyar c,⇑
a
Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
b
Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
c
Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA

a r t i c l e i n f o a b s t r a c t

Article history: Antibodies continue to be extremely utilized entities in myriad applications including basic research,
Received 2 November 2016 imaging, targeted delivery, chromatography, diagnostics, and therapeutics. At production stage, antibod-
Received in revised form 16 December 2016 ies are generally present in complex matrices and most of their intended applications necessitate purifi-
Accepted 17 December 2016
cation. Antibody purification has always been a major bottleneck in downstream processing of
Available online 22 December 2016
antibodies, due to the need of high quality products and associated high costs. Over the years, extensive
research has focused on finding better purification methodologies to overcome this holdup. Among a
Keywords:
plethora of different techniques, affinity chromatography is one of the most selective, rapid and easy
Antibody
Antibody purification
method for antibody purification. This review aims to provide a detailed overview on affinity chromatog-
Affinity chromatography raphy and the components involved in purification. An array of support matrices along with various
Affinity matrices classes of affinity ligands detailing their underlying working principles, together with the advantages
Affinity ligands and limitations of each system in purifying different types of antibodies, accompanying recent develop-
ments and important practical methodological considerations to optimize purification procedure are
discussed.
Ó 2016 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
2. Affinity chromatography: principle. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
3. Affinity chromatography: components . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
3.1. Chromatography matrix . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
3.2. Affinity ligands . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
3.2.1. Biospecific ligands . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
3.2.2. Pseudobiospecific ligands. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
3.2.3. Synthetic ligands . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90
3.2.4. Affinity tags. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90
4. Affinity chromatography: practical considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
4.1. Sample preparation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
4.2. Binding. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
4.3. Washing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
4.4. Elution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
5. Conclusions and future perspectives. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92

⇑ Corresponding author.
E-mail address: vijiayyar@gmail.com (B.V. Ayyar).

http://dx.doi.org/10.1016/j.ymeth.2016.12.010
1046-2023/Ó 2016 Elsevier Inc. All rights reserved.
S. Arora et al. / Methods 116 (2017) 84–94 85

1. Introduction specific antigenic determinants (epitopes). In the heavy and light


chains, there are three HVs/CDRs which are connected to each
Antibodies are inducible defensive components of host immune other by four FR regions. Functionally, antibodies are composed
system programmed to identify and neutralize non-self-entities of a fragment antigen binding (Fab), responsible for specific
such as pathogens. Antibodies are specific components of adaptive binding to the antigen; and a fragment crystallizable (Fc), impor-
immune system which not only directly engage in the elimination tant for effector mechanisms, entwining specific recognition of
of foreign particles but also trigger other non-specific effector func- antigen with non-specific effector mechanisms of the host
tions such as antibody-dependent cell-mediated cytotoxicity, immune system (Fig. 1).
complement-dependent cytotoxicity, and antibody-dependent Antibodies are categorized into polyclonal, monoclonal and
cell-mediated phagocytosis to eradicate the threat to host’s recombinant forms. A polyclonal antibody is a mixture of antibod-
homeostasis. ies to a given antigen produced by different B cells. Such a mixture
An antibody is typified by the immunoglobulin G (IgG) contains antibodies with different affinities and specificities to dif-
molecule, the most abundant subclass found in the serum of ferent epitopes on the antigen. Monoclonal antibodies are pro-
mammals [1,2]. IgG finds extensive application in assay develop- duced from a single clone of a B cell, having singular epitope
ment and antibody-based therapeutics [3,4] owing to their specificity. The antibodies or antibody fragments generated
relatively small size and stability during the isolation and purifi- in vitro using molecular techniques are classified as recombinant
cation processes. An IgG is represented as a Y-shaped structure antibodies.
consisting of four polypeptide chains, two heavy chains and Antibodies are being exploited more and more for the obvious
two light chains, connected by disulfide bonds (Fig. 1). These benefits they offer. They are increasingly being utilized in various
heavy and light chains are named based on the difference in scientific endeavors including but not limited to basic research,
their molecular weights. A light chain has a molecular weight chromatography, targeted delivery, imaging, diagnostics and ther-
of 25 kDa whereas a heavy chain has a molecular weight of apeutics [4–14]. Most of these applications require homogeneous
50 kDa. Based on variability in their amino acids, both heavy antibody preparations with high purity. Consequently, there is an
and light chains are further divided into constant (C) and vari- ever-increasing demand for robust, simple, efficient and cost-
able (V) regions. The constant region determines mechanisms effective purification methodologies for antibodies from complex
used to destroy antigen and segregates antibodies into five major mixtures such as plasma, serum, ascites fluid, cell culture medium,
classes (IgM, IgG, IgA, IgD and IgE) depending on their type and egg yolk, plant extracts, and bacterial and yeast cultures [15,16]. To
immune function. Variable region is further subdivided into this end, a variety of chromatographic methodologies based on dif-
hypervariable (HV) and framework (FR) regions. The relatively ferent biochemical properties (e.g. size, solubility, charge,
conserved amino acid sequences make-up the FR region. Con- hydrophobicity and binding affinity) have been tested [15,16].
versely, there is great amount of variability in the sequence of However, affinity chromatography based processes have been
amino acids in HV regions (also known as complementarity reported to be the most efficient and widely employed techniques
determining regions (CDRs)). Combination of different amino owing to their exceptional specificity, ease of operation, yield and
acids in the CDRs confers to antibody its ability to recognize relatively high throughput [16,17].

Fig. 1. Structure of a typical immunoglobulin, IgG. IgG are large molecules of approximately 150–155 kDa containing 2 pairs of heavy and light chains composed of different
domains. The heavy chain consists of a variable domain (VH) and three constant domains (CH1, CH2, and CH3). The two heavy chains are connected by disulfide bonds (SS) in
the hinge region. The light chain has one variable domain (VL) and only one constant domain (CL).
86 S. Arora et al. / Methods 116 (2017) 84–94

2. Affinity chromatography: principle ligand. Additionally, they should be uniformly distributable in a


column, have macroporosity and hydrophilicity along with the sta-
Affinity chromatography is the most selective and versatile bility to withstand various chemical and mechanical stresses. To
form of liquid chromatography [16,18,19] relying on the reversible obtain an ideal matrix material different support materials and
and specific binding between a protein and its cognate ligand, e.g. conjugation chemistries were explored. In 1936, Landsteiner and
binding of a hormone with its receptor, interaction of an enzyme van der Scheer used diazo coupling method to couple haptens to
with its substrate, and binding of an antibody with its target anti- chicken erythrocyte stroma to isolate antibodies that could bind
gen. This specificity of the interaction is used in affinity chro- to the immobilized haptens [28]. Another group, in 1951, used acti-
matography by immobilizing one of the interacting agents, called vated cellulose for the immobilization of bovine serum albumin
‘‘affinity ligand” on to the column as a stationary phase support. (BSA) for the isolation of anti-BSA antibodies from the serum of
Target protein is eluted from a complex mixture by selective BSA immunized rabbit [29]. Subsequently, several groups used
absorption on the affinity ligand. Complex mixture containing tar- binding agents coupled to solid supports such as polyaminostyrene
get protein (e.g. plasma or bacterial culture) is applied in a mobile [30] and glass beads [31], for antigen immobilization for the purifi-
phase that has the right composition and pH for binding of the tar- cation of enzyme or antibodies. However, two major breakthrough
get protein with affinity ligand, while other components pass discoveries changed the way chromatography matrix developed.
through. The retained target is then eluted from the column. Firstly, beaded agarose was developed in 1964 [32] which was fol-
Method of choice for the target elution varies according to the lowed by the development of cyanogen bromide (CNBr) immobi-
affinity of target and ligand interaction. Following elution, column lization method in 1967 [33]. These advances allowed for the
is allowed to regenerate prior to the application of next sample. development of convenient means for covalently coupling peptides
and proteins to the polysaccharide-based materials. CNBr based
coupling method found several usages in affinity chromatography,
3. Affinity chromatography: components
however, due to its toxic nature it became a health hazard for
users. Therefore, several other coupling reagents, such as carbodi-
Affinity chromatography employs different types of binding
imide, tresyl chloride, glutaraldehyde and phosphoryl chloride
agents (ligands) and solid phase supports (matrices). By optimizing
were developed (refer [34]). However, their activation remained
various properties, such as specificity, selectivity, reproducibility,
a challenge. Therefore, pre-activated matrices were developed
conjugation chemistries, and cost-effectiveness of these compo-
and made available commercially [16]. These pre-activated matri-
nents, this method can be applied for the large-scale purification
ces were developed using chemistries involving primary amines,
of antibodies to achieve desired yield and purity of the product.
sulfhydryls, aldehydes, hydroxyls, and carboxylic acids for cou-
Over the years, substantial efforts have been made by scientific fra-
pling ligands to matrices covalently. Based on the origin of com-
ternity on finding or generating quality matrices and affinity
pounds, matrices for use in affinity chromatography were divided
ligands to aid in the development of effective methodologies for
into three classes; natural, synthetic and inorganic (Table 1).
affinity chromatography, with an aim of increased purity and yield
Affinity separation based on the magnetic beads has gained
of the desired antibody in an economic and efficient way.
increased attention in the last two decades. Biocompatible mag-
netic beads were in use since 1970s in chemistry, medicine, and
3.1. Chromatography matrix biotechnology [35]. A detailed review describing their use in pro-
tein and peptide purification had been published earlier [36]. Mag-
A prerequisite for affinity chromatography is a support system netic beads coupled with appropriate ligand-based purification
for the ligands and targets. During early phase of chromatography employ simple steps of mixing and incubating beads with the tar-
method development natural compounds were used both as sup- get sample containing antibody of interest. Magnetic beads having
port and binding agents, mainly in the purification of enzyme. antibodies bound to them are then captured by the separator, fol-
Examples of such efforts include purification of amylase by insol- lowed by washing, and elution of the bound antibodies [36,37].
uble starch [20–24], alginic acid by polygalacturonase [25], pepsin Magnetic beads-based procedure is simple, fast, involves less sheer
by crystal form of edestin [26] and elastase by elastin [27] as sup- pressure, and is applicable to samples with very high protein con-
port and binding agent. In later years, the range of biomolecules of centrations. Therefore, magnetic beads are frequently used in high-
interest expanded, however, availability of ligand and support sys- throughput automation [38]. To prepare magnetic support, mag-
tem from natural sources remained limited. This led to the quest netic particles are entrapped by copolymerization with the poly-
for supporting materials from alternate sources. Desired properties mer, onto which the biomolecules can be immobilized [39].
expected of these supporting materials included exhibition of min- Different kind of supports such as polystyrene, agarose, cellulose,
imum non-specific absorption, insolubility in the buffer system silica, and porous glass can be entrapped onto the magnetic parti-
used, adaptability to the appropriate flow characteristics, availabil- cles of different sizes. Due to their low cost and robustness, mag-
ity of adequate surface area, and easy activation, for the coupling of netic beads are used in a variety of applications and are available
commercially in different formats.
The aforementioned chromatographic matrices can be applied
Table 1 to columns, however, their relatively high cost and a high pressure
Different matrices for use in affinity chromatography.
drop along the column is not appreciated in certain applications.
Matrix Class Examples Commercial Names References Therefore, an alternative method for the bioseparation of large pro-
Natural Agarose Sepharose [33] teins was used in the form of development of affinity membranes.
Dextrans Sephadex and Superdex [181] Affinity membranes offer improved mass-transfer efficiencies, ease
Cellulose beads Cellulose beads [182] of handling, macroporosity, larger surface area and higher dynamic
Synthetic Acrylamide Bio-Gel [183,184] binding capacity [40]. Due to compactness of bed, affinity mem-
Polystyrene Polystyrene [185,186] branes can be used in various sizes and formats with better acces-
Polymethacralate Separon HEMA [187,188]
sibility of ligands for affinity interaction, making them easier to
Inorganic Porous silica Porous silica [189,190] scale-up. Application of affinity membranes is similar to other
Glass fiber Glass fiber [191,192]
matrices described above, wherein membranes are activated for
TiO2 TiO2 [193]
the coupling of affinity ligands, and used for capturing antibodies
S. Arora et al. / Methods 116 (2017) 84–94 87

Table 2
Binding characteristics of different immunoglobulin-binding proteins.

Species Antibody Class Protein A Protein G Protein A/G Protein L§


Human
Total IgG +++ +++ +++ +++
IgG1 +++ +++ +++ +++
IgG2 +++ +++ +++ +++
IgG3 + +++ +++ +++
IgG4 +++ +++ +++ +++
IgM +  + +++
IgD    +++
IgE ++  ++ +++
IgA +  + +++
IgA1 +  + +++
IgA2 +  + +++
Fab + + + +++
scFv +  + +++
Mouse
Total IgG +++ +++ +++ +++
IgG1 + ++ ++ +++
IgG2a +++ +++ +++ +++
IgG2b +++ +++ +++ +++
IgG3 +++ +++ +++ +++
IgM    +++
Rat
Total IgG + ++ ++ +++
IgG1 + ++ ++ +++
IgG2a  +++ +++ +++
IgG2b  + + +++
IgG2b +++ +++ +++ +++
Cow
Total IgG + +++ +++ 
IgG1 + +++ +++ 
IgG2 +++ +++ +++ 
Goat
Total IgG + +++ +++ 
IgG1 + +++ +++ 
IgG2 +++ +++ +++ 
Sheep
Total IgG + +++ +++ 
IgG1 + +++ +++ 
IgG2 +++ +++ +++ 
Horse

Total IgG + +++ +++

IgG(ab) +  +

IgG(c) +  +

IgG(T)  +++ +++
Rabbit
Total IgG +++ +++ +++ +
Guinea pig
Total IgG +++ + +++ +
Pig
Total IgG +++ + +++ +++
Dog

Total IgG +++ + +++
Hamster
Total IgG ++ ++ ++ +++
Donkey

Total IgG ++ +++ +++
Cat

Total IgG +++ + +++
Monkey

Total IgG +++ +++ +++
Chicken
Total IgY    ++

+ = weak binding, ++ = medium binding, +++ = strong binding,  = no binding, * = information not available.
§
Binding occurs only if the appropriate kappa light chains are present. Reproduced with permission from [15].
88 S. Arora et al. / Methods 116 (2017) 84–94

[41]. Affinity membranes find widespread use in high-throughput mammalian serum, binds to mannose and N-acetyl-D-
applications and are available commercially as SwellGel disks. glucosamine residue [51], jacalin, isolated from jackfruit Artocarpus
integrifolia, binds to a-D-galactosyl groups [52]. Lectins are suitable
3.2. Affinity ligands for IgD, IgA and IgM purifications, which are otherwise difficult to
purify using bacterially derived ligands.
Affinity ligands are the principle component of a successful
affinity chromatographic set-up [19]. Typical qualities of a suitable 3.2.1.3. Antigens and anti-antibodies as ligands. Antigen-specific
ligand for affinity chromatography include affinity to the target, antibodies can be purified using whole antigen or specific peptides
specificity, immobilization feasibility, stability in harsh washing as ligands. Alternatively, ligands mimicking structure or sequence
and elution conditions, and retention of target binding capacity of the antigens can also be utilized. This approach leads to the
after attachment to the matrix. A plethora of research has been selection of antibodies with varying affinities, though requiring
conducted to develop and mend these ligands for improved protein further optimization of elution conditions based on the affinity of
purification. However, only a few of these ligands have found con- antibodies to be purified [53]. Similarly, anti-antibodies also offer
sistent use for antibody purification. These ligands are classified in high-specificity options for antibody purification. The constant
three broad groups: biospecific ligands, pseudobiospecific ligands domains of antibodies are used as potential targets for this process.
and synthetic ligands. Additionally, recombinant affinity tags serve However, antibodies have stability issues under harsh pH and high
as alternative ligands for purification of recombinant antibodies. salt concentrations [54], generally required for elution, leading to
leeching of antibodies from column. Single-domain antibodies
3.2.1. Biospecific ligands (sdAb) are especially useful for antibody purification as they have
Biospecific ligands are naturally derived molecules and usually small size, high affinity and stability and a three-dimensional
have high binding affinities to antibodies. This class includes anti- structure that enable binding to novel epitopes [55]. Additionally,
body binding proteins of biological origins, such as bacterially sdAb are easily produced in large scale using microbial systems
derived proteins, antigens, lectins and anti-antibodies. Biospecific and can be tweaked for specificity and affinity depending on the
ligands offer high specificity and selectiveness [42]. type of target. However, sdAb are monomeric, thus, limited in their
binding capacity (owing to their monovalency) during the purifica-
3.2.1.1. Bacterially derived proteins. Bacterially derived proteins tion process.
possess high affinity towards antibodies and, thus, are dominant Biospecific ligands, owing to their specificity and selectivity,
tool for antibody purification. Among these, protein A, G and L have been used extensively for purification of antibodies; however,
are the most commonly used ligands for purification of full- they also have various disadvantages associated with them, mak-
length antibodies. Protein A was isolated from the cell wall of Sta- ing them less attractive. Their manufacturing and processing is
phylococcus aureus and consists of five IgG-binding domains (desig- costly and laborious [56]. Other drawbacks associated with
nated E, D, A, B and C) [36]. Protein A binds to the Fc region of IgG biospecific ligands include low stability, lot-to-lot variability, high
at the junction between its CH2 and CH3 domains [38] and the risk of contamination and toxicity, ligand leakage, difficulty with
heavy chain variable region between CDR2 and CDR3 of VH3 family sterilization, low binding capacities, limited life cycles and low
[43]. Protein G was isolated from C and G groups of Streptococcus scale-up potential [56,57]. Additionally, they are difficult to immo-
spp. and binds strongly to the Fc region of IgG and with low affinity bilize in proper orientation owing to their large size. Consequently,
to the CH1 domain of Fab region [41,44]. In addition to IgG, protein alternative ligands have regularly been researched to overcome
G also binds to albumin, a2 macroglobulin and kinogen [45] which these limitations.
leads to reduced purification efficiencies. Both protein A and G
bind to antibodies from different species, with varied affinities 3.2.2. Pseudobiospecific ligands
based on their subclasses and source (Table 2). While protein A Pseudobiospecific ligands, a group alternative ligands, exploit
binds only to certain species and antibody subclasses; protein G intrinsic properties of the immunoglobulins as they are developed
is broad-spectrum and possess low binding capacity and is less on the basis of multiple non-covalent forces involved in the inter-
stable during harsh elution steps. Over the years recombinantly action of immunoglobulins with affinity ligands. These ligands are
engineered forms of protein A and G, overcoming their drawbacks, promising candidates as they are cheaper, robust, less toxic, struc-
were developed. For example, a genetically engineered protein A turally well defined, and highly stable as they meet the standards
and G fusion protein was developed, combining the IgG binding of good manufacturing protocols (GMPs) involving harsh sanita-
domains of both protein A and G. This fusion protein binds to all tion or sterilization conditions [58,59]. Their affinity is relatively
the human IgG subclasses (Table 2) [46–48]. lower than the biospecific ligands, but it is sufficient enough to
Protein L, isolated from Peptostreptococcus magnus, is another ensure specificity and selectivity towards target antibody [60].
bacterial protein that is useful for antibody purification. Protein L Some of the most common pseudobiospecific ligands used in the
has nanomolar affinity towards kappa (k) light chains of variable purification of antibodies include thiophilic [61,62], hydroxyap-
region of antibody. Though, it binds only to j1, j3, and j4 subclass atite [63], L-histidine [64–66], chelating metal ions [67,68] and
of the antibody light chains, not recognizing j2 and k subgroups mixed mode ligands [69,70]. The development of pseudobiospeci-
and thus, has the ability to bind to the different antibody isotypes fic ligands for the purification of antibodies begun in early nineties
(IgG, IgM, IgY, IgD and IgE) (Table 2) [17]. and since then several products, such as Thiosorb, T-gel, HA-
Ultrogel, and MEP HyperCel were commercialized and used
3.2.1.2. Lectins. Lectins are carbohydrate-binding proteins that bind extensively.
specifically and reversibly to the glycosylation sites on the antibod-
ies rendering them useful for antibody purification. Some of the 3.2.2.1. Thiophilic adsorption chromatography (TAC). TAC is based on
common lectins used for antibody purification include con- the specific propensity of the immunoglobulins to bind with the
canavalin A (Con A), wheat germ agglutinin (WGA), mannan- thioether-substituted organic sulfone compounds, in a reaction
binding protein (MBPs), and jacalin. Con A, isolated from Canavalia termed as thiophilic interaction [71]. TAC is based on the utiliza-
ensiformis, binds to a-D-mannose and a-D-glucose [49,50], wheat tion of a synthetic pseudo-affinity matrix, termed thiophilic gel
germ agglutinin (WGA) binds to the sialic acid and molecules con- (T-gel) [72]. T-gels are the most commonly used adsorbent, and
taining N-acetyl-D-glucosamine residue [50], MBPs, isolated from adsorption depends just on the presence of a kosmotropic salt.
S. Arora et al. / Methods 116 (2017) 84–94 89

T-gels are synthesized by a reaction between divinylsulfone and 2- effective when used along with chloride gradients as it helps in
mercaptoethanol and the chemical formula for an immobilized better removal of host cell proteins, leached protein A, DNA, endo-
ligand can be depicted as agarose-CH2-CH2-SO2-CH2-CH2-SCH2- toxin, and virus [95].
CH2OH [73], indicative of the presence of a linear ligand with
two sulfur atoms. Sulfur atoms provide specificity for selective 3.2.2.3. Hydrophobic charge-induction chromatography (HCIC). The
binding of the immunoglobulins under high salt concentrations. HCIC method is based on the usage of dual-mode ionizable
Due to their small size and high charge density, phosphates and hydrophobic ligands in a pH-dependent manner [96]. As protein
sulfates are the most commonly used salts in the buffer system binding occurs in almost neutral conditions, this method represent
applied in TAC. Composition of the buffer system is determined a near physiological condition for antibody purification. Both thio-
empirically, depending on the free energy of hydrogen bonding philic adsorption chromatography and hydroxyapatite chromatog-
or on the basis of Hofmeister series, so as to avoid protein precip- raphy, require high salt concentrations; whereas, HCIC only
itation in the presence of high salt concentrations [71]. Bound requires variation in pH as the modification of pH of the mobile
immunoglobulins can be eluted by lowering salt concentration, phase induces a net ionic charge on the ligand and the adsorbed
however, salt concentration for the elution of different antibody antibody, leading to the electrostatic repulsion-induced desorp-
formats require optimization to achieve highest recovery of the tion. The 4-mercapto-ethyl-pyridine (MEP) attached via a
antibody in its purest form. Boschetti has reviewed a range of thio- hydrophobic spacer arm to the hydrophilic matrix is a dual-mode
philic ligands elsewhere [62]. ligand of choice for selective adsorption of antibodies. Presence
The simplicity of control over the adsorption behavior as a func- of pyridine ring on MEP provides an added advantage in the sepa-
tion of salt concentration, chemical stability of the matrix, high ration of immunoglobulins [97,98]. When sample is applied with-
protein binding capacity, and affinity toward several classes of out adjustment of ionic strength or pH, the IgG dynamic binding
antibodies has made TAC a method of choice in several applica- capacity ranges from 25 to 35 mg/mL at pH 7–8 (physiological
tions [17]. Thiophilic chromatography has found application in pH). MEP, which is non-charged structure in neutral conditions
the purification of IgG from cows [74], mouse hybridoma [74,75], (pKa 4.8), binds efficiently with antibodies; and under acidic pH
IgG from human and rabbit [76]; IgY from egg yolk [77]; F(ab0 )2 (4.0–4.5) of the mobile phase, a net positive charge is imparted
[78]; and Fab fragment [61], and scFvs [79] from E. coli. on both the ligand and bound target molecules, resulting in the
Besides salt-dependent thiophilic ligands, development of the desorption of the antibody owing to the repulsion forces between
salt-independent mercaptoheterocyclic ligands to capture IgG with the sorbent and positively charged antibody [99]. Due to simplicity
higher adsorption capacity were reported [73]. For instance, Qian of the method, purification and concentration effects are achieved
et al. [80] synthesized thiophilic magnetic polymer beads which in a single step [100]. Chemical properties of MEP render it inert,
were found to have strong specificity against IgG in a salt- thus, not interacting with impurities of the sample; enhancing
independent manner. Magnetic beads were functionalized by using the washing-off of impurities in the flow-through [101]. Taken
a heterocyclic ligand of 2-mercaptonicotinic acid (MNic). Beads together, HCIC is an efficient method of purifying antibodies from
were prepared by microsuspension polymerization with divinyl- samples obtained from diverse sources, such as animal sera, ascites
benzene (DVB) and vinyl acetate (VAc). By using these beads fluid, and cell culture supernatant [84,102–104].
authors were able to purify antibodies, with >94% purity and
>99% bioactivity, from human serum in batch mode and under 3.2.2.4. Immobilized metal affinity chromatography (IMAC). In IMAC,
physiological conditions, with the assistance of magnetic decanta- a metal ion functions as an affinity ligand which is bound to a
tion [80]. chelating agent attached to a support. The adsorption of proteins
is based on the interaction between electron donor groups located
3.2.2.2. Hydroxyapatite chromatography (HAC). Hydroxyapatite on the protein surface and immobilized metal ions [105]. Amino
(HA), (Ca5(PO4)3OH)2, is a naturally occurring compound, and its acid moieties such as histidine, cysteine, serine, glutamate, aspar-
synthetic forms are available both as calcium and phosphate com- tate, and tryptophan present on the outer surface of a biomolecule,
pounds and as an alternative fluorapatite (FA), (Ca5(PO4)3F)2. HA is contain electron donor atoms either in their amino terminus or in
used as porous ceramic microsphere to generate mixed-mode their side chains and, therefore, have higher affinity towards
resin. HA mediates its interaction through (i) P-site, comprising transitional metal ions such as Co2+, Cu2+, Fe2+, Ni2+, and Zn2+
of a triplet of phosphate groups wherein each phosphate with a [106–111]. The ligands for IMAC are designed to exploit this very
pair of negatively charged oxygen residues interacts with amino property of biomolecules in general, and antibodies in particular,
groups of proteins, similar to cation exchangers; (ii) C-site, a dou- wherein ligands are coupled to a covalently linked chelating com-
blet of positively charged calcium residues that mediates calcium pound and spacer group attached to the matrix [112]. Chelating
chelation of protein through carboxyl clusters, and calcium coordi- compounds vary from bidentate (a-amino phenylalanine tetra-
nation on DNA through phosphoryl groups in a metal-affinity type zole) [113], tridentate (iminodiacetic acid, IDA), tetradentate
of mechanism. The P-site and C-site bindings are cooperative; (nitrilotriacetic acid, NTA), to pentadentate (tris (carboxymethyl)
exhibiting positive cooperation with antibodies and negative coop- ethylene diamine) compounds, based on the number of occupied
eration with other solutes. Traditional elution strategies involve coordination bonds.
use of phosphate [81–85] or sodium chloride gradients [81–83]; Several reports indicated use of IMAC for the purification of
however, recently sulfate and borate gradients were also employed antibodies from different species and subclasses [114–123]. Anti-
[83]. Application of HAC in the one-step purification of IgG was bodies purification up to a purity level exceeding 90% in a single
reported to be equally effective as protein A and protein G step can be achieved using IMAC [124,67]. This efficiency emanates
[81,82,86–88]. HAC method was also adopted to support IgA from the cluster of histidine residues present at the junction of the
[63,89,90] and IgM [89,91–93] capture, with capacities ranging second and third constant domains of the heavy chain of an anti-
from 10–20 mg/mL and yields with 90% purity. HAC’s application body [67,115,125]. These histidine moieties endow the antibodies
in removal of antibody aggregate during large-scale production with high affinity towards immobilized metal ions. According to an
for industrial setup has been highlighted [83,94,95]. In these estimate, copper-immobilized IDA can bound up to 14–16 mg/mL
applications, polyethylene glycol (PEG) is recommended. PEG is of polyclonal or monoclonal antibodies [68]. IMAC has been uti-
proposed to enhance aggregate removal as it imposes secondary lized in the purification of IgG from different sources e.g. human
size selectivity on HA [95]. PEG addition is suggested to be most [115], humanized murine IgG [125], and goat IgG [126].
90 S. Arora et al. / Methods 116 (2017) 84–94

IMAC has been the method of choice for the purification of goat, and rabbit. Another screening of a synthetic tetrapeptide
recombinant proteins, especially under denaturing conditions, library targeted against anti-granulocyte macrophage-colony stim-
e.g. inclusion bodies in E. coli expression system, which are nuclear ulating factor (GMCSF) mAb from mouse ascetic fluid resulted in
or cytoplasmic aggregates of protein and require high concentra- the identification of Ala-Pro-Ala-Arg peptide [140]. Similarly, Fas-
tions of chaotropic salts for solubilization. The recombinant anti- sina et al. [141] screened a synthetic multimeric peptide library
bodies could be synthesized so as to have a histidine tag at for purification of IgG from human serum.
amino- or carboxy-terminal of the protein to enhance millimolar Besides screening peptide library, a group of ligands were syn-
affinity towards metal ions. Following chromatographic step, the thesized as biomimetic ligands to extract properties of natural
fusion tag can be efficiently cleaved and removed by proteases ligands. Examples of combinatorial approach in rational design of
[127]. This approach has extensively been utilized in the purifica- ligand came from two different studies [142,143] which synthe-
tion of Fab fragments [128] and scFvs [5,129,130]. Depending on sized ligand mimicking protein sequence of the IgG-Fcc receptor,
the antibody properties, following purification, proper folding can implicated in the binding of proteins to IgG-Fc region for the purifi-
be achieved through a combination of different approaches [131]. cation of mouse and human IgG. Sugita et al. [142] generated an
Guo et al. used an on-column refolding system to obtain functional octameric peptide library using spot-synthesized peptide array
protein from inclusion bodies by fusing single chain Fv (scFv) anti- and identified two peptide sequences by amino acid substitution
body fragment with interferon-c inducible protein 10 (IP10), a che- assay. Similarly, Gong et al. [143] fine-tuned Fc-III peptide, identi-
mokine inducing chemotaxis of activated T- and NK-cells [132]. fied from a phage display screening with a cyclic peptide library
Overall, IMAC is superior to protein A or G methods owing to [144], and synthesized Fc-II-4C peptide containing two disulfide
the robustness of system in terms of IMAC matrices, lower cost, bonds, thus, displaying higher stability and affinity against IgG
and mild elution of proteins with salts [133]. However, proteins from various species. Several other groups had made attempts to
without affinity to metal ions are not applicable to IMAC. Addition- identify small ligands mimicking protein A [145–149].
ally, buffers containing chelating agents, such as EDTA, ammonium
salts, can strip metal ions from resin, posing limitations to IMAC 3.2.3.2. Non-peptidyl ligands. Non-peptidyl ligands, also known as
mediated purifications. non-peptidic ligands (NPL), were initially prepared following
exploration of potential binding site on the target protein. These
3.2.2.5. Boronate affinity chromatography (BAC). The BAC method ligands were developed to overcome weakness of synthetic-
utilizes boronic acid or its derivative as ligands [134]. Boronic acids peptide ligands, which had vulnerable peptide bonds that are fis-
form a stable cyclic ester in alkaline aqueous solution with a com- sile and subject to degradation. The basic element of NPL were
pound containing cis-diol groups. Dissociation of boronic-esters is developed based on chemistries involving peptoids, protease-
achieved by changing medium to an acidic pH. BAC utilizes pres- resistant peptides synthesized by placing replacement at the nitro-
ence of glycans in the Fc region of the antibody for purification gen atom rather than at the a-carbon, or by incorporating ethyl
[135,136]. Recently, Liu et al. [137] prepared a restricted access moiety between the two carbon atoms, thus, assigning novel sec-
boronate affinity porous monolith, as a mimic of protein A for ondary structures to polypeptides [150,151]. Conversely, non-
the specific capture of antibodies. This biomimetic was based on peptidic low molecular weight compounds were chemically syn-
using steric hindrance of the porous monolith and using boronic thesized with high affinity and specificity for IgG. These de novo
acid as specific ligand. BAC holds promise owing to its low cost, synthesized and optimized molecules had very high affinity, dura-
high stability, and fast elution kinetics based on changing pH bility and capacity, with very low cost for compound production.
condition. Examples of NPL include dichloropyridine (AvidAL) [152], thio-
philic gel (divinylsulfone activated agarose) and its derivatives
3.2.3. Synthetic ligands coupled with hetero-aromatic ligands [62,153], and histidyl-
Advancement in high-throughput lead generation and screen- Sepharose matrix [66,154]. Several other small ligands were
ing by high-resolution methods such as X-ray crystallography designed based on the properties of textile dyes as biomimetic
and nuclear magnetic resonance (NMR) has helped in the develop- ligands, by following NPL approach [155]. Among them, chlorotri-
ment of compounds that can overcome weaknesses associated azine based dyes led to the synthesis of de novo designs of several
with natural ligands, such as fragility, and extreme costs in produc- such compounds, e.g. ligand 22/8 synthesized from artificial pro-
tion and procedure optimizations. These new generation com- tein A (ApA) [156,157] and cyanuric chloride [158]. These
pounds are low in molecular weight and more environment triazine-based compounds were further modified and made avail-
friendly due to increased options of their recycling. These com- able commercially as MAbsorbent A1P and A2P. Another group of
pounds are generated either by using a template, based on ratio- NPL compounds derived from dyes, based on their ability to bind
nale design considering its structure and functional aspect or proteins selectively and in a reversible manner, include, Cibacron
using combinatorial approach to identify a template from a library; Blue FG-3A and mono- or di-chloro-triazine (developed by cou-
or a combination of both the approaches by generating a library pling azo, anthraquinone, and phatalocyanin choromophores con-
based on a template [138]. The product generated by this approach jugated with a chlorotriazine ring) [156,157]. The derivation of
could be further modified to obtain desired specificity and affinity these synthetic ligands opened up numerous possibilities of using
and, thus, minimizing efforts in process optimization. The products combinatorial strategies to develop low cost and stable ligands for
generated could be divided into two broad categories based on the the purification of antibodies.
starting material viz. peptidyl and non-peptidyl ligands.
3.2.4. Affinity tags
3.2.3.1. Peptidyl ligand. Amalgamate of different combinations of Affinity tags are short polypeptide sequences, proteins/protein
amino acids in form of a small peptide used in the purification of domains or enzymes, appended as fusion partners with the target
its cognate protein are termed as peptidyl ligands. Yang et al. proteins. These tags have high specificity to different ligands and,
[139] screened a hexamer peptide beads library composed of his- thus, they find considerable use in purification of recombinant
tidine at amino-terminus and positively charged amino acids at antibody formats owing to the associated high yields and purity
carboxy-terminus flanking the aromatic amino acids. This obtained in just a few steps [159,160]. The cDNA of the desired
approach led to the identification of a peptide, His-Trp-Arg-Gly- tag is incorporated either onto the N- or C-terminus of the anti-
Trp-Val, which can purify IgG subclasses from human, cow, mouse, body expressing gene, so that the tag peptide/protein is expressed
S. Arora et al. / Methods 116 (2017) 84–94 91

Table 3 or more soluble components of a homogeneous mixture by form-


Major contaminants in different sources of antibodies. ing a precipitate. Precipitation procedure can be used to reduce
Source Major contaminants the bulk of the material and can either be used to precipitate anti-
Serum Serum proteins, albumin, transferrin, a2- bodies (positive) or other proteins (negative); end product being
macroglobulin, haptoglobulin, ceruloplasmin antibody-enriched sample.
Ascitic fluid Lipids, albumin, transferrin, lipoproteins,
endogenous IgG, other host proteins
Hybridoma Serum proteins, phenol red, water, albumin,
4.2. Binding
transferrin, a2-macroglobulin, haptoglobulin,
ceruloplasmin, bovine IgG, viruses Once the sample is prepared, it is added to the column to bind
Bacterial expression of Proteins from the host bacterial cells and with the ligand. Based on the affinity of antibody to the ligand, spe-
recombinant antibodies growth media
cial considerations, such as slow flow rates and longer incubation
Reproduced with permission from [15]. of samples with the ligands, are often required when purifying
low affinity antibodies. Each ligand has its own specific optimum
binding properties such as temperature, pH, etc. It is necessary to
in the same open reading frame as the antibody, resulting in the carefully consider these conditions for binding buffer preparation
expression of antibodies fused to the tag [160]. Specific ligands to promote maximum binding between the antibody and the
are then used to purify these antibodies fused to the tag. If ligand. Binding can also be promoted by altering salt concentration
required, the fused tag can be removed to generate purified anti- if hydrophobic or ionic interactions are involved. In case of proteins
bodies [160]. containing disulfide bonds, addition of reducing agents such as b-
Various affinity tags are reported with their own sets of mercaptoethanol or dithiothreitol (DTT) can be helpful.
strengths and weaknesses [16,161–164]. An ideal affinity tag will
have the advantage of not disturbing antibody structure and func-
4.3. Washing
tion. Additionally, it should be easily removable from the final pro-
duct, if needed. However, affinity tags might alter affinity and
Once the antibody is allowed to interact with ligand, the col-
specificity, lead to antibody aggregations, cause incorrect folding,
umn is treated with adequate washing buffer to remove all the
are prone to protease degradation and can be difficult to cleave-
non-specific components (unbound or weakly-bound) sticking to
off [165–167]. Polyhistidine tag is the most commonly used tag
the matrix or ligand. Non-specific binding is generally contributed
for antibody purification. Other tags utilized for antibody purifica-
by weak ionic interactions or hydrophobic interactions, which are
tion include FLAG, c-myc, hemagglutinin (HA) and avidin tags. At
usually of low binding strength compared to specific interactions.
times, more than one tag, i.e. combinatorial tagging, offering extra
Binding of non-specific components can be easily dealt by addition
benefits such as enhanced protein solubility and improved purifi-
of salt (NaCl, MgCl2), by altering the pH and/or addition of deter-
cation, can be used [16,160,163].
gents (Tween-20, Triton X-100) to the buffers. Sometimes addition
of blocking agents such as bovine serum albumin or low quantities
4. Affinity chromatography: practical considerations of mimicking agents can be helpful.

Affinity purification of antibodies typically involves 4 steps; i) 4.4. Elution


preparing the sample to load on the affinity column; ii) loading
and incubation of the sample with the ligand to promote binding; Elution can be specific or non-specific depending on the
iii) washing away the non-bound components from the column; method. Specific elution is carried out by adding a mimicking agent
and iv) eluting (recovering) the bound antibodies from the immo- which can either compete for binding with the ligand or the anti-
bilized ligand to collect purified antibodies. Some practical consid- body, thus, displacing antibody from the ligand causing specific
erations associated with these steps are briefly discussed below. elution. Specific elution is a gentle, but slow method. This type of
For detailed protocols please refer [15]. elution requires optimization of the concentration of competitor
a priori to ensure complete displacement of the antibody from
4.1. Sample preparation ligand. For example, application of imidazole for elution in anti-
body purification using Ni-NTA IMAC.
Preparation of starting material is the first step in an affinity Elution performed by changing solvent conditions (pH, ionic
purification procedure. In case of working with mAbs, it is impor- strength and polarity) is referred to as non-specific elution. Non-
tant to identify their isotype to select ligand for efficient purifica- specific elution is faster compared to specific elution, however,
tion. Both ELISA- and lateral flow-based tests are available for due to being harsh, it can cause ligand leaching; limiting the num-
this purpose. Sample characterization to verify if the antibodies ber of times the column can be used. Elution under low pH is the
are expressed extracellularly or intracellularly should be carried most commonly used method among non-specific elution. How-
out to determine the type of extraction and clarification proce- ever, like other proteins, antibodies are also very sensitive to pH
dures to be used. Simple extraction procedures involve osmotic and using very low pH can result in their aggregation. Elution at
shock, French press or sonication to facilitate the release of anti- low temperature or by using additives such as salts (NaCl, MgCl2
bodies extracellularly. If the antibodies are expressed as aggre- or LiCl), guanidine hydrochloride, urea or sometimes organic sol-
gates, disruption by harsh conditions, using denaturing agents vents can be considered to enhance elution.
such as urea and guanidine hydrochloride, can be used.
Sources of antibodies (ascetic fluid, hybridoma supernatant and 5. Conclusions and future perspectives
microbial culture) contain a bulk of other proteins (Table 3) which
may interfere with the purification procedure. These contaminants Downstream processing is the final stage of antibody produc-
can be removed by sample clarification techniques using centrifu- tion that not only determines the quality of the end product but
gation or filtration. Precipitation, using caprylic acid or ammonium also massively impacts the commercial costs. There are many steps
sulfate, is another effective way of clarifying the starting material such as purification, quality assessment, virus testing and efficacy
along with providing enrichment by isolation or removal of one testing that an antibody has to go through before it makes way
92 S. Arora et al. / Methods 116 (2017) 84–94

to the market. Antibody purification serves as a limiting factor in [19] D.S. Hage, J.A. Anguizola, C. Bi, R. Li, R. Matsuda, E. Papastavros, E. Pfaunmiller,
J. Vargas, X. Zheng, J. Pharm. Biomed. Anal. 69 (2012) 93–105.
the antibody production [168] and has been the bottleneck when
[20] E. Starkenstein, Biochem. Z. 24 (1910) 210–218.
it comes to cost reduction [169,170], resulting in increasing the [21] L. Ambard, Bull. Soc. Chim. Biol. 3 (1921) 51–65.
overall costs of the antibody-related products. [22] O. Holmbergh, Biochem. Z. 258 (1933) 134–140.
Advances in commercial antibody market and the growing need [23] Y. Tokuoka, J. Agric. Chem. Soc. Jpn. 13 (1937) 586–594.
[24] D. Hockenhull, D. Herbert, Biochem. J. 39 (1945) 102–106.
for antibody-based therapeutic applications has augmented the [25] H. Lineweaver, R. Jang, E. Jansen, Arch. Biochem. 20 (1949) 137–152.
pressure on industries to reduce the cost and time of deliverables [26] J.H. Northrop, J. Gen. Physiol. 17 (1934) 359–363.
to market. Last five years have witnessed a tremendous surge in [27] N.H. Grant, K.C. Robbins, Arch. Biochem. Biophys. 66 (1957) 396–403.
[28] K. Landsteiner, J. Van der Scheer, J. Exp. Med. 63 (1936) 325–339.
the FDA approved antibody-based therapeutics [4], making anti- [29] D.H. Campbell, E. Luescher, L.S. Lerman, Proc. Natl. Acad. Sci. U.S.A. 37 (1951)
bodies the most promising and fastest growing niches within the 575–578.
biopharmaceuticals sector [171]. But forlornly, there is no such [30] G. Manecke, K.-E. Gillert, Naturwissenschaften 42 (1955) 212–213.
[31] G.B. Sutherland, D.H. Campbell, J. Immunol. 80 (1958) 294–298.
growth evident in the development of downstream processing [32] S. Hjerten, Biochim. Biophys. Acta 79 (1964) 393–398.
methodologies, which is lagging way behind, even after the mark- [33] R. Axen, J. Porath, S. Ernback, Nature 214 (1967) 1302–1304.
edly higher requirements for robust and scalable large scale purifi- [34] J.H. Luong, W.H. Scouten, Curr. Protoc. Protein Sci. (2008). Chapter 9, Unit 9.3.
[35] P.L. Kronick, G.L. Campbell, K. Joseph, Science 200 (1978) 1074–1076.
cation processes [172]. The downstream processing alone accounts [36] I. Safarik, M. Safarikova, Biomagn. Res. Technol. 2 (2004) 7.
for 50–80% of the total manufacturing costs of the therapeutics [37] D. Wilkinson, Scientist 14 (2000) 25–28.
antibodies [53,173]. It is noteworthy that development of [38] R.S. Sista, A.E. Eckhardt, V. Srinivasan, M.G. Pollack, S. Palanki, V.K. Pamula,
Lab. Chip 8 (2008) 2188–2196.
antibody-based therapeutics requires purified antibodies at
[39] M. Koneracka, P. Kopčanský, M. Timko, C. Ramchand, A. De Sequeira, M.
numerous additional stages and not just at the final stage of their Trevan, J. Mol. Catal. B Enzym. 18 (2002) 13–18.
production [174], which expectedly leads to remarkably high sell- [40] M. Teeters, S. Conrardy, B. Thomas, T. Root, E. Lightfoot, J. Chromatogr. A
ing costs of these therapeutics. (2003) 165–173.
[41] H. Sun, B. Ge, S. Liu, H. Chen, J. Sep. Sci. 31 (2008) 1201–1206.
Over the years, protein A-based purification of antibodies has [42] Z. Liu, P.V. Gurgel, R.G. Carbonell, Biotechnol. Prog. 29 (2013) 91–98.
been the stalwart technology for industrial antibody purification [43] C. Boi, J. Chromatogr. B 848 (2007) 19–27.
[174,175], as majority of commercial antibodies have been either [44] S.P. Chambers, Drug Discov. Today 7 (2002) 759–765.
[45] P.J. Haney, C. Draveling, W. Durski, K. Romanowich, M. Walid Qoronfleh,
monoclonal or polyclonal [176]. It is unmistakable apparent that Protein Expr. Purif. 28 (2003) 270–279.
it will require a big leap to move from tested and established chro- [46] J.J. Marchalonis, J.L. Atwell, J.W. Goding, Immunology 34 (1978) 97–103.
matography methods that have been the mainstay of antibody [47] S. Kabir, Immunol. Invest. 31 (2002) 263–278.
[48] O. Corey, H. David, B. Raychelle, B. Min, W. Chunling, K. Elizabeth, Bioaffinity
purification for many decades. Consequently, a lot of recent work chromatography, in: D.S. Hage, J. Cazes (Eds.), Handbook of Affinity
has been directed at improving protein A for stability and binding Chromatography, 2nd ed., CRC Press, 2005, pp. 101–126.
capacity [177,178]. However, the entry of recombinant antibodies [49] K. Kwack, J. Biochem. Mol. Biol. 33 (2000) 188–192.
[50] T. Ishihara, T. Kadoya, H. Yoshida, T. Tamada, S. Yamamoto, J. Chromatogr. A
in the market is surely going to dent this dominance, though not 1093 (2005) 126–138.
in near future. Additionally, there are augmented efforts concen- [51] A.C.A. Roque, C.R. Lowe, M.Â. Taipa, Biotechnol. Prog. 20 (2004) 639–654.
trated at increasing the throughput of antibody purification, pro- [52] K. Hagiwara, D. Collet-Cassart, K. Kunihiko, J.-P. Vaerman, Mol. Immunol. 25
(1988) 69–83.
viding additional benefits of robustness, automation, and time-
[53] B. Chackerian, L. Briglio, P.S. Albert, D.R. Lowy, J.T. Schiller, J. Virol. 78 (2004)
and cost-effectiveness [172,174,179,180]. However, these trials 4037–4047.
are still in their infancy and have a lot of challenges to address [54] A. Wörn, A. Plückthun, J. Mol. Biol. 305 (2001) 989–1010.
[172,179] before they find extensive implementation. [55] A.L. Horenstein, I. Durelli, F. Malavasi, Purification of clinical-grade
monoclonal antibodies by chromatographic methods, in: C.M. Smales, D.C.
James (Eds.), Therapeutic Proteins, Humana Press, 2005, pp. 191–208.
[56] S. Sheng, F. Kong, Pharm. Biol. 50 (2012) 1038–1044.
Conflict of interest
[57] J.N. Arnold, R.A. Dwek, P.M. Rudd, R.B. Sim, Immunol. Lett. 106 (2006) 103–
110.
The authors declare no conflict of interest. [58] J. Champagne, C. Delattre, C. Shanthi, B. Satheesh, L. Duverneuil, M.A.
Vijayalakshmi, Chromatographia 65 (2007) 639–648.
[59] R.R. Prasanna, A.S. Kamalanathan, M.A. Vijayalakshmi, J. Mol. Recognit. 28
References (2015) 129–141.
[60] C.R. Lowe, J.C. Pearson, Methods Enzymol. 104 (1984) 97–113.
[1] S.E. Salmon, B.A. Smith, J. Clin. Invest. 49 (1970) 1114–1121. [61] M. Fiedler, A. Skerra, Protein Expr. Purif. 17 (1999) 421–427.
[2] B.L. Larson, H.L. Heary Jr., J.E. Devery, J. Dairy Sci. 63 (1980) 665–671. [62] E. Boschetti, J. Biochem, Biophys. Methods 49 (2001) 361–389.
[3] I. Correia, mAbs 2 (2010) 221–232. [63] E. Luellau, U. von Stockar, S. Vogt, R. Freitag, J. Chromatogr. A 796 (1998) 165–
[4] B.V. Ayyar, S. Arora, R. O’Kennedy, Trends Pharmacol. Sci. 37 (2016) 1009– 175.
1028. [64] S.M. Bueno, K. Haupt, M.A. Vijayalakshmi, J. Chromatogr. B Biomed. Appl. 667
[5] B.V. Ayyar, S. Hearty, R. O’Kennedy, Anal. Biochem. 407 (2010) 165–171. (1995) 57–67.
[6] K. Welbeck, P. Leonard, N. Gilmartin, B. Byrne, C. Viguier, S. Arora, R. [65] K. Haupt, S.M. Bueno, M.A. Vijayalakshmi, J. Chromatogr. B Biomed. Appl. 674
O’Kennedy, J. Immunol. Methods 364 (2011) 14–20. (1995) 13–21.
[7] V. Saxena, C.K. Lai, T.C. Chao, K.S. Jeng, M.M. Lai, J. Virol. 86 (2012) 4139–4150. [66] Q. Luo, H. Zou, Q. Zhang, X. Xiao, J. Ni, Biotechnol. Bioeng. 80 (2002) 481–489.
[8] J.A.G.L. van Buggenum, J.P. Gerlach, S. Eising, L. Schoonen, R.A.P.M. van Eijl, S. [67] D. Todorova-Balvay, O. Pitiot, M. Bourhim, T. Srikrishnan, M. Vijayalakshmi, J.
E.J. Tanis, M. Hogeweg, N.C. Hubner, J.C. van Hest, K.M. Bonger, K.W. Mulder, Chromatogr. B Analyt. Technol. Biomed. Life Sci. 808 (2004) 57–62.
Sci. Rep. 6 (2016) 22675. [68] R.R. Prasanna, M.A. Vijayalakshmi, J. Chromatogr. A 1217 (2010) 3660–3667.
[9] S.N. Rylova, L. Del Pozzo, C. Klingeberg, R. Tönnesmann, A.L. Illert, P.T. Meyer, [69] V. Brenac Brochier, A. Schapman, P. Santambien, L. Britsch, J. Chromatogr. A
H.R. Maecke, J.P. Holland, J. Nucl. Med. 57 (2016) 96–102. 1177 (2008) 226–233.
[10] F. Bootz, B. Ziffels, D. Neri, Inflamm. Bowel Dis. 22 (2016) 2098–2105. [70] S.S. Ranjini, D. Bimal, A.P. Dhivya, M.A. Vijayalakshmi, J. Chromatogr. B Analyt.
[11] H.J. Kim, M.H. Jeong, H.J. Park, W.C. Kim, J.E. Kim, Food Chem. 196 (2016) Technol. Biomed. Life Sci. 878 (2010) 1031–1037.
1144–1149. [71] J. Porath, M. Belew, Trends Biotechnol. 5 (1987) 225–229.
[12] L. Xue, T. Hickling, R. Song, J. Nowak, B. Rup, Clin. Exp. Immunol. 183 (2016) [72] J. Porath, F. Maisano, M. Belew, FEBS Lett. 185 (1985) 306–310.
102–113. [73] H. Lakhiari, D. Muller, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 818
[13] B.V. Ayyar, K.R. Aoki, M.Z. Atassi, Infect. Immun. 83 (2015) 1465–1476. (2005) 53–59.
[14] P. Delfani, L. Dexlin Mellby, M. Nordström, A. Holmér, M. Ohlsson, C.A.K. [74] P. Konecny, R.J. Brown, W.H. Scouten, J. Chromatogr. A 673 (1994) 45–53.
Borrebaeck, C. Wingren, PLoS One 11 (2016) e0159138. [75] U.B. Finger, W. Brummer, E. Knieps, J. Thommes, M.R. Kula, J. Chromatogr. B
[15] S. Arora, B.V. Ayyar, R. O’Kennedy, in: N.E. Labrou (Ed.), Protein Downstream Biomed. Appl. 675 (1996) 197–204.
Processing: Design, Development and Application of High and Low- [76] A. Lihme, P.M. Heegaard, Anal. Biochem. 192 (1991) 64–69.
Resolution Methods, Humana Press, Totowa, NJ, 2014, pp. 497–516. [77] P. Hansen, J.A. Scoble, B. Hanson, N.J. Hoogenraad, J. Immunol. Methods 215
[16] B.V. Ayyar, S. Arora, C. Murphy, R. O’Kennedy, Methods 56 (2012) 116–129. (1998) 1–7.
[17] A.C.A. Roque, C.S.O. Silva, M.Â. Taipa, J. Chromatogr. A 1160 (2007) 44–55. [78] G.K. Yurov, G.L. Neugodova, O.A. Verkhovsky, B.S. Naroditsky, J. Immunol.
[18] R.R. Walters, Anal. Chem. 57 (1985) 1099A–1101A. 1102A-1106A passim. Methods 177 (1994) 29–33.
S. Arora et al. / Methods 116 (2017) 84–94 93

[79] K. Trisler, L.L. Looger, V. Sharma, M. Baker, D.E. Benson, S. Trauger, P.G. [133] G. Serpa, E.F. Augusto, W.M. Tamashiro, M.B. Ribeiro, E.A. Miranda, S.M.
Schultz, V.V. Smider, J. Biol. Chem. 282 (2007) 26344–26353. Bueno, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 816 (2005) 259–
[80] H. Qian, C. Li, Z. Lin, Y. Zhang, Colloids Surf. B Biointerfaces 75 (2010) 342– 268.
348. [134] P. Muhammad, D. Li, Z. Liu, Encyclopedia of Analytical Chemistry, John Wiley
[81] R. Giovannini, R. Freitag, Biotechnol. Bioeng. 73 (2001) 522–529. & Sons Ltd, Chichester, 2006, pp. 1–18.
[82] S. Schubert, R. Freitag, J. Chromatogr. A 1142 (2007) 106–113. [135] L. Borlido, A.M. Azevedo, A.C. Roque, M.R. Aires-Barros, J. Chromatogr. A 1218
[83] P. Gagnon, Nat. Biotechnol. 25 (2009) 287–293. (2011) 7821–7827.
[84] L. Guerrier, I. Flayeux, E. Boschetti, J. Chromatogr. B. Biomed. Sci. Appl. 755 [136] A.M. Azevedo, A.G. Gomes, L. Borlido, I.F. Santos, D.M. Prazeres, M.R. Aires-
(2001) 37–46. Barros, J. Mol. Recognit. 23 (2010) 569–576.
[85] A. Jungbauer, R. Hahn, K. Deinhofer, P. Luo, Biotechnol. Bioeng. 87 (2004) [137] Y. Liu, Y. Lu, Z. Liu, Chem. Sci. 3 (2012) 1467–1471.
364–375. [138] Y.D. Clonis, J. Chromatogr. A 1101 (2006) 1–24.
[86] L.H. Stanker, M. Vanderlaan, H. Juarez-Salinas, J. Immunol. Methods 76 (1985) [139] H. Yang, P.V. Gurgel, R.G. Carbonell, J. Pept. Res. 66 (2005) 120–137.
157–169. [140] S.A. Camperi, N.B. Iannucci, G.J. Albanesi, M. Oggero Eberhardt, M.
[87] A. Jungbauer, C. Tauer, M. Reiter, M. Purtscher, E. Wenisch, F. Steindl, A. Etcheverrigaray, A. Messeguer, F. Albericio, O. Cascone, Biotechnol. Lett. 25
Buchacher, H. Katinger, J. Chromatogr. 476 (1989) 257–268. (2003) 1545–1548.
[88] C. Poiesi, A. Tamanini, S. Ghielmi, A. Albertini, J. Chromatogr. 465 (1989) 101– [141] G. Fassina, A. Verdoliva, G. Palombo, M. Ruvo, G. Cassani, J. Mol. Recognit. 11
111. (1998) 128–133.
[89] K. Aoyama, J. Chiba, J. Immunol. Methods 162 (1993) 201–210. [142] T. Sugita, M. Katayama, M. Okochi, R. Kato, T. Ichihara, H. Honda, Biochem.
[90] H. Leibl, R. Tomasits, H.M. Wolf, M.M. Eibl, J.W. Mannhalter, J. Chromatogr. B Eng. J. 79 (2013) 33–40.
Biomed. Appl. 678 (1996) 173–180. [143] Y. Gong, L. Zhang, J. Li, S. Feng, H. Deng, Bioconjug. Chem. 27 (2016) 1569–
[91] G. Coppola, J. Underwood, G. Cartwright, M.T. Hearn, J. Chromatogr. 476 1573.
(1989) 269–290. [144] W.L. DeLano, M.H. Ultsch, A.M. de Vos, J.A. Wells, Science 287 (2000) 1279–
[92] A.J. Henniker, K.F. Bradstock, Biomed. Chromatogr. 7 (1993) 121–125. 1283.
[93] E. Fishbein, D. Alarcon-Segovia, J. Immunol. Methods 33 (1980) 93–99. [145] B. D’Agostino, P. Bellofiore, T. De Martino, C. Punzo, V. Rivieccio, A. Verdoliva,
[94] P. Gagnon, K. Beam, Curr. Pharm. Biotechnol. 10 (2009) 440–446. J. Immunol. Methods 333 (2008) 126–138.
[95] P. Gagnon, J. Immunol. Methods 336 (2008) 222–228. [146] H. Yang, P.V. Gurgel, R.G. Carbonell, J. Chromatogr. A 1216 (2009) 910–918.
[96] S.C. Burton, D.R. Harding, J. Chromatogr. A 814 (1998) 71–81. [147] K. Sakamoto, Y. Ito, T. Hatanaka, P.B. Soni, T. Mori, K. Sugimura, J. Biol. Chem.
[97] S. Oscarsson, J. Porath, J. Chromatogr. 499 (1990) 235–247. 284 (2009) 9986–9993.
[98] K.L. Knudsen, M.B. Hansen, L.R. Henriksen, B.K. Andersen, A. Lihme, Anal. [148] L.N. Lund, P.-E. Gustavsson, R. Michael, J. Lindgren, L. Nørskov-Lauritsen, M.
Biochem. 201 (1992) 170–177. Lund, G. Houen, A. Staby, P.M. St. Hilaire, J. Chromatogr. A 1225 (2012) 158–
[99] G. Zhao, Y. Sun, J. Chromatogr. A 1165 (2007) 109–115. 167.
[100] W. Schwart, D. Judd, M. Wysocki, L. Guerrier, E. Birck-Wilson, E. Boschetti, J. [149] S. Menegatti, A.D. Naik, P.V. Gurgel, R.G. Carbonell, J. Sep. Sci. 35 (2012)
Chromatogr. A 908 (2001) 251–263. 3139–3148.
[101] E. Boschetti, Trends Biotechnol. 20 (2002) 333–337. [150] T. Clackson, J.A. Wells, Trends Biotechnol. 12 (1994) 173–184.
[102] R.Z. Wang, D.Q. Lin, H.F. Tong, H.L. Lu, S.J. Yao, J. Chromatogr. B Analyt. [151] H.U. Saragovi, M.I. Greene, R.A. Chrusciel, M. Kahn, Biotechnology N.Y. 10
Technol. Biomed. Life Sci. 936 (2013) 33–41. (1992) 773–778.
[103] L. Guerrier, P. Girot, W. Schwartz, E. Boschetti, Bioseparation 9 (2000) 211– [152] T.T. Ngo, N. Khatter, J. Chromatogr. A 597 (1992) 101–109.
221. [153] F. Anspach, D. Petsch, W. Deckwer, Bioseparation 6 (1996) 165–184.
[104] A. Schwarz, F. Kohen, M. Wilchek, J. Chromatogr. B Biomed. Appl. 664 (1995) [154] A. El-kak, M.A. Vijayalakshmi, J. Chromatogr. B Biomed. Sci. Appl. 570 (1991)
83–88. 29–41.
[105] J. Porath, J. Carlsson, I. Olsson, G. Belfrage, Nature 258 (1975) 598–599. [155] C.R. Lowe, S.J. Burton, N.P. Burton, W.K. Alderton, J.M. Pitts, J.A. Thomas,
[106] P.G. Taylor, C. Martinez-Torres, E.L. Romano, M. Layrisse, Am. J. Clin. Nutr. 43 Trends Biotechnol. 10 (1992) 442–448.
(1986) 68–71. [156] R. Li, V. Dowd, D.J. Stewart, S.J. Burton, C.R. Lowe, Nat. Biotechnol. 16 (1998)
[107] J.H. Swain, L.B. Tabatabai, M.B. Reddy, J. Nutr. 132 (2002) 245–251. 190–195.
[108] S. Storcksdieck genannt Bonsmann, R.F. Hurrell, J. Food Sci. 72 (2007) S019– [157] S.F. Teng, K. Sproule, A. Husain, C.R. Lowe, J. Chromatogr. B Biomed. Sci. Appl.
S029. 740 (2000) 1–15.
[109] J. Porath, Protein Expr. Purif. 3 (1992) 263–281. [158] S.F. Teng, K. Sproule, A. Hussain, C.R. Lowe, J. Mol. Recognit. 12 (1999) 67–75.
[110] E. Sulkowski, BioEssays 10 (1989) 170–175. [159] D.W. Wood, Curr. Opin. Struct. Biol. 26 (2014) 54–61.
[111] F.H. Arnold, Biotechnology N.Y. 9 (1991) 151–156. [160] X. Zhao, G. Li, S. Liang, J. Anal. Methods Chem. 2013 (2013) 581093.
[112] F. Helfferich, Nature 189 (1961) 1001–1002. [161] U.D. Palanisamy, A. Hussain, S. Iqbal, K. Sproule, C.R. Lowe, J. Mol. Recognit.
[113] G. Lei, L. Liu, X. Xiong, Y. Wei, X. Zheng, J. Sep. Sci. 31 (2008) 3002–3008. 12 (1999) 57–66.
[114] S. Vançan, E.A. Miranda, S.M.A. Bueno, Process Biochem. 37 (2002) 573–579. [162] A.C.A. Roque, M.Â. Taipa, C.R. Lowe, J. Chromatogr. A 1064 (2005) 157–167.
[115] J. Porath, B. Olin, Biochemistry 22 (1983) 1621–1630. [163] G. Gupta, C.R. Lowe, J. Mol. Recognit. 17 (2004) 218–235.
[116] G. Serpa, E.F.P. Augusto, W.M.S.C. Tamashiro, M.B. Ribeiro, E.A. Miranda, S.M. [164] D. Dong, H. Liu, Q. Xiao, R. Li, J. Chromatogr. B 870 (2008) 51–54.
A. Bueno, J. Chromatogr. B 816 (2005) 259–268. [165] M.L. Isaksen, K. Fitzgerald, in: R. Kontermann, S. Dübel (Eds.), Antibody
[117] D. Das, T.M. Allen, M.R. Suresh, Protein Expr. Purif. 39 (2005) 199–208. Engineering, Springer, Berlin Heidelberg, Berlin, Heidelberg, 2001, pp. 282–
[118] T. Zimmerman, C. Petit Frère, M. Satzger, M. Raba, M. Weisbach, K. Döhn, A. 291.
Popp, M. Donzeau, J. Immunol. Methods 314 (2006) 67–73. [166] A. Goel, D. Colcher, J.-S. Koo, B.J.M. Booth, G. Pavlinkova, S.K. Batra, Biochim.
[119] S. Martins, A. Karmali, J. Andrade, M.L. Serralheiro, Mol. Biotechnol. 33 (2006) Biophys. Acta 1523 (2000) 13–20.
103–113. [167] H. Schmeisser, P. Kontsek, D. Esposito, W. Gillette, G. Schreiber, K.C. Zoon, J.
[120] D. Platis, J. Drossard, R. Fischer, J.K.C. Ma, N.E. Labrou, J. Chromatogr. A 1211 Interferon Cytokine Res. 26 (2006) 866–876.
(2008) 80–89. [168] F. Li, N. Vijayasankaran, A. Shen, R. Kiss, A. Amanullah, mAbs 2 (2010) 466–
[121] M.B. Ribeiro, M. Vijayalakshmi, D. Todorova-Balvay, S.M.A. Bueno, J. 479.
Chromatogr. B 861 (2008) 64–73. [169] P. Gronemeyer, R. Ditz, J. Strube, Bioengineering 1 (2014) 188–212.
[122] S. Jain, M.N. Gupta, Biotechnol. Appl. Biochem. 39 (2004) 319–322. [170] G.E. Khoury, B. Khogeer, C. Chen, K.T. Ng, S.I. Jacob, C.R. Lowe, Pharm.
[123] K. Ramessar, T. Rademacher, M. Sack, J. Stadlmann, D. Platis, G. Stiegler, N. Bioprocess. 3 (2015) 139–152.
Labrou, F. Altmann, J. Ma, E. Stoger, T. Capell, P. Christou, Proc. Natl. Acad. Sci. [171] D.M. Ecker, S.D. Jones, H.L. Levine, mAbs 7 (2015) 9–14.
U.S.A. 105 (2008) 3727–3732. [172] J. Welsh, Pharm. Bioprocess. 3 (2015) 1–3.
[124] R.C. Cheung, J.H. Wong, T.B. Ng, Appl. Microbiol. Biotechnol. 96 (2012) 1411– [173] C.R. Lowe, Curr. Opin. Chem. Biol. 5 (2001) 248–256.
1420. [174] P.M. Schmidt, M. Abdo, R.E. Butcher, M.-Y. Yap, P.D. Scotney, M.L. Ramunno,
[125] J.E. Hale, D.E. Beidler, Anal. Biochem. 222 (1994) 29–33. G. Martin-Roussety, C. Owczarek, M.P. Hardy, C.-G. Chen, L.J. Fabri, J.
[126] V. Boden, J.J. Winzerling, M. Vijayalakshmi, J. Porath, J. Immunol. Methods Chromatogr. A 1455 (2016) 9–19.
181 (1995) 225–232. [175] G.R. Bolton, K.K. Mehta, Biotechnol. Prog. 32 (2016) 1193–1202.
[127] P.A. Walker, L.E. Leong, P.W. Ng, S.H. Tan, S. Waller, D. Murphy, A.G. Porter, [176] M. Hornsby, M. Paduch, S. Miersch, A. Sääf, T. Matsuguchi, B. Lee, K.
Biotechnology N.Y. 12 (1994) 601–605. Wypisniak, A. Doak, D. King, S. Usatyuk, K. Perry, V. Lu, W. Thomas, J. Luke, J.
[128] Y. Zhao, L. Gutshall, H. Jiang, A. Baker, E. Beil, G. Obmolova, J. Carton, S. Goodman, R.J. Hoey, D. Lai, C. Griffin, Z. Li, F.J. Vizeacoumar, D. Dong, E.
Taudte, B. Amegadzie, Protein Expr. Purif. 67 (2009) 182–189. Campbell, S. Anderson, N. Zhong, S. Gräslund, S. Koide, J. Moffat, S. Sidhu, A.
[129] B. McDonnell, S. Hearty, W.J.J. Finlay, R. O’Kennedy, Anal. Biochem. 410 Kossiakoff, J. Wells, Mol. Cell. Proteomics 14 (2015) 2833–2847.
(2011) 1–6. [177] R. Godawat, K. Brower, S. Jain, K. Konstantinov, F. Riske, V. Warikoo,
[130] S. Sakamoto, F. Taura, R. Tsuchihashi, W. Putalun, J. Kinjo, H. Tanaka, S. Biotechnol. J. 7 (2012) 1496–1508.
Morimoto, Hybridoma 29 (2010) 481–488. [178] J. Pollock, G. Bolton, J. Coffman, S.V. Ho, D.G. Bracewell, S.S. Farid, J.
[131] R. Vincentelli, S. Canaan, V. Campanacci, C. Valencia, D. Maurin, F. Frassinetti, Chromatogr. A 1284 (2013) 17–27.
L. Scappucini-Calvo, Y. Bourne, C. Cambillau, C. Bignon, Protein Sci. 13 (2004) [179] V. Girard, N.-J. Hilbold, C.K.S. Ng, L. Pegon, W. Chahim, F. Rousset, V.
2782–2792. Monchois, J. Biotechnol. 213 (2015) 65–73.
[132] J.Q. Guo, Q.M. Li, J.Y. Zhou, G.P. Zhang, Y.Y. Yang, G.X. Xing, D. Zhao, S.Y. You, [180] J. Spooner, T. Wilkinson, B.P. Kemp, Pharm. Bioprocess. 3 (2015) 411–424.
C.Y. Zhang, Protein Expr. Purif. 45 (2006) 168–174. [181] J. Turková, Journal of Chromatography Library, Elsevier, 1993.
94 S. Arora et al. / Methods 116 (2017) 84–94

[182] J. Štamberg, Sep. Purif. Methods 17 (1988) 155–183. [188] M.W.H. Roberts, C.M. Ongkudon, G.M. Forde, M.K. Danquah, J. Sep. Sci. 32
[183] Z. Jedliński, J. Paprotny, J. Polym. Sci. A Polym. Chem. 5 (1967) 2957–2960. (2009) 2485–2494.
[184] A. Verdoliva, F. Pannone, M. Rossi, S. Catello, V. Manfredi, J. Immunol. [189] K.K. Unger, Journal of Chromatography Library, Elsevier, 1979.
Methods 271 (2002) 77–88. [190] F. Xi, J. Wu, J. Chromatogr. A 1057 (2004) 41–47.
[185] E. Klein, E. Eichholz, D.H. Yeager, J. Membr. Sci. 90 (1994) 69–80. [191] G.C. Serafica, G. Belfort, J. Pimbley, Biotechnol. Bioeng. 43 (1994) 21–36.
[186] C. Staak, F. Salchow, P.H. Clausen, E. Luge, J. Immunol. Methods 194 (1996) [192] M. Schuster, E. Wasserbauer, A. Neubauer, A. Jungbauer, Bioseparation 9
141. (2000) 259–268.
[187] O. Wichterle, D. Lim, Nature 185 (1960) 117–118. [193] Y. Li, H.G. Spencer, J. Biotechnol. 26 (1992) 203–211.

You might also like