Antigen Generation and Display in Therapeutic Antibody Drug Discovery - A Neglected But Critical Player

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

ISSN 1860-6768 · BJIOAM 7 (12) 1421–1532 (2012) · Vol.

7 · Dezember 2012

Systems & Synthetic Biology ·


Nanobiotech · Medicine

12/2012 Cover illustration


Biopharmaceuticals
Purification
Protein engineering
Antibodies

Special issue Biopharmaceuticals


Drug discovery and production are highly interlinked, because the main product
attributes need to be defined early on. This Special issue edited by Alois Jung-
bauer (University of Natural Resources and Life Sciences, Vienna, Austria) and
Klaus Graumann (Sandoz GmbH, Kundl, Austria) covers several areas relevant
to the discovery, development and manufacturing of biopharmaceuticals.
Topics include antibodies, protein engineering and purification.
Images: Antibodies © Elena Pankova – Fotolia.com; Biopharmaceutical Produc-
www.biotechnology-journal.com
tion © Sandoz.

Biotechnology Journal – list of articles published in the December 2012 issue.


Review Research Article
Antigen generation and display in therapeutic antibody Periodic counter-current chromatography – design and
drug discovery – a neglected but critical player operational considerations for integrated and continuous
Hilmar Ebersbach and Sabine Geisse purification of proteins
http://dx.doi.org/10.1002/biot.201200066 Rahul Godawat, Kevin Brower, Sujit Jain, Konstantin
Konstantinov, Frank Riske and Veena Warikoo
Mini-Review http://dx.doi.org/10.1002/biot.201200068
Current strategies in antibody engineering: Fc engineering
and pH-dependent antigen binding, bispecific antibodies Technical Report
and antibody drug conjugates Endotoxin removal and prevention for pre-clinical biologics
Karen J. Vincent and Mauro Zurini production
http://dx.doi.org/10.1002/biot.201200250 Anne Serdakowski London, Brendan Kerins,
William R. Tschantz and Kasey Mackay
Review http://dx.doi.org/10.1002/biot.201200220
Structure-function studies with G protein-coupled receptors
as a paradigm for improving drug discovery and development Technical Report
of therapeutics A high-throughput assay for enzymatic polyester hydrolysis
Patrick M. McNeely, Andrea N. Naranjo and activity by fluorimetric detection
Anne S. Robinson Ren Wei, Thorsten Oeser, Susan Billig and Wolfgang
http://dx.doi.org/10.1002/biot.201200076 Zimmermann
http://dx.doi.org/10.1002/biot.201200119
Review
The sweet tooth of biopharmaceuticals: Importance Perspective
of recombinant protein glycosylation analysis How can measurement, monitoring, modeling and control
Nico Lingg, Peiqing Zhang, Zhiwei Song and advance cell culture in industrial biotechnology?
Muriel Bardor Manuel J. T. Carrondo, Paula M. Alves, Nuno Carinhas,
http://dx.doi.org/10.1002/biot.201200078 Jarka Glassey, Friedemann Hesse, Otto-Wilhelm Merten,
Martina Micheletti, Thomas Noll, Rui Oliveira, Udo Reichl,
Review Arne Staby, Ana P. Teixeira, Henry Weichert and
Minimizing immunogenicity of biopharmaceuticals Carl-Fredrik Mandenius
by controlling critical quality attributes of proteins http://dx.doi.org/10.1002/biot.201200226
Miranda M.C. van Beers and Muriel Bardor
http://dx.doi.org/10.1002/biot.201200065

Review
Multimodal chromatography: An efficient tool in downstream
processing of proteins
Kristian Kallberg, Hans-Olof Johansson and Leif Bulow
http://dx.doi.org/10.1002/biot.201200074

© 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.biotechnology-journal.com


Biotechnol. J. 2012, 7, 1433–1443 DOI 10.1002/biot.201200066 www.biotechnology-journal.com

Review

Antigen generation and display in therapeutic antibody drug


discovery – a neglected but critical player

Hilmar Ebersbach1 and Sabine Geisse2


1 NBC/NT, Novartis Institutes for BioMedical Research, Basel, Switzerland
2 NBC/PPA, Novartis Institutes for BioMedical Research, Basel, Switzerland

Disease intervention by targeting a critical pathway molecule through a blocking antibody or


Received 08 JUN 2012
interference by therapeutic proteins is currently en vogue. Generation of blocking antibodies or Revised 26 AUG 2012
therapeutic proteins inevitably requires the production of recombinant proteins or cell-based Accepted 25 SEP 2012
immunogens. Thus, one could call the antigen molecule the neglected player in antibody drug dis-
covery. The variety of methods available for making recombinant proteins or recombinant cell lines
that present the target on the cell surface is extensive. These need to be addressed in conjunction
with biochemical and biophysical quality criteria and the experimental application intended. Fun-
damentally, successful production and isolation of monoclonal antibodies requires optimized
antigen preparation and presentation to the immune host. This review summarizes the most im-
portant aspects of antigen generation and display, enabling logical decision making to give rise to
potent high-affinity antibodies.

Keywords: Antigen preparation · Expression systems · Immunization · Phage display · Quality parameters

1 Introduction preparation and its presentation because the underlying


main principle of immunization is the presentation of the
Recently, considerable progress has been made in gener- antigen to the immune host in a format that elicits the
ating full antibodies, fragments thereof, or other function- strongest and, at the same time, most specific immune
ally blocking molecules based on various scaffolds as bio- response. Therefore, the quality, integrity, and folding
pharmaceuticals, and the different routes to achieve this state of an antigen protein are essential parameters for
have been extensively discussed in the public domain. successful antibody generation and its presentation
However, none of the reviews [1–5] recently compiled should mimic the situation encountered in vivo as close-
mention the generation of antigen material in sufficient ly as possible.
quantity and quality; a crucial aspect that is clearly neg- Yet, the nature and inherent characteristics of target
lected. molecules selected for detection or intervention by anti-
Fundamental to the successful isolation of a mono- bodies or related molecules vary considerably, and like-
clonal antibody (mAb) or binding molecule is antigen wise, the formats and production modes can vary as well.
In this review, several approaches to antigen generation
are discussed and summarized with particular attention
Correspondence: Dr. Hilmar Ebersbach, NBC/NT, Novartis Institutes for paid to their application and product quality.
BioMedical Research, Novartis Campus, WSJ-506.313A, CH-4002 Basel,
Switzerland
E-mail: Hilmar.Ebersbach@novartis.com 2 Soluble antigen generation by chemical
synthesis or recombinant expression
Abbreviations: CHO, Chinese hamster ovary; FACS, fluorescence-activated
cell sorting; GPCR, G-protein coupled receptor; HEK293, human embryon-
ic kidney cell line; LAL, limulus amoebocyte lysate; mAb, monoclonal anti- The vast majority of antigens currently under investiga-
body; SEC, size-exclusion chromatography; MALS, multiangle light scatter- tion comprise soluble protein entities, such as full-length
ing proteins (such as cytokines), sub-domains of soluble pro-

© 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim 1433


Biotechnology Biotechnol. J. 2012, 7, 1433–1443
Journal
www.biotechnology-journal.com

teins, or full extracellular domains of cell-surface recep- from inclusion body isolation and solubilization to refold-
tors (such as G-protein coupled receptors, GPCRs). Tech- ing techniques, such as chromatographic methods, mem-
niques such as chemical peptide synthesis as well as the brane controlled denaturant removal, or direct dilution in
entire repertoire of recombinant protein expression tech- conjunction with optimization of redox conditions in the
nologies in prokaryotic and eukaryotic hosts can be case of disulfide-bonded proteins.
applied to generate these molecules and are summarized Improved refolding strategies, the use of enhancers
in the following sections. Two considerations should such as low-molecular-weight additives, and the deter-
govern the decision for one expression technology over mination of optimum refolding parameters by coupling
another: (i) the nature of the molecule to be expressed dynamic and static light scattering methodologies has
and (ii) its biochemical and biophysical characteristics. driven recent progress and achievements, as document-
The yield should be a homogenous, properly folded and ed impressively in multiple reviews [13–18].
displayed antigen such that the critical epitopes are ac- Likewise, the solubility of the antigen protein can be
cessible. enhanced by fine-tuning of expression parameters, rang-
ing from plasmid construct design, selection of E. coli host
2.1 Synthetic peptide antigens strain, to modulation of process parameters [19].
For purification of soluble recombinant proteins
Synthetic peptides are often used as immunogens, in that expressed in E. coli, classical methods based on the
they represent surrogates for protein domains or even physico-chemical properties of the molecule (such as size,
complete proteins. This approach is applied in hybridoma charge, hydrophobicity, solubility, or binding affinity) are
generation, for example, because it is fast, straightfor- applied. Certain techniques, such as hydrophobic inter-
ward, and comes into play when the required amount of action, ion exchange or size exclusion chromatography,
soluble, functionally active antigen is difficult to produce enable individual proteins to be purified from a crude
by recombinant expression technologies. Moreover, for source. The biological affinity of a protein interacting with
cell-surface receptors, representing a class of highly a specific partner, such as enzyme–substrate, ligand–
interesting targets for antibody intervention, peptide receptor, and antibody–antigen interactions, can be
antigen mimics have been successfully used in immu- exploited for affinity chromatography. Alternatively, artifi-
nization or phage display [6, 7]. Yet, the selection of suit- cial N- or C-terminal extensions to the gene of interest can
able peptides derived from cell-surface receptors provides be introduced into the molecule to serve as affinity purifi-
challenges. Apart from immunogenic attributes, they also cation tags. Comprehensive reviews of affinity tags in the
need to have the appropriate conformational properties to context of bacterial expression were recently compiled by
enable selection of mAbs capable of recognizing the Young et al. [20] and Walls and Loughran [21].
native antigen displayed on the cell surface, in particular, As an example, polyhistidine tags are very common,
if the retrieval of functionally blocking antibodies is easy to use, and usually result in relatively pure protein
envisaged. Detailed information on all aspects that should samples [22, 23]. “Pseudo-affinity” resins contain bound
be considered when working with peptide antigens was metal ions, either nickel or cobalt, to which the polyhisti-
nicely summarized in a recent review by Trier et al. [8]. dine tag binds with micromolar affinity. High concentra-
tions of imidazole, EDTA or buffers with low pH in the
2.2 Recombinant antigen expression in bacteria sample elution fraction can be exchanged against a phys-
iological buffer by dialysis or application of size exclusion
Recombinant protein expression in Escherichia coli chromatography as a subsequent purification step.
strains is undoubtedly the fastest and most cost-effective
approach to protein generation, but frequently results in 2.3 Recombinant expression in eukaryotic systems
the formation of inclusion bodies. Here, the protein of in-
terest accumulates in high concentration and purity, but E. coli-derived antigens, lacking the natural sugar
is insoluble and non-functional. If the antigen exceeds residues attached by glycosylation, may give rise to anti-
small to medium size, features multiple disulfide bonds, or bodies directed against exposed epitopes that are not
needs to undergo other secondary modifications essential accessible in the mammalian system and hence are of lit-
for function, the choice of a bacterial expression system tle value [24]. In contrast, yeast expression systems using,
may thus be suboptimal [9, 10]. Periplasmic expression by for example, the strain Saccharomyces cerevisiae as a
co-expression of folding chaperones has shown some host may yield hyper-glycosylated proteins, which in turn
benefits in this respect; specific release from only the could lead to masking of immunogenic epitopes [25],
periplasm is achieved by osmotic shock or leakage [11, unless a yeast strain modified by metabolic engineering
12]. towards a human glycosylation pattern is employed [26,
Moreover, remarkable progress has been made in the 27].
successful refolding of complex proteins from inclusion Infecting insect cell lines, such as Spodoptera frugi-
bodies. The refolding process comprises several steps perda (Sf) or Trichoplusia ni (T.ni) derivatives, with re-

1434 © 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim


Biotechnol. J. 2012, 7, 1433–1443

www.biotecvisions.com

combinant baculoviruses is very effective for the produc- turing tags is often the method of choice to allow for its re-
tion of recombinant proteins of an intracellular, cell-sur- trieval from the rather complex composition of cell culture
face-bound, or secreted nature [28–30]. Insect cells, in media. A variety of small to large tags, such as HA-tag,
comparison to mammalian cells, are easy to handle and FLAG-tag, his6-tag, and Fc-fusions, have been described.
cultivate and require less complex and thus less expen- Small tags usually remain attached to the recombinant
sive culture media and cell culture equipment. Moreover, protein and binders raised against these entities during
the efficient infectious process and short run time facili- the primary antibody selection process require a subse-
tates the generation of multiprotein complexes, either quent counter-selection step for de-selection. Large tags
from a mixture of single recombinant baculoviruses by co- can also be removed by an additional cleavage and purifi-
infection, or from the so-called MultiBac system [31]. cation step, if a protease cleavage site is introduced into
Insect cells are capable of modifying proteins by phos- the cDNA construct (e.g. PreScission, enterokinase,
phorylation, fatty acid acylation, N-terminal acetylation, thrombin) [21, 46, 47].
carboxymethylation, isoprenylation, and N- and O-glyco- In some instances, a capturing tag on the antigen may
sylation. Yet, an insect-specific N-glycosylation pattern of impair correct folding and/or biological activity. In these
the high-mannose type has led to efforts in pathway cases, a tagless protein can be produced and retrieved by
engineering of insect cells to exhibit a human-like glyco- affinity chromatography, providing that a suitable anti-
sylation pattern [32, 33]. body against the target is available in sufficient quantity.
Rapidly growing interest in the development of bio- To conclude this section on soluble expression of anti-
pharmaceuticals is paralleled by a steep rise in recombi- gens, there is increasing interest to tackle complex multi-
nant protein production using mammalian cells as hosts, domain antigens in different indications. Each domain is
not only for manufacturing purposes, but also to supply capable of possessing a separate independent function
the discovery process with high-quality proteins, such as and three-dimensional structure and could harbor func-
antigens. For the rapid generation of material on a scale tional epitopes. Both isolated individual expression of sin-
of 1–100 mg of purified protein, transient expression in gle domain entities and expression of the full-length
the human embryonic kidney cell line (HEK293) and Chi- extracellular domain can be exploited for antigen genera-
nese hamster ovary (CHO) cells has therefore become the tion. However, the determination of the protein domain
method of choice in recent years [34–36]. In many in- boundaries as an extension or deletion of a single amino
stances, these host cell lines have been engineered by acid is critical and may result in soluble expression in rea-
stable integration of viral elements derived from sonable yields. Unfortunately, the respective entries in rel-
Epstein–Barr virus or simian virus 40 (SV40), which, in evant databases may be incorrect and ill-described at
conjunction with the corresponding interacting virus-de- times [48].
rived elements incorporated into the expression plasmid,
give rise to high-level expression. For both host cell lines, 2.4 Cell-surface displayed antigens
detailed protocols for suspension cultivation of cells under
serum-free conditions and transfection by lipoplexes and 2.4.1 Naturally presented antigens
polycations on a small to large scale have been worked out Instead of defined polypeptides, naturally occurring
and described [37–40]. whole cells can be used for immunization and in vitro
The availability of disposable bioreactors, such as the display; an approach especially useful for the identifica-
WAVE™ system [41], along with inexpensive transfection tion and characterization of novel cell-surface biomark-
reagents, such as polyethylenimine (PEI) [42], has enabled ers. For example, normal or cancerous human cells are
transient protein production up to a scale of >100 L at a potent immunogens for the generation of mAbs, result-
reasonable price, giving rise to protein yields in the multi- ing in a strong immune response in animals [49–51]. For
milligram to gram range [43, 44]. The impact of this might cell-based immunization, cells can be collected from tis-
not be obvious, since only small amounts of antigen are sue culture or isolated directly from tissue samples or
required to initiate the antibody generation process, but blood samples, such as blood mononuclear cells. With
advanced antibody drug candidates in preclinical or clin- this approach, a prerequisite for success is the high
ical phases still rely on antigen supply in increasing overexpression of the target molecule versus all other
amounts at the highest quality for manufacturing of the cell-surface entities displayed. Additionally, isogenic
product [45]. Furthermore, the expression process and control cells expressing low or no target molecule are
scale can be flexibly tailored to expression levels and needed for counter-selection. Overall, extensive screen-
requested amounts of an individual protein. ing must be successful in isolating highly specific bind-
Applying techniques paralleling those used for pro- ing molecules when non-recombinant technologies are
teins derived from bacteria is possible, provided that the applied [52, 53].
antigen is secreted from cells and the protein is subse- A further specialized technique for antibody genera-
quently captured from the culture medium of insect or tion, which uses naturally occurring and presented anti-
mammalian cells. Tagging of the protein with various cap- gens, is in vivo phage display, for which antibody libraries

© 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim 1435


Biotechnology Biotechnol. J. 2012, 7, 1433–1443
Journal
www.biotechnology-journal.com

are injected into animals. The phage enriched for binding larly, expression of a second detector molecule, such as
to the target is then recovered by isolation of cells or even green fluorescent protein, or an unrelated short receptor
tissues of interest. This technique is often used for spe- fragment, either by direct fusion to the molecule of inter-
cific cancer cell targets [54]. est or by co-expression, has been tried successfully
[57–59].
2.4.2 Recombinant antigens presented on cell surface Commonly used cell lines for recombinant receptor
To circumvent the inefficiency of screening for mAbs that expression comprise HEK293 and CHO derivatives, but to
recognize the native cell-displayed antigens, receptor maximize the specific antibody response in the mouse
molecules can be overexpressed recombinantly. If the in- upon whole-cell immunization, human target antigens
tended application involves cell-based screening, a tran- can also be cloned into and overexpressed on the surface
sient expression approach may be less desirable due to of mouse cell lines derived from a strain syngenic to the
the heterogeneity of expression frequently encountered immunization host strain, for example, BALB/c mice. The
in these cell pool populations. Furthermore, if repeated stably transfected cells are used for immunizing the syn-
cell batches are required, the inherent inter-experimental genic mouse strain and subsequent screening of hybrido-
variation observed in transient expression trials could be ma clones derived from cell fusions, while the non-trans-
problematic. Novel transfection strategies with very high fected cell line serves as the negative control (Fig. 1). In
transfection rates, such as large-scale electroporation contrast to human cells, recombinant murine cells will
using the MaxCyte™ device [55] combined with a proto- typically exhibit a significantly reduced background
col for freezing of transiently transfected cell batches [56], response when using the syngenic inbred mouse strain
are potential means to overcome these problems. because the cells are genetically identical to the mouse
Alternatively, the generation of stable episomal cell strain apart from the transfected and expressed target
pools can be applied. Subsequent to transfection, cell pop- molecule. In addition, it is also possible to use cell-based
ulations exhibiting resistance to commonly used selection immunization to generate mAbs against the secreted
antibiotics, such as hygromycin or geneticin, are estab- forms of antigens by tethering the secreted proteins, such
lished. To circumvent any lengthy and tedious single-cell as serum proteins and cytokines, to the cell surface as
cloning procedure, enriched fractions of positive recombi- antigens for immunization.
nant pools can be isolated by fluorescence-based cell sort- A condensed overview for the selection of recombi-
ing (FACS). Two to three rounds of FACS enrichment usu- nant expression hosts in relation to the antigen to be pro-
ally give rise to nicely positive, albeit not fully homoge- duced is given in Fig. 2. To conclude, awareness at the
nous, cell populations. A caveat to this approach is the beginning of a discovery project should be raised that
necessity to monitor the expression stability of the usually more than one antigen needs to be prepared per
enriched pool populations over time. Moreover, the avail- project. Depending on the sequence homology of the tar-
ability of a detection antibody capable of recognizing a na- get between human, rodents, and monkeys, antigens of
tive epitope and thus suitable for immunofluorescence all species may be required for cross-reactivity assess-
staining and cell sorting may pose a constraint, in partic- ment. Notably, even if a protocol for successful expression
ular, for novel targets. In cases where no detection anti- of a human antigen has been worked out, this may not
body is available against the target protein, if the attach- translate into similarly successful production of the relat-
ment of a small peptide detection tag, such as the his6 tag, ed, but not sequence-identical cynomolgus or mouse pro-
at the N terminus of the receptor is not desired, or the N tein. Furthermore, paralogue antigens for counter-screen-
and C termini of the receptor molecule reside intracellu- ing and selection should also be included in planning.

Figure 1. Transfected cells as antigens


for immunization and subsequent mAb
screening. A human target antigen is
overexpressed on the surface of a mouse
cell line derived from the BALB/c strain.
The stable transfected cells are used as
an antigen for immunizing BALB/c mice
and for the differential screening of
hybridoma supernatants of the transfect-
ed versus non-transfected cells by flow
cytometry. Expression profiles of an anti-
gen on transfected and control cells are
shown.

1436 © 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim


Biotechnol. J. 2012, 7, 1433–1443

www.biotecvisions.com

Figure 2. Which approach to choose for which


type of antigen? In an abbreviated format, an
overview of the different choices for expression
hosts, depending on the type of protein and its
sub-cellular localization, is shown. Dashed
arrows indicate potentially less preferable
choices. BEVS, baculovirus expression vector
system; ORF, open reading frame.

2.4.3 Complex membrane proteins as antigens from the parental cell line. It is also very easy to produce
Isolation of soluble membrane proteins, such as most “null” particles in an identical parental cell line not ex-
GPCRs, following recombinant expression in either mam- pressing the target antigen for counter-selection purpos-
malian or bacterial systems remains a challenging task. es. Exosomes, which are microparticles naturally shed by
The identification of functional binding molecules, a variety of cell types, can also be engineered towards
regardless of whether they are agonistic or antagonistic, presentation of antigens [64–66].
requires expression and purification of GPCRs in a func- Finally, cell-free expression offers an interesting alter-
tional state. The amount of recombinant membrane- native method to produce proteins that lead to cell toxic-
inserted receptors produced depends on the optimal com- ity in other systems. Potential translation directly into
bination of a particular GPCR and the production host, membrane mimetics is beneficial for membrane proteins.
which needs to be determined experimentally. In addition Further stabilization of the expressed proteins can be
to classical membrane preparation from overexpressing achieved by almost unlimited supplementation of cofac-
cells, receptors can be extracted from membranes by the tors, ligands, or chaperones to the translation reaction
use of detergents. The choice of an appropriate detergent mixture. Recent developments have resulted in well-
for solubilization and purification is crucial for retaining established systems of prokaryotic and eukaryotic origins
the functionality of the receptor. Many GPCRs become [67–69].
unstable upon detergent extraction from lipid membranes
and their quality should be controlled as described below, 2.4.4 DNA Immunization
for example, by ligand binding assays [60–62]. For the sake of completeness, an approach that does not
Alternatively, Lebon et al. [63] described a method to rely on a preformulated proteinaceous antigen directly
engineer a GPCR gene by point mutagenesis to obtain should be mentioned. An additional strategy for dealing
sufficient amounts of soluble and thermostable trans- with difficult to express antigens is to use a DNA immu-
membrane protein for crystallization trials and biochemi- nization approach. This method exploits the flexibility
cal analysis. In fact, thermostabilized transmembrane and ease of working with plasmid DNA, allowing the gen-
proteins obtained by protein engineering are a valuable eration of many different DNA constructs encoding pep-
antigen source and might be the method of choice if oth- tides, protein domains, or full-length proteins. DNA can
er trials for antigen preparation are unsuccessful. also be delivered by several different means, including
Another option for antigen presentation as an injection (hypodermic needle), a gene gun using DNA-
immunogen (especially suited for membrane proteins) is coated gold beads, pneumatic injection, and lipid formu-
the generation of virus-like particles, as shown for cell re- lations [70]. It is thought that the resulting antigenic pep-
ceptors and ion channels. A number of different ap- tides are presented by antigen presenting cells, such as
proaches exist, however, they all follow the same princi- dendritic cells, and elicit both a strong humoral and cellu-
ple of co-overexpression of the antigen with a virus coat lar immune response [71, 72].
protein for encapsulation of the antigen into so-called The main limitation of this approach is the resulting
virus-like particles. The main benefit of this approach over lack of available protein antigen for subsequent screening
merely transfected cells is a higher expression or concen- purposes. Thus, combination with the approaches
tration of the antigen combined with a better defined sur- described above using cells, which endogenously express
face, that is, reduced presentation of irrelevant proteins the protein of interest, or even more recent techniques

© 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim 1437


Biotechnology Biotechnol. J. 2012, 7, 1433–1443
Journal
www.biotechnology-journal.com

employing next-generation sequencing have to be con- 4 Quality-control criteria and methodologies


sidered [73, 74]. Sequencing of the complete antibody applied
repertoire versus the sequencing results obtained from a
naïve animal may lead to the identification of highly spe-
cific binders albeit at low frequency. Careful monitoring of protein quality with respect to iden-
tity, integrity, and functionality is essential to ensure the
use of antigens, which mimic natural presentation and
3 Chemical modification of antigens provide the highest chance for isolation of binding mole-
for immunization and screening cules with the desired specificity. The analytical methods
applied are highly diverse and cover a broad range with
The generation of mAbs to peptide antigens by classical regard to technology as well as equipment. The most
immunization usually requires coupling of the peptide to important parameters to be assessed are summarized
a carrier protein, for example, keyhole limpet hemocyanin below.
or BSA. Likewise, in vitro display systems require the use First, an exact determination of protein concentration
of carrier proteins due to the small size of peptide anti- is a crucial parameter for affinity determination of binding
gens. The coating of the plastic surface of microtiter molecules towards purified antigen. Apart from classical
plates with unconjugated peptides renders them masked methods, such as Bradford and Lowry, Nanodrop offers a
and inaccessible during blocking of free binding sites of fast analysis technique with minimal sample consump-
the plastic surface with BSA or milk. tion by measurement of the absorbance at 280 nm with a
Protein antigens can also be biotinylated either site- known extinction coefficient. Alternatively, HPLC pro-
specifically at the C or N terminus by attachment of the vides data on protein quantity and purity, but clearly has
so-called Avi-tag to the cDNA [75] or through endoge- limitations for glycosylated proteins. Another convincing
nously present lysines by random biotinylation. Con- method, which combines analysis of quantity and purity,
versely, chemically derived peptides can be conjugated is LabChip (Caliper Life Sciences, Zurich, Switzerland).
during synthesis. This allows the addition of antigen to Here SDS-PAGE-like size separation is electronically eval-
displayed binding molecules and, following a standard uated to obtain concentrations of individual protein
panning cycle, facilitates capture of the full binding com- bands based on an internal reference standard.
plex via streptavidin or NeutrAvidin. This serves as an Likewise, confirmation of the protein identity should
alternative strategy for antigen presentation apart from be a routinely performed analysis and can be achieved by
direct coating onto plastic surfaces [76, 77]. different methods. Most commonly used and powerful is
For immunization peptide or protein antigens are usu- HPLC coupled to mass spectrometry (LC-MS), which sep-
ally formulated with an adjuvant. The use of an adjuvant, arates proteins on a reverse-phase column following
such as Freund’s adjuvant, not only lowers the amount of analysis of protein mass by TOF electrospray ionization
antigen required, but also prolongs the antigen stimula- (TOF-MS). Evaluation of individual results can confirm
tion of the immune host. Even though in recent years the identity, purity, and integrity of the protein, but also
there has been a growing number of new adjuvant allow identification of post-translational modifications,
reagents available, Freund’s adjuvant has remained the such as phosphorylation, deamidation, and glycosylation
first choice for working with antigens that have poor im- [85, 86]. More sophisticated and laborious methods to
munogenicity in the immune host [78–80]. Freund’s com- confirm the protein identity comprise mass spectrometry
plete adjuvant contains heat-inactivated mycobacterial after proteolytic cleavage (peptide mapping), N-terminal
elements, which enhance the immune response by sequencing or tandem MS [87–89].
directly stimulating the activity of antigen-presenting In addition, classical SDS-PAGE analysis under reduc-
cells. However, it also elicits a local inflammatory reaction ing and non-reducing conditions gives an indication of
that may cause a necrotic appearance at the site of injec- protein purity and amount of contaminants by host cell
tion in the animal. To minimize or eliminate this side proteins, but also oxidation state and degradation of the
effect of Freund’s adjuvant, it is necessary to utilize a min- respective protein.
imal dose of the antigen and complete Freund’s adjuvant Importantly, quantified concentrations of purified
mixture in the first injection; subsequent injections are protein may differ dramatically from effective functional
then performed using incomplete Freund’s adjuvant. The protein concentration due to multimerization and aggre-
(partially) denaturating effect of Freund’s adjuvant gives gation because these non-native folding states are
rise to a tendency to isolate antibodies that recognize lin- unlikely to contribute to the specific interaction with the
ear exposed epitopes, even if properly folded protein binding partner. Analytical size-exclusion chromatogra-
preparations are used for immunizations [81–84]. phy (SEC) or different light-scattering methods (multian-
gle light scattering (SEC-MALS) or dynamic light scat-
tering (DLS)) assist in quality control regarding aggrega-
tion [90].

1438 © 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim


Biotechnol. J. 2012, 7, 1433–1443

www.biotecvisions.com

Table 1. Overview on quality parameters, methods, and their readout

Quality parameter Method Readout


Quantity Absorbance 280 nm Protein concentration [mg/mL]
Lab chip (Caliper)
HPLC
Purity HPLC Purity of protein [%], amount of contaminations/impurities
SDS-PAGE of expression host
Lab chip (Caliper)
LC-MS
Identity LC-MS Expected protein
Peptide mapping
N-terminal protein sequencing
Tandem MS
Immunoassays (western blot, ELISA)
Integrity LC-MS Intact protein, degradation by proteolytic cleavage,
SDS-PAGE folding state, and secondary structure
Circular dichroism (CD) spectroscopy
Dispersity Analytical SEC Multimerization and aggregation
SEC-MALS
DLS
Homogeneity LC-MS Post-translational modifications
(e.g. phosphorylation, deamidation, glycosylation)
Pyrogenicity LAL assay (turbidimetric or chromogenic) Endotoxin level [EU/mg]
Functionality BiaCORE Affinity [M]
Cellular assays Binding
FACS Specificity
Immunoassays (ELISA)
Ligand binding assays

Furthermore, protein oxidation, aggregation, and deg- teria-derived protein preparations. Another major source
radation are known issues that can affect the outcome of of endotoxins are hydrolysates, which are frequently
antibody generation severely. Therefore, there is a need to added to cell culture media as a substitute for fetal bovine
carefully monitor, for example, by MS or functional assays, serum. Due to their deleterious effect in animal studies, it
the quality of the antigen produced. Additional post- is clearly mandatory to remove endotoxins from immuno-
translational modification, such as deamidation, may like- gens. This can be very demanding because, despite the
wise be critical to the successful isolation of antibodies development of novel methods, there is no universal
and needs to be appreciated [91, 92]. recipe for removal. To determine the pyrogenicity of pro-
Glycosylation is another aspect that can affect antigen tein samples the turbidimetric LAL (limulus amoebocyte
functionality. As outlined in previous sections, the glyco- lysate) technique and the chromogenic LAL technique
sylation pattern is dependent on the host organism as well are most frequently used [95–97].
as on cultivation conditions [93]. Thus, it can be essential Last but not least, one of the most important and
in some instances to characterize the sugar moieties of meaningful parameters is the functionality of the antigen
recombinantly expressed proteins in detail to assess preparation. The type of functional assessment is very
product consistency as well as to understand the rela- dependent on the nature and class of the antigen and
tionship between glycan structure and function. Several should provide information on overall binding, affinity, or
methods have been described for detailed characteriza- specificity. Interactions of soluble antigens to their bind-
tion of the glycan profiles of proteins, ranging from HPLC ing partners are examined by immunoassays (e.g. ELISA)
over capillary electrophoresis to MS-based methods [94]. if the reagents for antigen detection are available. Other-
In the context of in vivo application of antigens, wise, label-free assays, such as surface plasmon reso-
attention should be paid to the presence and, if necessary, nance, come into play [98, 99]. Transmembrane proteins,
removal of endotoxins. Endotoxins, also called lipopoly- either as antigens or as antigen-binding partners, require
saccharides, are major contaminants often found in bac- specific assays with a functional readout, such as fluores-

© 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim 1439


Biotechnology Biotechnol. J. 2012, 7, 1433–1443
Journal
www.biotechnology-journal.com

cent imaging plate reader (FLIPR) technology or reporter


gene assays [100, 101]. The detailed setup of cell-based or Hilmar Ebersbach entered the field of
ligand-binding assays needs to be tailored to the individ- protein engineering during his Ph.D.,
ual antigen [102, 103]. which was a joint venture project
The quality criteria discussed herein are by no means between the University of Halle and a
exhaustive, but should give an overview of the key pa- small biotech company, Scil Proteins,
rameters summarized in Table 1. All described tech- Halle, Germany. He continued to
niques should be properly considered before application broaden his experience in display tech-
in any antibody generation campaign to allow the pro- nologies with a post-doctoral project in
duction of a highly valuable binding molecule. the Plückthun lab at the University of
Zürich, Switzerland. In 2007 he joined Novartis Research, Biologics
Center, in Basel as an expert in antibody generation. In this position
5 Discussion he developed several novel libraries of different binding molecules for
drug discovery.
This review covers a plethora of different options for anti-
gen generation and validation. As an extract and to
bridge the descriptive contents with applications, we
summarize briefly our strategies for antigen generation.
The selection of an appropriate expression system for a Sabine Geisse is a recognized expert in
given antigen is based on the nature of the target and its eukaryotic cell biology with >20 years of
anticipated biochemical and biophysical behavior. It is experience gathered during her Ph.D.
our experience that a multiparallel approach in different studies in Nutrition and Human Biolo-
systems (e.g. E. coli and mammalian cells) with multiple gy at the Justus-Liebig-University in
constructs greatly enhances the chances of success, Giessen and Philipps-University in Mar-
especially for novel targets. The candidate proteins are burg and as a post-doctoral researcher
mostly tagged, including differently sized tags with var- at the University of Essen, Germany.
ied positions. This enables in-process analytics during She joined Sandoz/Novartis in 1988
small-scale trial expression preceding upscaling of cul- within Discovery Technologies and is a scientific expert for biomole-
tures, as well as affinity-based purification as a first step. cule production. In 2004, she received the Novartis Leading Scientist
The vast majority of proteins derived from mammalian award. Sabine Geisse built up and implemented an expression plat-
cells are purified by affinity-based capturing, followed by form for transient and stable expression technologies in eukaryotic cell
a size-exclusion polishing step, if necessary. The purified lines and developed, among others, a HEK293 expression system for
proteins are characterized by SDS-PAGE analysis under transient expression on small and large scale for the rapid production
non-reducing and reducing conditions, as well as by MS, of recombinant proteins.
which can be preceded by a de-glycosylation step, if
needed. The aggregation and degradation rate of the final
product is assessed by analytical SEC, likewise the endo-
toxin level is determined. More elaborate techniques, We would like to acknowledge the many scientific contri-
such as SEC-MALS, N-terminal sequencing, tryptic butions from all our colleagues in the Novartis Biologics
digest followed by peptide mapping, and assessment of Center, subgroups ‘Protein Production and Antibodies’
the correct folding state of E. coli-derived antigens by DLS and ‘Novel Technologies’, that enabled us to compile the
are carried out on demand. Functionality of the protein is content of this review. We are grateful to our colleague,
always analyzed by appropriate assays developed by the Dr. Anne London, for careful proofreading of the manu-
project teams. script.
Lastly, a brief comment on the rather large set of com-
mercially available antigen preparations: the possibility of The authors declare no conflict of interest.
acquiring antigens as a starting material for target vali-
dation and assay development is certainly a huge advan-
tage in research. Unfortunately, apart from the cost issue, 6 References
the quality of the purchased material often does not live
[1] Dimitrov, D. S., Marks, J. D., Therapeutic antibodies: Current state
up to expectations. Thus, in addition to the product char-
and future trends – is a paradigm change coming soon? Methods
acteristics described by the vendor being carefully looked Mol. Biol. 2009, 525, 1–27, xiii.
at, a quality check by biochemical, biophysical, and/or [2] Enever, C., Batuwangala, T., Plummer, C., Sepp, A., Next generation
functional means is clearly advisable to avoid unpleasant immunotherapeutics – honing the magic bullet. Curr. Opin. Biotech-
surprises at later stages of the project. nol. 2009, 20, 405–411.

1440 © 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim


Biotechnol. J. 2012, 7, 1433–1443

www.biotecvisions.com

[3] Beck, A., Wurch, T., Bailly, C., Corvaia, N., Strategies and challenges [27] Hamilton, S. R., Gerngross, T. U., Glycosylation engineering in yeast:
for the next generation of therapeutic antibodies. Nat. Rev. Immunol. The advent of fully humanized yeast. Curr. Opin. Biotechnol. 2007,
2010, 10, 345–352. 18, 387–392.
[4] Elbakri, A., Nelson, P. N., Abu Odeh, R. O., The state of antibody [28] Drugmand, J. C., Schneider, Y. J., Agathos, S. N., Insect cells as fac-
therapy. Hum. Immunol. 2010, 71, 1243–1250. tories for biomanufacturing. Biotechnol. Adv. 2012, 30, 1140–1157.
[5] Carter, P. J., Introduction to current and future protein therapeutics: [29] Trowitzsch, S., Bieniossek, C., Nie, Y., Garzoni, F. et al., New bac-
A protein engineering perspective. Exp. Cell Res. 2011, 317, 1261– ulovirus expression tools for recombinant protein complex produc-
1269. tion. J. Struct. Biol. 2010, 172, 45–54.
[6] Axelsen, T. V., Holm, A., Christiansen, G., Birkelund, S., Identifica- [30] Hitchman, R. B., Locanto, E., Possee, R. D., King, L. A., Optimizing
tion of the shortest Aβ-peptide generating highly specific antibod- the baculovirus expression vector system. Methods 2011, 55, 52–57.
ies against the C-terminal end of amyloid-beta42. Vaccine 2011, 29, [31] Bieniossek, C., Imasaki, T., Takagi, Y., Berger, I., MultiBac: Expand-
3260–3269. ing the research toolbox for multiprotein complexes. Trends
[7] Huang, L., Sato, A. K., Sachdeva, M., Fleming, T. et al., Discovery of Biochem. Sci. 2012, 37, 49–57.
human antibodies against the C5aR target using phage display [32] Aumiller, J. J., Mabashi-Asazuma, H., Hillar, A., Shi, X. et al., A new
technology. J. Mol. Recognit. 2005, 18, 327–333. glycoengineered insect cell line with an inducibly mammalianized
[8] Trier, N. H., Hansen, P. R., Houen, G., Production and characteriza- protein N-glycosylation pathway. Glycobiology 2012, 22, 417–428.
tion of peptide antibodies. Methods 2012, 56, 136–144. [33] van Oers, M. M., Opportunities and challenges for the baculovirus
[9] Berkmen, M., Production of disulfide-bonded proteins in Escherichia expression system. J. Invertebr. Pathol. 2011, 107 Suppl, S3–15.
coli. Protein Expression Purif. 2012, 82, 240–251. [34] Rajendra, Y., Kiseljak, D., Baldi, L., Hacker, D. L. et al., A simple high-
[10] Makino, T., Skretas, G., Georgiou, G., Strain engineering for im- yielding process for transient gene expression in CHO cells.
proved expression of recombinant proteins in bacteria. Microb. Cell J. Biotechnol. 2011, 153, 22–26.
Fact. 2011, 10, 32. [35] Geisse, S., Reflections on more than 10 years of TGE approaches.
[11] Schlapschy, M., Skerra, A., Periplasmic chaperones used to enhance Protein Expression Purif. 2009, 64, 99–107.
functional secretion of proteins in E. coli. Methods Mol. Biol. 2011, [36] Sun, X., Hia, H. C., Goh, P. E., Yap, M. G., High-density transient
705, 211–224. gene expression in suspension-adapted 293 EBNA1 cells. Biotech-
[12] Yoon, S. H., Kim, S. K., Kim, J. F., Secretory production of recombi- nol. Bioeng. 2008, 99, 108–116.
nant proteins in Escherichia coli. Recent Pat. Biotechnol. 2010, 4, [37] Raymond, C., Tom, R., Perret, S., Moussouami, P. et al., A simplified
23–29. polyethylenimine-mediated transfection process for large-scale and
[13] Basu, A., Li, X., Leong, S. S., Refolding of proteins from inclusion high-throughput applications. Methods 2011, 55, 44–51.
bodies: Rational design and recipes. Appl. Microbiol. Biotechnol. [38] Kunert, R., Vorauer-Uhl, K., Strategies for efficient transfection of
2011, 92, 241–251. CHO-cells with plasmid DNA. Methods Mol. Biol. 2012, 801, 213–
[14] Mayer, M., Buchner, J., Refolding of inclusion body proteins. Meth- 226.
ods Mol. Med. 2004, 94, 239–254. [39] Hopkins, R. F., Wall, V. E., Esposito, D., Optimizing transient recom-
[15] Sahdev, S., Khattar, S. K., Saini, K. S., Production of active eukaryot- binant protein expression in mammalian cells. Methods Mol. Biol.
ic proteins through bacterial expression systems: A review of the ex- 2012, 801, 251–268.
isting biotechnology strategies. Mol. Cell. Biochem. 2008, 307, 249– [40] Bollin, F., Dechavanne, V., Chevalet, L., Design of Experiment in
264. CHO and HEK transient transfection condition optimization. Protein
[16] Eiberle, M. K., Jungbauer, A., Technical refolding of proteins: Do we Expression Purif. 2011, 78, 61–68.
have freedom to operate? Biotechnol. J. 2010, 5, 547–559. [41] Eibl, R., Kaiser, S., Lombriser, R., Eibl, D., Disposable bioreactors:
[17] Burgess, R. R., Refolding solubilized inclusion body proteins. Meth- The current state-of-the-art and recommended applications in
ods Enzymol. 2009, 463, 259–282. biotechnology. Appl. Microbiol. Biotechnol. 2010, 86, 41–49.
[18] Gautam, S., Dubey, P., Rather, G. M., Gupta, M. N., Non-chromato- [42] Boussif, O., Lezoualc’h, F., Zanta, M. A., Mergny, M. D. et al., A ver-
graphic strategies for protein refolding. Recent Pat. Biotechnol. satile vector for gene and oligonucleotide transfer into cells in cul-
2012, 6, 57–68. ture and in vivo: Polyethylenimine. Proc. Natl. Acad. Sci. USA 1995,
[19] Correa, A., Oppezzo, P., Tuning different expression parameters to 92, 7297–7301.
achieve soluble recombinant proteins in E. coli: Advantages of high- [43] Baldi, L., Hacker, D. L., Meerschman, C., Wurm, F. M., Large-scale
throughput screening. Biotechnol. J. 2011, 6, 715–730. transfection of mammalian cells. Methods Mol. Biol. 2012, 801, 13–
[20] Young, C. L., Britton, Z. T., Robinson, A. S., Recombinant protein ex- 26.
pression and purification: A comprehensive review of affinity tags [44] Pham, P. L., Kamen, A., Durocher, Y., Large-scale transfection of
and microbial applications. Biotechnol. J. 2012, 7, 620–634. mammalian cells for the fast production of recombinant protein. Mol.
[21] Walls, D., Loughran, S. T., Tagging recombinant proteins to enhance Biotechnol. 2006, 34, 225–237.
solubility and aid purification. Methods Mol. Biol. 2011, 681, 151– [45] Staack, R. F., Stracke, J. O., Stubenrauch, K., Vogel, R. et al., Quality
175. requirements for critical assay reagents used in bioanalysis of ther-
[22] Hengen, P., Purification of His-Tag fusion proteins from Escherichia apeutic proteins: What bioanalysts should know about their
coli. Trends Biochem. Sci. 1995, 20, 285–286. reagents. Bioanalysis 2011, 3, 523–534.
[23] Lichty, J. J., Malecki, J. L., Agnew, H. D., Michelson-Horowitz, D. J. [46] Waugh, D. S., Making the most of affinity tags. Trends Biotechnol.
et al., Comparison of affinity tags for protein purification. Protein Ex- 2005, 23, 316–320.
pression Purif. 2005, 41, 98–105. [47] Malhotra, A., Tagging for protein expression. Methods Enzymol.
[24] Konthur, Z., Hust, M., Dubel, S., Perspectives for systematic in vitro 2009, 463, 239–258.
antibody generation. Gene 2005, 364, 19–29. [48] Dyson, M. R., Selection of soluble protein expression constructs: The
[25] Singh, M. B., Bhalla, P. L., Recombinant expression systems for al- experimental determination of protein domain boundaries. Bio-
lergen vaccines. Inflammation Allergy: Drug Targets 2006, 5, 53–59. chem. Soc. Trans. 2010, 38, 908–913.
[26] Mattanovich, D., Branduardi, P., Dato, L., Gasser, B. et al., Recombi- [49] Kung, P., Goldstein, G., Reinherz, E. L., Schlossman, S. F., Mono-
nant protein production in yeasts. Methods Mol. Biol. 2012, 824, clonal antibodies defining distinctive human T cell surface antigens.
329–358. Science 1979, 206, 347–349.

© 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim 1441


Biotechnology Biotechnol. J. 2012, 7, 1433–1443
Journal
www.biotechnology-journal.com

[50] Zhang, C., Xu, Y., Gu, J., Schlossman, S. F., A cell surface receptor [71] Casares, S., Inaba, K., Brumeanu, T. D., Steinman, R. M. et al., Anti-
defined by a mAb mediates a unique type of cell death similar to on- gen presentation by dendritic cells after immunization with DNA
cosis. Proc. Natl. Acad. Sci. USA 1998, 95, 6290–6295. encoding a major histocompatibility complex class II-restricted viral
[51] Zhang, C. H., Davis, W. C., Grunig, G., Antczak, D. F., The equine ho- epitope. J. Exp. Med. 1997, 186, 1481–1486.
mologue of LFA-1 (CD11a/CD18): Cellular distribution and differen- [72] Shedlock, D. J., Weiner, D. B., DNA vaccination: Antigen presenta-
tial determinants. Vet. Immunol. Immunopathol. 1998, 62, 167–183. tion and the induction of immunity. J. Leukocyte Biol. 2000, 68,
[52] Jahnichen, S., Blanchetot, C., Maussang, D., Gonzalez-Pajuelo, M. et 793–806.
al., CXCR4 nanobodies (VHH-based single variable domains) po- [73] ’t Hoen, P. A., Jirka, S. M., ten Broeke, B. R., Schultes, E. A. et al.,
tently inhibit chemotaxis and HIV-1 replication and mobilize stem Phage display screening without repetitious selection rounds. Anal.
cells. Proc. Natl. Acad. Sci. USA 2010, 107, 20 565–20 570. Biochem. 2012, 421, 622–631.
[53] Popkov, M., Rader, C., Barbas III, C. F., Isolation of human prostate [74] Matochko, W., Chu, K., Jin, B., Lee, S. W. et al., Deep sequencing
cancer cell reactive antibodies using phage display technology. analysis of phage libraries using Illumina platform. Methods 2012,
J. Immunol. Methods 2004, 291, 137–151. DOI: 10.1016/j.ymeth.2012.07.006.
[54] Rivinoja, A., Laakkonen, P., Identification of homing peptides using [75] de Boer, E., Rodriguez, P., Bonte, E., Krijgsveld, J. et al., Efficient bi-
the in vivo phage display technology. Methods Mol. Biol. 2011, 683, otinylation and single-step purification of tagged transcription fac-
401–415. tors in mammalian cells and transgenic mice. Proc. Natl. Acad. Sci.
[55] Witting, S. R., Li, L. H., Jasti, A., Allen, C. et al., Efficient large vol- USA 2003, 100, 7480–7485.
ume lentiviral vector production using flow electroporation. Human [76] Zahnd, C., Sarkar, C. A., Pluckthun, A., Computational analysis of
Gene Therapy 2012, 23, 243–249. off-rate selection experiments to optimize affinity maturation by di-
[56] Kleman, M. I., Oellers, K., Lullau, E., Optimal conditions for freezing rected evolution. Protein Eng., Des. Sel. 2010, 23, 175–184.
CHO-S and HEK293-EBNA cell lines: Influence of Me2SO, freeze [77] Derda, R., Tang, S. K., Li, S. C., Ng, S. et al., Diversity of phage-dis-
density, and PEI-mediated transfection on revitalization and growth played libraries of peptides during panning and amplification. Mol-
of cells, and expression of recombinant protein. Biotechnol. Bioeng. ecules 2011, 16, 1776–1803.
2008, 100, 911–922. [78] Sanchez, Y., Ionescu-Matiu, I., Dreesman, G. R., Kramp, W. et al., Hu-
[57] Hsieh, J. M., Besserer, G. M., Madej, M. G., Bui, H. Q. et al., Bridging moral and cellular immunity to hepatitis B virus-derived antigens:
the gap: A GFP-based strategy for overexpression and purification Comparative activity of Freund complete adjuvant alum, and lipo-
of membrane proteins with intra and extracellular C-termini. Protein somes. Infect. Immun. 1980, 30, 728–733.
Sci. 2010, 19, 868–880. [79] Billiau, A., Matthys, P., Modes of action of Freund’s adjuvants in ex-
[58] Ashikawa, Y., Ihara, M., Matsuura, N., Fukunaga, Y. et al., GFP- perimental models of autoimmune diseases. J. Leukocyte Biol. 2001,
based evaluation system of recombinant expression through the se- 70, 849–860.
cretory pathway in insect cells and its application to the extracellu- [80] Odunsi, K., Qian, F., Matsuzaki, J., Mhawech-Fauceglia, P. et al.,
lar domains of class C GPCRs. Protein Sci. 2011, 20, 1720–1734. Vaccination with an NY-ESO-1 peptide of HLA class I/II specificities
[59] Cairns, V. R., DeMaria, C. T., Poulin, F., Sancho, J. et al., Utilization of induces integrated humoral and T cell responses in ovarian cancer.
non-AUG initiation codons in a flow cytometric method for efficient Proc. Natl. Acad. Sci. USA 2007, 104, 12837–12842.
selection of recombinant cell lines. Biotechnol. Bioeng. 2011, 108, [81] Paus, D., Winter, G., Mapping epitopes and antigenicity by site-di-
2611–2622. rected masking. Proc. Natl. Acad. Sci. USA 2006, 103, 9172–9177.
[60] Grisshammer, R., Purification of recombinant G-protein-coupled re- [82] Stills, H. F., Jr., Adjuvants and antibody production: Dispelling the
ceptors. Methods Enzymol. 2009, 463, 631–645. myths associated with Freund’s complete and other adjuvants. ILAR
[61] Chiu, M. L., Introduction to membrane proteins. Curr. Protoc. Protein J. 2005, 46, 280–293.
Sci. 2012, Chapter 29, Unit 29 21. [83] Perrie, Y., Mohammed, A. R., Kirby, D. J., McNeil, S. E. et al., Vaccine
[62] Drew, D., Newstead, S., Sonoda, Y., Kim, H. et al., GFP-based opti- adjuvant systems: Enhancing the efficacy of sub-unit protein anti-
mization scheme for the overexpression and purification of eukary- gens. Int. J. Pharm. 2008, 364, 272–280.
otic membrane proteins in Saccharomyces cerevisiae. Nat. Protoc. [84] Brunner, R., Jensen-Jarolim, E., Pali-Scholl, I., The ABC of clinical
2008, 3, 784–798. and experimental adjuvants – a brief overview. Immunol. Lett. 2010,
[63] Lebon, G., Bennett, K., Jazayeri, A., Tate, C. G., Thermostabilisation 128, 29–35.
of an agonist-bound conformation of the human adenosine A(2A) re- [85] Johnson, H., Eyers, C. E., Analysis of post-translational modifica-
ceptor. J. Mol. Biol. 2011, 409, 298–310. tions by LC-MS/MS. Methods Mol. Biol. 2010, 658, 93–108.
[64] Willis, S., Davidoff, C., Schilling, J., Wanless, A. et al., Virus-like par- [86] Timms, J. F., Cutillas, P. R., Overview of quantitative LC-MS tech-
ticles as quantitative probes of membrane protein interactions. Bio- niques for proteomics and activitomics. Methods Mol. Biol. 2010,
chemistry 2008, 47, 6988–6990. 658, 19–45.
[65] Yao, Q., Bu, Z., Vzorov, A., Yang, C. et al., Virus-like particle and [87] Ewles, M., Goodwin, L., Bioanalytical approaches to analyzing pep-
DNA-based candidate AIDS vaccines. Vaccine 2003, 21, 638–643. tides and proteins by LC–MS/MS. Bioanalysis 2011, 3, 1379–1397.
[66] Delcayre, A., Le Pecq, J. B., Exosomes as novel therapeutic nanode- [88] Doucet, A., Overall, C. M., Amino-terminal oriented mass spectrom-
vices. Curr. Opin. Mol. Ther. 2006, 8, 31–38. etry of substrates (ATOMS) N-terminal sequencing of proteins and
[67] Junge, F., Schneider, B., Reckel, S., Schwarz, D. et al., Large-scale proteolytic cleavage sites by quantitative mass spectrometry. Meth-
production of functional membrane proteins. Cell. Mol. Life Sci. ods Enzymol. 2011, 501, 275–293.
2008, 65, 1729–1755. [89] Egelhofer, V., Gobom, J., Seitz, H., Giavalisco, P. et al., Protein iden-
[68] He, M., Cell-free protein synthesis: Applications in proteomics and tification by MALDI-TOF-MS peptide mapping: A new strategy.
biotechnology. N. Biotechnol. 2008, 25, 126–132. Anal. Chem. 2002, 74, 1760–1771.
[69] Reckel, S., Sobhanifar, S., Durst, F., Lohr, F. et al., Strategies for the [90] Li, Y., Weiss, W. F. T., Roberts, C. J., Characterization of high-molec-
cell-free expression of membrane proteins. Methods Mol. Biol. 2009, ular-weight nonnative aggregates and aggregation kinetics by size
607, 187–212. exclusion chromatography with inline multi-angle laser light scat-
[70] Robinson, H. L., Pertmer, T. M., Nucleic acid immunizations. Curr. tering. J. Pharm. Sci. 2009, 98, 3997–4016.
Protoc. Immunol. 2001, Chapter 2, Unit 2 14.

1442 © 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim


Biotechnol. J. 2012, 7, 1433–1443

www.biotecvisions.com

[91] Srebalus Barnes, C. A., Lim, A., Applications of mass spectrometry [98] Lunn, C. A., Label-free screening assays: A strategy for finding bet-
for the structural characterization of recombinant protein pharma- ter drug candidates. Future Med. Chem. 2010, 2, 1703–1716.
ceuticals. Mass Spectrom. Rev. 2007, 26, 370–388. [99] Murphy, M., Jason-Moller, L., Bruno, J., Using Biacore to measure
[92] Jenkins, N., Meleady, P., Tyther, R., Murphy, L., Strategies for the binding kinetics of an antibody-antigen interaction. Curr. Protoc.
analysing and improving the expression and quality of recombinant Protein Sci. 2006, Chapter 19, Unit 19 14.
proteins made in mammalian cells. Biotechnol. Appl. Biochem. 2009, [100] Miraglia, L. J., King, F. J., Damoiseaux, R., Seeing the light: Lumi-
53, 73–83. nescent reporter gene assays. Comb. Chem. High Throughput
[93] Baker, K. N., Rendall, M. H., Hills, A. E., Hoare, M. et al., Metabolic Screening 2011, 14, 648–657.
control of recombinant protein N-glycan processing in NS0 and CHO [101] Arkin, M. R., Connor, P. R., Emkey, R., Garbison, K. E. et al., FLIPR
cells. Biotechnol. Bioeng. 2001, 73, 188–202. Assays for GPCR and Ion Channel Targets, In: Sittapalam, G. S.,
[94] Karg, S. R., Frey, A. D., Ferrara, C., Streich, D. K. et al., A small-scale Gal-Edd, N., Arkin, N., Auld, D. et al. (Eds), Assay Guidance Man-
method for the preparation of plant N-linked glycans from soluble ual, Eli Lily & Company and the National Center for Advancing
proteins for analysis by MALDI-TOF mass spectrometry. Plant Phys- Translational Sciences, Bethestda, MD 2004–2012.
iol. Biochem. 2009, 47, 160–166. [102] Michelini, E., Cevenini, L., Mezzanotte, L., Coppa, A. et al., Cell-
[95] Magalhaes, P. O., Lopes, A. M., Mazzola, P. G., Rangel-Yagui, C. et based assays: Fuelling drug discovery. Anal. Bioanal. Chem. 2010,
al., Methods of endotoxin removal from biological preparations: A re- 398, 227–238.
view. J. Pharm. Pharm. Sci. 2007, 10, 388–404. [103] Strange, P. G., Use of the GTPgammaS ([35S]GTPgammaS and Eu-
[96] Wilson, M. J., Haggart, C. L., Gallagher, S. P., Walsh, D., Removal of GTPgammaS) binding assay for analysis of ligand potency and ef-
tightly bound endotoxin from biological products. J. Biotechnol. ficacy at G protein-coupled receptors. Br. J. Pharmacol. 2010, 161,
2001, 88, 67–75. 1238–1249.
[97] Petsch, D., Anspach, F. B., Endotoxin removal from protein solutions.
J. Biotechnol. 2000, 76, 97–119.

© 2012 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim 1443

You might also like