You are on page 1of 54

C H A P T E R

20
Antibody Modification and Conjugation

The ability to conjugate an antibody to another preparations are isolated from antisera by affinity chro-
protein or molecule is critically important for many matography using the corresponding immobilized anti-
applications in life science research, diagnostics, and gen (see Chapter  15). These preparations thus contain
therapeutics. Antibody conjugates have become one of only that population of antibody molecules which has
the most important classes of biological agents associ- the desired antigenic specificity. Modification or conju-
ated with targeted therapy for cancer and other dis- gation of whole immunoglobulin fractions should be
eases. There literally are dozens of markers that have avoided, since other antibody populations will be pres-
been identified on tumor cells to which monoclonal ent and cause considerable nonspecificity in the resul-
antibodies have been developed for targeted therapy tant activity of the reagent. Even secondary antibodies
(Carter et al., 2004). The preparation of antibody conju- should be affinity purified and highly cross-adsorbed
gates to find and destroy cancer cells in vivo has become against immunoglobulins of other species’ antibody
one of the leading strategies of research into investi- types to prevent nonspecific interactions.
gational new drugs (McCarron et  al., 2005; Sievers and Monoclonal antibodies also should be purified by
Senter, 2012). In most cases, the site-specific delivery of affinity chromatography prior to undergoing biocon-
drugs involves the successful development of defined jugation. This can be accomplished using an immobi-
monoclonal antibody conjugates that can target dis- lized antigen or, if the antigen is not available in large
eased cells without affecting normal ones (see Chapter 1 enough quantities, an immobilized immunoglobulin
for an extensive review of antibody conjugates used in binding protein (such as protein A) may be employed.
the treatment of disease). Most monoclonals that can be successfully purified
In addition, the use of antibody molecules in immu- while maintaining activity also will be stable enough
noassay or detection techniques encompasses a broad to withstand the rigors of chemical modification.
variety of applications affecting nearly every field of Occasionally, however, a particular monoclonal will be
research. The availability of relatively inexpensive poly- partially or completely inactivated through the modi-
clonal and monoclonal antibodies of exacting specific- fication reaction. Sometimes this activity loss is caused
ity has made possible the design of reagent systems by physically blocking the antigen binding sites during
that can interact in high affinity with virtually any conjugation. In other cases, conformational changes in
conceivable analyte. The directed specificity of puri- the complementarity-determining regions (CDRs) are
fied immunoglobulins provides powerful tools for con- the cause of the problem. If the antigen binding site is
structing immunological reagents. Using a number of merely being blocked, then choosing an appropriate
conjugation and modification techniques, these specific site-directed chemistry may solve the problem. On the
antibodies can be modified to allow easy tracking in other hand, some monoclonals are too labile to undergo
complex mixtures. For instance, an antibody molecule modification reactions, regardless of the coupling
labeled with an enzyme, a fluorescent compound, or method. Trial and error often is necessary when work-
biotin provides a detectable complex able to be quanti- ing with monoclonals to determine if modification will
fied or visualized through its tag. See Chapter 1 for an severely affect activity.
overview of how antibody conjugates are being used in The unique structural characteristics of antibody
a wide variety of applications in research, diagnostics, molecules supply a number of choices for modification
and therapeutics. and conjugation schemes (Roitt, 1977; Goding, 1986;
To maintain specificity in antibody conjugates Harlow and Lane, 1988). The chemistry used to effect
derived from polyclonal antisera, only affinity-puri- conjugate formation should be chosen to yield the best
fied immunoglobulins should be used. Such purified possible retention of antigen binding activity. A detailed

Bioconjugate Techniques, Third Edition.


DOI: http://dx.doi.org/10.1016/B978-0-12-382239-0.00020-0 867 © 2013 Elsevier Inc. All rights reserved.
868 20.  Antibody Modification and Conjugation

Heavy-chain
Intrachain hypervariable
disulfides regions
Antigen
binding site
Interchain
disulfides
Light-chain
hypervariable
regions

Light chain
Complement
binding region

Hinge region

Glycosylation
Protein A
between heavy
binding region
chains

Heavy chains

FIGURE 20.1  Detailed structure of an immunoglobulin G (IgG) antibody molecule.

illustration of antibody structure is shown in Figure containing two light and two heavy chains. By con-
20.1. The most basic immunoglobulin G molecule is trast, IgA molecules can exist as a singlet, doublet, or
composed of two light and two heavy chains, held triplet of this basic Ig monomeric structure, while IgM
together by noncovalent interactions as well as a num- molecules are large pentameric constructs (Figure
ber of disulfide bonds. The light chains are disulfide- 20.2). Both IgA and IgM contain an additional subunit,
bonded to the heavy chains in the CL and CH1 regions, called the J chain—a very acidic polypeptide of molec-
respectively. The heavy chains are, in turn, disulfide- ular weight 15,000 that is very rich in carbohydrate.
bonded to each other in the hinge region. The heavy chains of immunoglobulin molecules also
The two heavy chains in each immunoglobulin mol- are glycosylated, typically in the CH2 domain within
ecule are identical. Depending on the class of immuno- the Fc fragment region, but also may contain carbohy-
globulin, the molecular weight of these subunits ranges drate near the antigen binding sites.
from about 50,000 to around 75,000. Similarly, the two There are two antigen binding sites on each of the
light chains of an antibody are identical and have a basic Ig-type monomeric structures, formed by the
molecular weight of about 25,000. For IgG molecules, heavy-/light-chain proximity in the N-terminal, hyper-
the intact molecular weight representing all four sub- variable region at the tips of the “y” structure. The
units is in the range of 150,000 to 160,000. unique tertiary structure created by these subunit pair-
There are two forms of light chains that may be ings produces the conformation necessary to interact
found in antibodies. A single antibody will have with a complementary antigen molecule. The points of
light-chain subunits of either lambda (λ) or kappa (κ) interaction on the immunoglobulin molecule with an
variety, but not both types in the same molecule. The antigen involve noncovalent forces that may encom-
immunoglobulin class, however, is determined by an pass numerous non-sequential amino acids within the
antibody’s heavy-chain variety. A single antibody will heavy and light chains. In other words, the binding site
also possess only one type of heavy chain (designated is formed not strictly from a linear sequence of amino
as γ, μ, α, ε, or δ). Thus, there are five major classes acids on each chain, but from the unique orientation
of antibody molecules, each determined from their of these groups in three-dimensional space as the beta-
heavy-chain type and designated as IgG, IgM, IgA, sheet structure turns at the tips of the Fab regions. The
IgE, or IgD. Three of these antibody classes, IgG, IgE, binding site thus has affinity for a particular antigen
and IgD, consist of the basic Ig monomeric structure molecule due to both structural complementarity as

BIOCONJUGATE TECHNIQUES
ANTIBODY MODIFICATION AND CONJUGATION  869

µ Heavy Chains

Light Chains α Heavy Chains

Secretory
Piece

J Chain J Chain

Disulfide
Linkages

Antigen
Binding Sites
Antigen
Binding Sites
Pentameric IgM Dimeric IgA

FIGURE 20.2  General structures of IgA and IgM antibodies.

well as the combination of van der Waals, ionic, hydro- purposes. Crosslinking reagents may be used to tar-
phobic, and hydrogen-bonding forces which may be get lysine ε-amine and N-terminal α-amine groups.
created at each point of contact. Carboxylate groups also may be coupled to another
Useful enzymatic derivatives of antibody molecules molecule using the C-terminal end as well as aspartic
may be prepared that still retain the antigen binding acid and glutamic acid residues. Although both amine
sites. Two principal digested forms of IgG antibod- and carboxylate groups are as plentiful in antibodies
ies are useful for creating immunological reagents. as they are in most proteins, the distribution of them
Enzymatic digestion with papain produces two small within the three-dimensional structure of an immuno-
fragments of the immunoglobulin molecule, each con- globulin is nearly uniform throughout the surface topol-
taining an antigen binding site (called Fab fragments), ogy. For this reason, conjugation procedures that utilize
and one larger fragment containing only the lower these functionalities will crosslink to the most accessible
portions of the two heavy chains (called Fc, for “frag- ones in a somewhat random fashion on nearly all parts
ment crystallizable”) (Section 1.4) (Coulter and Harris, of the antibody molecule. This, in turn, leads to a ran-
1983). Alternatively, pepsin cleavage produces one large dom orientation of the antibody within the conjugate
fragment containing two antigen binding sites (called structure, which may block the antigen binding sites
F(ab′)2) and many smaller fragments formed from against the surface of another coupled protein or mole-
extensive degradation of the Fc region (Rousseaux et al., cule. Obscuring the binding sites in this manner results
1983). The F(ab′)2 fragment is held together by reten- in decreased antigen binding activity in the conjugate
tion of the disulfide bonds in the hinge region. Specific compared to that observed for the unconjugated anti-
reduction of these disulfides using 2-mercaptoethyl- body. This effect also is dependent on how many points
amine (MEA), dithiothreitol (DTT), tris(2-carboxyethyl) of modification are made on the antibody molecules.
phosphine (TCEP), or other reducing agents (Chapter 2, Controlling the level of modification and conjugation
Section 4.1) produces two Fab′ fragments, each of will typically lead to antibody conjugates having the
which has one antigen binding site. highest possible activity in the anticipated application.
Antibody molecules possess a number of func- Conjugation chemistry done with antibody mol-
tional groups suitable for modification or conjugation ecules may be more successful at preserving activity

BIOCONJUGATE TECHNIQUES
870 20.  Antibody Modification and Conjugation

if the functional groups that are utilized are present in post-translationally modified with carbohydrate after
limiting quantities or only at discrete sites on the mol- hybridoma synthesis. Recombinant antibodies grown
ecule. Such “site-directed conjugation” schemes make in bacteria may also be devoid of carbohydrate. Before
use of crosslinking reagents that can specifically react attempting to use a conjugation method that couples
with residues that are only in certain positions on the through polysaccharide regions, it is best to test the anti-
immunoglobulin surface—usually chosen to be well body to see if it contains carbohydrate—especially if the
removed from the antigen binding sites. By proper immunoglobulin is of hybridoma or recombinant origin.
selection of the conjugation chemistry and knowledge
of antibody structure, the immunoglobulin molecule
can be oriented so that its bivalent binding potential for 1.  PREPARATION OF ANTIBODY–
antigen remains maximally available. ENZYME CONJUGATES
Two site-directed chemical reactions are especially
useful in this regard. The disulfides in the hinge region The most extensive application of antibody conjuga-
that hold the heavy chains together can be cleaved tion using crosslinking reagents is for the preparation
with a reducing agent (such as MEA, DTT, or TCEP) of antibody–enzyme conjugates. Since the develop-
to reveal some thiols in the intact antibody or in some ment of enzyme-linked immunosorbent assay (ELISA)
cases to create two half-antibody molecules that have systems, the ability to make conjugates of specific anti-
thiols available, each containing an antigen bind- bodies with enzymes has provided the means to quan-
ing site (Palmer and Nissonoff, 1963; Sun et  al., 2005) tify or detect hundreds, if not thousands, of important
(Chapter  2, Section 4.1). Alternatively, smaller antigen analytes. The use of enzymes as labels in immunoassay
binding fragments may be made from pepsin digestion procedures surpassed radioactive tags as the means
(which makes F(ab′)2 fragments lacking most of the Fc of detection, primarily due to the long-term stabil-
region) and similarly reduced to form monovalent Fab′ ity potential of an enzyme system and the hazards
molecules containing thiols. Both of these preparations and waste problems associated with radioisotopes.
contain free sulfhydryl groups that can be targeted for Designed properly, an antibody–enzyme conjugate
conjugation using thiol-reactive probes or crosslinkers. assay system can be just as sensitive as a radiolabeled
Conjugations done using hinge-region SH groups will antibody system.
orient the attached protein or other molecule away from The development of viable methods for crosslink-
the antigen binding sites, thus preventing blockage of ing antibody and enzyme molecules—methods that
these regions and better preserving activity. retain high antigen binding activity coupled with high
The second method of site-directed conjugation of enzymatic activity—has formed the basis for much
antibody molecules takes advantage of the glycan car- of today’s diagnostic industries, literally a multi-bil-
bohydrate chains typically attached to the CH2 domain lion dollar enterprise with enormous impact on world
between the heavy chains in the Fc region. Mild oxida- health (see Chapter  1 for an extensive review). The
tion of the polysaccharide sugar residues with sodium conjugation chemistries that make this possible are
periodate will generate aldehyde groups. A crosslinking designed around a knowledge of both antibody and
or modification reagent containing a hydrazide or ami- enzyme structure. The best methods make use of defini-
nooxy functional group then can be used to specifically tive site-directed chemistries that target both molecules
target these aldehydes for coupling to another mol- in regions removed from their respective active centers.
ecule. Directed conjugation through antibody carbo- The major enzymes used in ELISA technology
hydrate chains may avoid the antigen binding regions include horseradish peroxidase (HRP), alkaline phos-
while allowing for use of intact antibody molecules. phatase (AP), β-galactosidase (β-gal), and glucose oxi-
This method can result in the highest retention of anti- dase (GO). See Chapter 22 for a detailed description of
gen binding activity within the ensuing conjugate. each enzyme’s properties and activities. HRP is by far
However, care should be taken in using this method, the most popular enzyme used in antibody–enzyme
because some antibody molecules can be glycosylated conjugates. One survey of enzyme use stated that HRP
near the antigen binding area, thus potentially inter- is incorporated in about 80% of all antibody conjugates,
fering with activity upon conjugate formation (Wright most of them utilized in diagnostic assay systems. AP
et al., 1991). is the second-most popular choice for antibody–enzyme
Another limitation to the use of this strategy is the conjugation, being used in almost 20% of all commer-
necessity for the antibody molecule to be glycosylated. cial enzyme-linked assays. Although β-gal and GO are
Antibodies of polyclonal origin (from antisera) are usu- used frequently in research labs and cited numerous
ally glycosylated and work well in this procedure, but times in the literature, their utilization for commercial
other antibody preparations may not possess polysac- ELISA applications represents less than 1% of the total
charide. In particular, some monoclonals may not be assays available.

BIOCONJUGATE TECHNIQUES
1.  Preparation of Antibody–Enzyme Conjugates 871
Conjugation methods for attaching these enzymes a maleimide–PEGn–NHS ester compound actually
to antibody molecules vary according to the functional increases the solubility of the antibody and may help
groups available. HRP is a glycoprotein and easily can to maintain better stability for certain sensitive mono-
be periodate oxidized for coupling via reductive amina- clonals than the traditional aliphatic crosslinkers. The
tion to the amino groups on immunoglobulins. β-Gal methods described below for SMCC may be used with
contains abundant free sulfhydryl groups in its native success for PEG-based reagents or other maleimide–
state. The thiols can be utilized for coupling to the sulf- NHS ester heterobifunctionals.
hydryl-reactive end of heterobifunctional crosslinkers In protocols involving enzyme activation with SMCC
such as SMCC (Chapter  6, Section 1.3). Any of these and subsequent conjugation with an antibody molecule
enzymes can be conjugated through their amine groups (the most common method of producing antibody–
using crosslinking agents such as glutaraldehyde or enzyme conjugates with this crosslinker), the antibody
various heterobifunctional agents. The catalytic prop- usually has to be prepared for coupling to the maleimide
erties and activation methods often used with these groups on the enzyme by introduction of sulfhydryl
enzymes are discussed in detail in Chapter 22. residues. Since antibodies typically do not contain free
The following sections describe the most common sulfhydryls accessible for conjugation, they must be
reagents and reactions used to create antibody–enzyme fabricated by chemical means. Two main options are
conjugates. available for creating sulfhydryl functions on immuno-
globulin molecules. The disulfide residues in the hinge
1.1.  NHS Ester–Maleimide-Mediated region of the IgG structure may be reduced with DTT,
TCEP, or MEA to cleave the immunoglobulin into two
Conjugation
half-antibody molecules each possessing one antigen
Heterobifunctional reagents containing an amine- binding site and the requisite sulfhydryls. Alternatively,
reactive NHS ester on one end and a sulfhydryl-reactive a thiolation reagent may be used to modify the intact
maleimide group on the other end generally have great antibody to contain sulfhydryls (Chapter 2, Section 4.1).
utility for producing antibody–enzyme conjugates (see Both options are described below. Although there are
Chapter 6, Section 1). Crosslinking reagents possessing numerous thiolation reagents from which to choose, only
these reactive groups can be used in controlled, multi- SATA and Traut’s reagent are discussed in this section,
step procedures that yield conjugates of defined com- since they are the most popular.
position and high activity. Among the most popular
of these NHS ester–maleimide crosslinkers are SMCC Activation of Enzymes with NHS Ester–Maleimide
(Chapter  6, Section 1.3), MBS (Chapter  6, Section 1.4), Crosslinkers
and GMBS (Chapter 6, Section 1.7). The use of any one The first step in conjugation of antibody molecules
of these crosslinkers in the following protocol can result and enzymes using NHS ester–maleimide crosslinkers
in useful conjugates. However, SMCC and its water usually is modification of the enzyme with the NHS
soluble analog, sulfo-SMCC, possess the most stable ester end of the reagent to produce a maleimide-acti-
maleimide functionalities and are probably the most vated derivative (Figure 20.3). The protocol described
widely used crosslinkers of this type. This increased here uses sulfo-SMCC as the crosslinking agent due to
stability to hydrolysis of SMCC’s hindered maleimide the enhanced stability of its maleimide group and the
group allows activation of either enzyme or antibody water-solubility afforded by the negatively charged
via the amine-reactive NHS ester end, resulting in a sulfonate on its sulfo-NHS ring. Other NHS ester–
maleimide-activated intermediate. The intermediate maleimide crosslinkers may be substituted without dif-
species can then be purified away from excess cross- ficulty; however, water-insoluble varieties should be
linker and reaction byproducts before mixing with the solubilized in DMSO or DMF prior to addition to the
second protein to be conjugated. The multi-step nature aqueous reaction mixture.
of this process limits polymerization of the conjugated One note should be mentioned before proceed-
proteins and provides control over the extent and sites ing: when conjugating antibody molecules with
of crosslinking. Antibody conjugation using SMCC or β-galactosidase, the antibody is usually activated with
sulfo-SMCC continues to be a popular route to the for- sulfo-SMCC first to take advantage of the indigenous
mation of conjugates useful in detection and targeted sulfhydryl groups on the enzyme. Therefore, if β-gal
therapeutic purposes (Nwe et  al., 2011; Lee et  al., 2012; is being used, substitute the antibody for the enzyme
Sukhanova et al., 2012). mentioned in this protocol, and then after the purifica-
In addition, the PEG-based heterobifunctional cross- tion step add the enzyme in the desired molar excess to
linkers described in Chapter  18, Section 1.2, provide produce the final conjugation.
enhanced water solubility for antibody conjugation The following protocol describes the activation
applications. Conjugation of antibody molecules using of horseradish peroxidase (HRP) with sulfo-SMCC.

BIOCONJUGATE TECHNIQUES
872 20.  Antibody Modification and Conjugation

O chromatography buffer. At this concentration, HRP


O can be observed visually as it flows through the
H2N E + N O N column due to the color of its heme ring. Pool the
O O fractions containing the HRP peak. After elution,
O
adjust the HRP concentration to 10 mg/ml for the
Enzyme Containing SMCC
conjugation reaction. At this point, the maleimide-
Amine Groups activated enzyme may be frozen and lyophilized to
O preserve its maleimide activity. The modified enzyme
is stable for at least 1 year in a freeze-dried state. If
N OH
kept in solution, the maleimide-activated HRP should
O
be used immediately to conjugate with an antibody
NHS following one of the three options outlined below.

O Conjugation with Reduced Antibodies


H
N N E One method of introducing sulfhydryl residues into
O
antibody molecules for conjugation with maleimide-
O activated enzymes is to reduce indigenous disulfide
groups in the hinge region of the immunoglobulin
SMCC-activated enzyme structure. Reduction with low concentrations of DTT,
containing sulfhydryl-reactive
maleimide groups
TCEP, or 2-mercaptoethylamine (MEA) will principally
cleave the disulfide bonds holding the heavy chains
FIGURE 20.3  The reaction of SMCC with the amine groups on together, but the disulfides between the heavy and light
enzyme molecules yields a maleimide-activated derivative capable of chains may also be partially reduced to some extent.
coupling with sulfhydryl-containing antibody molecules.
In a comparative study of disulfide reducing agents, it
was determined that use of the relatively strong reduc-
tants DTT and TCEP required only 3.25 and 2.75 mole
Activation of other enzymes is performed similarly, equivalents per mole equivalent of antibody molecule
with the appropriate adjustments in the mass of to achieve the reduction of two inter-chain disulfide
enzyme added to the reaction to account for differences bonds between the heavy chains of a monoclonal IgG
in molecular weight. (Sun et al., 2005). This limited reduction strategy retains
The gel filtration column described in step 3 should intact bispecific antibody molecules while providing
be prepared and equilibrated prior to starting the modi- discrete sites for conjugation to thiols. Using higher
fication reaction. Enzymes activated with sulfo-SMCC concentrations of DTT, TCEP, or MEA will result in
are available from Thermo Fisher. complete cleavage of the disulfides between the heavy
chains and formation of two half antibody molecules,
Protocol each containing an antigen binding site. Under these
1. Dissolve 18 mg of HRP in 0.1-M sodium phosphate, conditions, some inter-chain cleavage also will occur
0.15-M NaCl, pH 7.2, at a concentration of 20 to and result in some smaller fragments being produced.
30 mg/ml. The more highly concentrated the enzyme Similar reduction can be done with F(ab′)2 fragments
solution, the more efficient the modification reaction produced from pepsin digestion of IgG molecules.
will be. For conjugating smaller quantities of enzyme Either of these reduction steps creates half-antibody
and antibody, proportionally decrease the amount of fragments, each containing one heavy and one light
the reagents used, while attempting to maintain the chain and one antigen binding site (Figure 20.4). The
same relative concentrations in solution. sulfhydryl groups produced by this reduction are able
2. Add 6 mg of sulfo-SMCC (Thermo Fisher) to the HRP to couple with maleimide-activated enzymes without
solution. Mix to dissolve and react for 30 min at room blocking the antigen binding area.
temperature. Alternatively, two 3-mg additions of Antibody reduction is usually carried out in the pres-
crosslinker may be performed—the second one after ence of EDTA to prevent re-oxidation of the sulfhydryls
15 min of incubation—to obtain even more efficient by metal catalysis. In phosphate buffer at pH 6 to 7 and
modification. 4°C, one report stated that the number of available thi-
3. Immediately purify the maleimide-activated HRP ols decreased only by about 7% in the presence of EDTA
away from excess crosslinker and reaction byproducts over a 40-h time span. In the absence of EDTA, this sulf-
by gel filtration using a desalting resin. Use 0.1-M hydryl loss increased to 63 to 90% in the same period
sodium phosphate, 0.15-M NaCl, pH 7.2, as the (Yoshitake et al., 1979).

BIOCONJUGATE TECHNIQUES
1.  Preparation of Antibody–Enzyme Conjugates 873

SS SH HS
SS 2-MEA SH HS

IgG molecule Reduction in hinge region


yielding two half antibody
molecules containing sulfhydryls

O
H
N N

O
E
O

Maleimide-activated enzyme

O
S H
N N
SH E
O
O

Antibody–enzyme conjugate
formation through thioether bond

FIGURE 20.4  Reduction of the disulfide bonds within the hinge region of an IgG molecule can produce half-antibody molecules contain-
ing thiol groups. More limited reduction with low concentrations of reducing agents can be used to retain the intact antibody structure while
revealing several thiols for conjugation. Reaction of these reduced antibodies with a maleimide-activated enzyme creates a conjugate through
thioether bond formation.

In the following protocol, the most critical aspects are reductant. Polyclonal populations typically work well
the concentration of reducing agent and EDTA in the in this procedure.
reaction mixture. Good reduction of IgG will take place A final consideration is to provide adequate desalt-
with 50- to 100-mM MEA and 1- to 100-mM EDTA. ing of the reduced antibody molecule from excess
For DTT or TCEP, the concentration of reducing agent reducing agent. If even a small amount of a thiol-con-
should be lowered to a 3-fold molar excess over the taining reductant remains, subsequent conjugation with
amount of antibody present. The pH of the reaction can a maleimide-activated enzyme will be inhibited.
vary from pH 6 to 9, with about pH 8 being optimal.
The absolute concentration of antibody can vary and Protocol
still yield acceptable results. With some monoclonals, 1. Dissolve the IgG to be reduced at a concentration of
however, reduction may not be completely efficient in 1 to 10 mg/ml in 0.1-M sodium phosphate, 0.15-M
cleaving the antibody between the heavy-chain pairs. NaCl, pH 7.2, containing 10-mM EDTA.
Particularly, some subclasses of immunoglobulins con- 2. Add 6 mg of MEA to each ml of antibody solution.
tain structures with unusually high numbers of disul- Alternatively, add DTT or TCEP to a final
fides in the hinge region, and some of them may not be concentration equal to 3 mole equivalents per mole
reduced except under much higher concentrations of equivalent of antibody present. Mix to dissolve.

BIOCONJUGATE TECHNIQUES
874 20.  Antibody Modification and Conjugation

3. Incubate for 90 min at 37°C. described in the previous protocol. Each time a cysteine
4. Purify the reduced IgG by gel filtration using a disulfide is reduced to two thiols, the subsequent con-
desalting resin. Perform the chromatography using jugation step usually involves the reaction of only one
0.1-M sodium phosphate, 0.15-M NaCl, pH 7.2, of the thiols with a reactive group, such as in the use of
containing 10-mM EDTA as the buffer. To obtain a maleimide, haloacetyl, vinyl sulfone, or epoxy group.
efficient separation between the reduced antibody Using these common reagents, one of the two sulfhy-
and excess reductant, the sample size applied to dryls gets covalently linked to the crosslinker, but the
the column should be at a ratio of no more than 5% other one may remain free without attachment. A bro-
sample volume to column volume. Collect 0.5-ml ken disulfide linkage within the antibody may allow
fractions and monitor for protein at 280 nm. Since for subunit dissociation to occur even if the conjugation
the reducing agents typically have no absorbance at reaction is successful. However, this situation may be
280 nm, the elution profile also may be monitored overcome if the conjugation reaction is able to link both
by use of the BCA Protein Assay method (Thermo thiols simultaneously using a single crosslinking agent.
Fisher). The BCA–copper reagent reacts with the Both thiols of a reduced disulfide within a protein
reductants to produce a colored product. EDTA in can be targeted concurrently using a unique com-
the chromatography buffer will inhibit the BCA pound containing two reactive components consisting
method somewhat, but a color response to the of an α,α-bis[(p-tolylsulfonyl)methyl]-m-aminoaceto-
reducing agent peak will still be obtained. A micro- phenone group, termed an “equilibrium transfer alkyl-
method for monitoring each fraction is as follows: ation crosslink” reagent (ETAC) (Liberatore et  al.,
a. Take 5 μl from each fraction collected and place in 1990; Rosario et  al., 1990; Wilbur et  al., 1994). The two
a separate well of a microtiter plate. p-tolylsulfonyl groups on this compound react with
b. Add 200 μl of BCA working reagent. neighboring thiols in a multi-step reaction sequence
c. Incubate at room temperature or 37°C for 15 to leading to the formation of two thioether bonds with
30 min or until color develops. The color response a three-carbon bridge between the thiols (Figure 20.5).
may be measured visually or by absorbance at The molecular distance between the two thiols after
562 nm. To ensure good separation between the disulfide reduction is nearly perfect for covalent con-
antibody peak and excess MEA, at least one jugation with both reactive p-tolylsulfonyl groups,
fraction of little or no color should separate the thus forming a linkage which maintains subunit asso-
two peaks. ciation in proteins where disulfides occur between
5. Pool the fractions containing antibody and immediately the subunits of multi-subunit proteins, such as within
mix with an amount of maleimide-activated enzyme to antibodies. The modification of hinge region disulfides
obtain the desired molar ratio of antibody-to-enzyme with this reagent yields highly specific site-directed
in the conjugate. Use of a 4:1 (enzyme:antibody) molar conjugation while maintaining the bifunctional
ratio in the conjugation reaction usually results in high- nature of the intact antibody’s antigen binding sites.
activity conjugates suitable for use in many enzyme- Similarly, a F(ab′)2 fragment can be labeled with an
linked immunoassay procedures. Higher molar ratios ETAC reagent and maintain the bifunctional nature of
also have been used with success. the antibody, which normally does not occur when the
6. React for 30 to 60 min at 37°C or 2 h at room reduced disulfides of the fragments are labeled using
temperature. The conjugation reaction also may be standard alkylation reagents.
carried out at 4°C overnight. ETAC derivatives have been used to biotinylate pro-
7. The conjugate may be further purified away from teins and antibodies after disulfide reduction and to
unconjugated enzyme by the procedures described add PEG modifications to selected sites (Brocchini et al.,
in Section 1.5. For storage, the conjugate should be 2008; Choi et  al., 2009). At the time of writing, ETAC
kept frozen, lyophilized, or sterile filtered and kept conjugation strategies are being applied to drug devel-
at 4°C. Stability studies may have to be carried out to opment and biotherapeutic conjugates, but no reagent
determine the optimal method of long-term storage systems are commercially available.
for a particular conjugate.
Conjugation with 2-Iminothiolane-Modified
Antibodies
ETAC REAGENTS FOR DITHIOL CONJUGATION Traut’s reagent, or 2-iminothiolane, is described in
The use of disulfide reduction within antibod- Chapter  2, Section 4.1. The reagent reacts with amine
ies to create sites for bioconjugation has the potential groups in proteins or other molecules in a ring-opening
deficiency of breaking down the subunit structure of reaction to result in covalent modifications containing
an antibody. This may occur even when using limit- terminal sulfhydryl residues (Figure 20.6). Antibodies
ing quantities of reducing agent, such as the method may be modified with Traut’s reagent to create the

BIOCONJUGATE TECHNIQUES
1.  Preparation of Antibody–Enzyme Conjugates 875

DTT
S SH
S HS

Disulfide-linked
Reduction forms two thiols
subunits in protein

ETAC-
O O reactive
R O S R O group
N N O S O
H H O
S
O S O O

Dithio crosslink
with 3-carbon spacer 2 ETAC reagent
holds subunits together R = biotin, PEG
O S O
fluorescent probe, etc.
O

FIGURE 20.5  ETAC reagents can be used to form a covalent linkage between two thiols that were previously a part of a disulfide bond, thus
retaining the subunit structure of reduced proteins or antibodies.

requisite sulfhydryls necessary for conjugation with Protocol


a maleimide-activated enzyme. Unlike the disulfide 1. Dissolve the antibody to be modified at a
reduction method described in the previous section, concentration of 1 to 10 mg/ml in 0.1-M sodium
this protocol better retains the divalent nature of the phosphate, 0.15-M NaCl, pH 7.2, containing 10-mM
antibody molecule. However, since amine modification EDTA. High levels of EDTA often are required
of antibodies can take place at virtually any available to completely stop metal-catalyzed oxidation of
lysine ε-amine location, the resultant sulfhydryls are sulfhydryl groups when working with serum
distributed almost randomly over the immunoglobu- proteins—especially polyclonal antibodies purified
lin structure. Conjugation through these SH groups from antisera. Presumably, carry-over of iron from
may result in a certain population of antibodies that partially hemolyzed blood is the contaminating
have their antigen binding sites obscured or blocked culprit.
by enzyme molecules. Typically, however, enough free 2. Add 2-iminothiolane (Thermo Fisher) to this
antigen binding sites are available in the conjugate solution to give a molar excess of 20 to 40 times
to result in high-activity complexes useful in ELISA over the amount of antibody present (MW of
procedures. Traut’s reagent is 137.63). Solid 2-iminothiolane
The number of sulfhydryls created on the immuno- may be added despite the fact that the compound
globulin using thiolation procedures such as this one is relatively insoluble in aqueous solution. As the
is more critical to the yield of conjugated enzyme mol- reagent reacts, it will be completely drawn into
ecules than the molar excess of maleimide-activated solution. Alternatively, a stock solution of Traut’s
enzyme used in the conjugation reaction. Therefore, it is may be made in DMF and an aliquot added to the
important to use a sufficient excess of Traut’s reagent to antibody solution (not to exceed 10% DMF in the
obtain a sufficient number of available sulfhydryls. In final solution).
addition, the thiolated antibody should be used imme- 3. React for 30 min at 37°C or 1 h at room
diately to prevent loss of thiols due to re-cyclization, temperature.
which can tie up the available sulfhydryls in an unde- 4. Purify the thiolated antibody by gel filtration using
sired side reaction (see Chapter 2, Section 4.1). a desalting resin. Perform the chromatography using

BIOCONJUGATE TECHNIQUES
876 20.  Antibody Modification and Conjugation

SS + SS
NH2 Cl SS
SS
S +
+ NH2 Cl
SH
N
2-Iminothiolane H

Antibody molecule Modification producing


containing amine groups a terminal sulfhydryl group

O
H
N N
E
O
O

Maleimide-activated enzyme

SS
SS
+
NH2 Cl O
S H
N N
N
H
O
E
O

Antibody–enzyme conjugate
formation through thioether bond

FIGURE 20.6  Antibodies may be modified with 2-iminothiolane at their amine groups to create sulfhydryls for conjugation with SMCC-
activated enzymes. The maleimide groups on the derivatized enzyme react with the thiols on the antibody to form thioether bonds.

0.1-M sodium phosphate, 0.15-M NaCl, pH 7.2, of antibody-to-enzyme in the conjugate. Use of a
containing 10-mM EDTA as the buffer. To obtain 4:1 to 15:1 (enzyme:antibody) molar ratio in the
efficient separation between the reduced antibody conjugation reaction usually results in high-activity
and excess reductant, the sample size applied to conjugates suitable for use in many enzyme-linked
the column should be at a ratio of no more than immunoassay procedures.
5% sample volume to the total column volume. 6. React for 30 to 60 min at 37°C or 2 h at room
Collect 0.5-ml fractions and monitor for protein at temperature. The conjugation reaction also may be
280 nm. To monitor the separation of the second performed at 4°C overnight.
peak (excess Traut’s reagent), the BCA Protein Assay 7. The conjugate may be further purified away from
reagent (Thermo Fisher) may be used according to unconjugated enzyme by the procedures described
the procedure described in the previous section, in Section 1.5. For storage, the conjugate should be
protocol step 4. kept frozen, lyophilized, or sterile filtered and kept
5. Pool the fractions containing antibody and at 4°C. Stability studies may have to be performed to
immediately mix with an amount of maleimide- determine the optimal method of long-term storage
activated enzyme to obtain the desired molar ratio for a particular conjugate.

BIOCONJUGATE TECHNIQUES
1.  Preparation of Antibody–Enzyme Conjugates 877

SS SS
SS O O SS
O
CH3 CH 3
H S
NH2 + N O S
N

O O O
O
Antibody molecule SATA Modification producing
containing amine groups N OH protected sulfhydryl groups

O
NHS
NH2OH

O
H SS
SS
N N
E + H
O N SH
O
O

Maleimide-activated enzyme Deprotection of sulfhydryl

SS
O
N
E
SS
N O
H O
N S

Antibody–enzyme conjugate
formation through thioether bond

FIGURE 20.7  Available amine groups on an antibody molecule may be modified with the NHS ester end of SATA to produce amide bond
derivatives containing terminal protected sulfhydryls. The acetylated thiols may be deprotected by treatment with hydroxylamine at alkaline
pH. Reaction of the thiolated antibody with a maleimide-activated enzyme results in thioether crosslinks.

Conjugation with SATA-Modified Antibodies and deacetylated as needed. Unlike thiolation proce-
N-Succinimidyl-S-acetylthioacetate (SATA) is a dures which immediately form a free sulfhydryl resi-
thiolation reagent described in detail in Chapter  2, due, the protected sulfhydryl group of SATA-modified
Section 4.1. The compound reacts with primary amines proteins is stable to long-term storage without
via its NHS ester end to form stable amide linkages. The degradation.
acetylated sulfhydryl group is stable until deacetylated Although amine-reactive protocols, such as SATA
with hydroxylamine. Thus, antibody molecules may thiolation, result in nearly random attachment over the
be thiolated with SATA to create the sulfhydryl target surface of the antibody structure, it has been shown that
groups necessary to couple with a maleimide-activated modification with up to six SATAs per antibody mol-
enzyme (Figure 20.7). Using this reagent, stock prepa- ecule typically results in no decrease in antigen bind-
rations of SATA-modified antibodies may be prepared ing activity (Duncan et  al., 1983). Even higher ratios of

BIOCONJUGATE TECHNIQUES
878 20.  Antibody Modification and Conjugation

SATA to antibody are possible with excellent retention 6. Deprotect the acetylated sulfhydryl groups on the
of activity. SATA-modified antibody according to the following
The following protocol should be compared to the protocol:
method described for SATA thiolation in Chapter  2, a. Prepare a 0.5-M hydroxylamine (Thermo Fisher)
Section 4.1. Although the procedures are slightly dissim- solution in 0.1-M sodium phosphate, pH 7.2,
ilar, the differences indicate the flexibility inherent in the containing 10-mM EDTA.
chemistry. For convenience, the buffer composition indi- b. Add 100 μl of the hydroxylamine stock solution
cated here was chosen to be consistent throughout this to each ml of the SATA-modified antibody. Final
section on enzyme–antibody conjugation using SMCC. concentration of hydroxylamine in the antibody
Other buffers and alternate protocols can be found in solution is 50-mM.
the literature. c. React for 2 h at room temperature.
d. Purify the thiolated antibody by gel filtration on
Protocol a desalting resin using 0.1-M sodium phosphate,
1. Dissolve the antibody to be modified in 0.1-M 0.1-M NaCl, pH 7.2, containing 10-mM EDTA as
sodium phosphate, 0.15-M NaCl, pH 7.2, at a the chromatography buffer. To obtain efficient
concentration of 1 to 5 mg/ml. Note: Phosphate separation between the thiolated antibody and
buffers at various pH values between 7.0 and 7.6 excess hydroxylamine and reaction byproducts,
have been used successfully with this protocol. the sample size applied to the column should
Other mildly alkaline buffers may be substituted be at a ratio of no more than 5% sample volume
for phosphate in this reaction, providing they to the total column volume. Collect 0.5-ml
do not contain extraneous amines (e.g., Tris) or fractions. Pool the fractions containing protein
promote hydrolysis of SATA’s NHS ester (e.g., by measuring the absorbance of each fraction at
imidazole). 280 nm.
2. Prepare a stock solution of SATA (Thermo Fisher) 7. Immediately mix the thiolated antibody with an
by dissolving it in DMF or DMSO at a concentration amount of maleimide-activated enzyme to obtain
of 8 mg/ml. Use a fume hood to handle the organic the desired molar ratio of antibody-to-enzyme in
solvents. the conjugate. Use of a 4:1 to 15:1 (enzyme:antibody)
3. Add 10 to 40 μl of the SATA stock solution per molar ratio in the conjugation reaction usually results
milliliter of 1 mg/ml antibody solution. This will in high-activity conjugates suitable for use in many
result in a molar excess of approximately 12- to enzyme-linked immunoassay procedures.
50-fold of SATA over the antibody concentration 8. React for 30 to 60 min at 37°C or 2 h at room
(for an initial antibody concentration of 1 mg/ml). A temperature. The conjugation reaction also may be
12-fold molar excess works well, but higher levels performed at 4°C overnight.
of SATA incorporation will potentially result in 9. The conjugate may be further purified away from
more maleimide-activated enzyme molecules able unconjugated enzyme by the procedures described
to couple to each thiolated antibody molecule. For in Section 1.5. For storage, the conjugate should be
higher concentrations of antibody in the reaction kept frozen, lyophilized, or sterile filtered and kept
medium, proportionally increase the amount of at 4°C. Stability studies may have to be carried out to
SATA addition; however, do not exceed 10% DMF determine the optimal method of long-term storage
in the aqueous reaction medium. for a particular conjugate.
4. React for 30 min at room temperature.
5. To purify the SATA-modified antibody,
1.2.  Glutaraldehyde-Mediated Conjugation
perform a gel filtration separation using
desalting resin or by dialysis against 0.1-M sodium Glutaraldehyde was one of the first crosslinking
phosphate, 0.15-M NaCl, pH 7.2, containing agents used for creating antibody–enzyme conjugates,
10-mM EDTA. Purification is not absolutely and it is still being used today in manufacturing pro-
required, since the following deprotection step is cesses despite its shortcomings. The crosslinking pro-
performed using hydroxylamine at a significant cess using glutaraldehyde is believed to proceed by
molar excess over the initial amount of SATA added. a number of mechanisms, including Schiff base for-
Whether a purification step is carried out or not, mation with possible rearrangement to a stable prod-
at this point, the derivative is stable and may be uct, addition to hemiacetal ring structures, or through
stored under conditions which favor long-term a Michael-type addition reaction that takes place at
antibody activity (i.e., sterile filtered at 4°C, frozen, points of double-bond unsaturation created by polym-
or lyophilized). erization of the reagent in solution (see Chapter  2,

BIOCONJUGATE TECHNIQUES
1.  Preparation of Antibody–Enzyme Conjugates 879

H H
+ E + O O

Antibody Enzyme Glutaraldehyde

NaCNBH3

HN
E H
H N N N
N H H
H
H N
HN N
E H
E N
N N
H H N N
H H H
H N
H N
N
H H E
H N N
N
Large polymeric H
N
E conjugate

FIGURE 20.8  Glutaraldehyde antibody–enzyme crosslinking procedures usually produce a wide range of high-molecular-weight com-
plexes, some of which may precipitate from solution. Note that there are many additional crosslinked forms that may occur upon crosslinking,
including species that involve addition reactions to hemiacetal and aldol forms of glutaraldehyde which may involve high-molecular-weight
polymers with multiple attachment points.

Section 4.4; Chapter  5, Section 6.2; Chapter  15, Section of insoluble polymer which causes yield and activity
2.1) (Avrameas, 1969). Reduction of Schiff base interme- losses in the preparation of antibody–enzyme conjugates.
diates is also possible using sodium cyanoborohydride This is especially true when the conjugation is performed
to form stable secondary amine linkages. using the one-step method where glutaraldehyde is sim-
The problem of indeterminate reaction products is ply added to a solution containing the two proteins to be
a deficiency that plagues all conjugations performed crosslinked (Figure 20.8). Enzymatic activity yields using
using glutaraldehyde. Part of this difficulty is due to this process can be as little as 10% in the final antibody–
the reagent’s bifunctional nature, but a significant part enzyme conjugate. To somewhat overcome the polym-
of the problem is also due to the ambiguous nature of erization problem, a two-step procedure was developed
the commercial product. In aqueous solutions at alka- which involves first activating one of the proteins with
line pH, glutaraldehyde can undergo hemiacetal cycli- glutaraldehyde, purifying the intermediate from excess
zation and aldol condensation reactions with itself to reagent, and then adding the second protein to effect the
form a variety of structures, some of which contain final conjugation. Unfortunately, even when using the
α, β-unsaturated aldehydes (Hardy et  al., 1969, 1976). two-step method, it results in significant formation of
Another disadvantage of the reagent is the tendency to large-molecular-weight species that may precipitate out
form high-molecular-weight conjugates due to uncon- of solution. The only enzyme that the two-step method
trollable polymerization during the crosslinking process. seems to work well with is HRP, since it only contains a
The resultant conjugates often have a significant amount limited number of available lysine amine groups.

BIOCONJUGATE TECHNIQUES
880 20.  Antibody Modification and Conjugation

Despite these deficiencies, antibody–enzyme conju- alternative to reduction, add 50 μl of 0.2-M lysine in
gates are still being made using glutaraldehyde—par- 0.5-M sodium carbonate, pH 9.5 to each milliliter of
ticularly for many commercial diagnostic ELISA kits the conjugation reaction to block excess reactive sites.
which were developed before the advent of more con- Block for 2 h at room temperature. Other amine-
trollable, heterobifunctional crosslinking procedures. containing small molecules may be substituted for
Today, choosing another method of producing anti- lysine—such as glycine, Tris buffer, or ethanolamine.
body–enzyme conjugates will result in much better con- 8. Reduce for 1 h at 4°C.
jugates of higher activity and higher yield. 9. To remove any insoluble polymers that may have
formed, centrifuge the conjugate or filter it through a
One-Step Glutaraldehyde Protocol 0.45-μm filter. Purify the conjugate by gel filtration or
1. Prepare a solution containing 2 mg/ml antibody and dialysis using PBS, pH 7.4.
5 mg/ml enzyme in 0.02-M sodium phosphate, 0.15-
M NaCl, pH 7.4, chilled to 4°C. 1.3.  Reductive Amination-Mediated
2. In a fume hood, add 10 μl of 25% glutaraldehyde Conjugation
(Sigma) per milliliter of antibody/enzyme solution.
Mix well. Oxidation of polysaccharide residues in glycopro-
3. React for 2 h at 4°C. teins with sodium periodate provides an efficient way
4. To reduce the resultant Schiff bases and any excess of generating reactive aldehyde groups for subsequent
aldehydes, add sodium borohydride (Aldrich) conjugation with amine- or hydrazide-containing mol-
to a final concentration of 10 mg/ml. Note: Some ecules via reductive amination (Chapter  2, Section 4.4,
protocols do not call for a reduction step. As an and Chapter 4, Section 4). Some selectivity of monosac-
alternative to reduction, add 50 μl of 0.2-M lysine in charide oxidation may be accomplished by regulating
0.5-M sodium carbonate, pH 9.5, to each milliliter of the concentration of periodate in the reaction medium.
the conjugation reaction to block excess reactive sites. In the presence of 1-mM sodium periodate at approxi-
Block for 2 h at room temperature. Other amine- mately 0°C, sialic acid groups will be preferentially oxi-
containing small molecules may be substituted for dized at their adjacent hydroxyl residues on the Nos.
lysine—such as glycine, Tris buffer, or ethanolamine. 7, 8, and 9 carbon atoms, cleaving off two molecules of
5. Reduce for 1 h at 4°C. formaldehyde and leaving one aldehyde group on the
6. To remove any insoluble polymers that may have No. 7 carbon. At higher concentrations of sodium peri-
formed, centrifuge the conjugate or filter it through a odate (10-mM or greater) at room temperature, other
0.45-μm filter. Purify the conjugate by gel filtration or sugar residues will be oxidized at points where adjacent
dialysis using PBS, pH 7.4. carbon atoms contain hydroxyl groups.
Thus, glycoproteins such as horseradish peroxidase,
glucose oxidase, or most antibody molecules can be
Two-Step Glutaraldehyde Protocol activated for conjugation by brief treatment with peri-
1. Dissolve the enzyme at a concentration of 10 mg/ml odate. Crosslinking with an amine-containing protein
in 0.1-M sodium phosphate, 0.15-M NaCl, pH 6.8. takes place under alkaline pH conditions through the
2. Add glutaraldehyde to a final concentration of formation of Schiff base intermediates. These relatively
1.25%. labile intermediates can be stabilized by reduction to a
3. React overnight at room temperature. secondary amine linkage with sodium cyanoborohy-
4. Purify the activated enzyme from excess dride (Figure 20.9).
glutaraldehyde by gel filtration using a desalting The use of periodate coupling chemistry for HRP
resin or by dialysis against PBS, pH 6.8. first was introduced by Nakane and Kawaoi (1974; see
5. Dissolve the antibody to be conjugated at a also Nakane, 1975). In the first step of their protocol, the
concentration of 10 mg/ml in 0.5-M sodium few amine groups on HRP were initially blocked with
carbonate, pH 9.5. Mix the activated enzyme with 2,4-dinitrofluorobenzene (DNFB) before periodate oxi-
the antibody at the desired molar ratio to effect dation. The blocking step was designed to eliminate
the conjugation. Mixing the equivalent of 4 mg of the possibility of self-conjugation of enzyme molecules
enzyme per milligram of antibody usually results in during reductive amination with an immunoglobulin.
acceptable conjugates. However, Boorsma and Streefkerk (1976a,b) determined
6. React overnight at 4°C. that HRP can still dimerize even after DNFB blocking,
7. To reduce any Schiff bases that may have formed as perhaps by a mechanism similar to Mannich conden-
well as reduce any excess aldehydes, add sodium sation (Chapter  3, Section 6.2) or through aldol forma-
borohydride to a final concentration of 10 mg/ml. tion. In fact, amine-blocked, periodate-oxidized HRP
Note: Some protocols avoid a reduction step. As an will form insoluble complexes during storage after just

BIOCONJUGATE TECHNIQUES
1.  Preparation of Antibody–Enzyme Conjugates 881

HO E HO E
O O
NaIO4
O O O O

HO OH O O

HRP containing Oxidation producing


polysaccharide chains reactive aldehyde groups
NaCNBH3

O
E
NH
NH2
O
O

Antibody molecule
O
H

containing amine groups


Reductive amination coupling
forming secondary aminelinkage

FIGURE 20.9  Enzymes that are glycoproteins like HRP may be oxidized with sodium periodate to produce reactive aldehyde residues.
Conjugation with an antibody then may be done by reductive animation using sodium cyanoborohydride.

weeks in solution at room temperature or at 4°C, indi- through membrane barriers is an important consid-
cating that another route of conjugation is taking place. eration. Washing steps also more effectively remove
Reductive amination crosslinking has been carried excess reagent if the conjugate is of low molecular
out using sodium borohydride or sodium cyanobo- weight, thus maintaining a low background signal in an
rohydride; however, cyanoborohydride is the better assay. By contrast, conjugates of high molecular weight
choice since it is more specific for reducing Schiff bases are more appropriate for ELISA procedures in a micro-
and will not reduce aldehydes. Small blocking agents plate or array format, where high sensitivity is impor-
such as lysine, glycine, ethanolamine, or Tris can be tant, but washing off excess conjugate is not a problem.
added after conjugation to quench any unreacted alde- The protocols appearing in the literature vary
hyde sites (Mannik and Downey, 1973; Mattiasson and according to the amount of periodate used during poly-
Nilsson, 1977; Barbour, 1976). Ethanolamine and Tris saccharide oxidation, the type of reductant and block-
are the best choices for blocking agents, since they con- ing agent employed for reductive amination, and the
tain hydrophilic hydroxyl groups with no charged func- pH at which the various reactions are done. This vari-
tional groups. ability indicates considerable flexibility in the protocols,
The pH of the reductive amination reaction can be all of which yield usable antibody–enzyme conjugates.
controlled to affect the efficiency of the crosslinking There are, however, several conclusions that can be
process and the size of the resultant antibody–enzyme drawn from these studies: Investigations performed
complexes formed. At physiological pH, the initial using HRP indicate the optimal concentration of
Schiff base formation is less efficient and conjugates of sodium periodate during oxidation to be approximately
lower molecular weight will result. At more alkaline pH 4 to 8-mM (Tussen and Kurstak, 1984). This reaction
values (i.e., pH 9–10), Schiff base formation occurs rap- should be performed in the dark to prevent periodate
idly and with high efficiency, resulting in conjugates of breakdown and for a limited period of time (no more
higher molecular weight and greater incorporation of than 15–30 min) to avoid loss of enzymatic activity. The
enzyme (when oxidized HRP is reacted in excess). conjugation reaction should be carried out at alkaline
The ability to select the relative size of the antibody– pH (7.2–9.5) in the presence of a reducing agent to sta-
enzyme complex is important depending on the assay bilize the Schiff base intermediates. If sodium cyano-
application. Low-molecular-weight conjugates may borohydride is used as the reductant, a blocking agent
be more optimal for immunohistochemical staining or should be added at the completion of the conjugation
blotting techniques where penetration of the complex reaction to cap excess aldehyde sites. The following

BIOCONJUGATE TECHNIQUES
882 20.  Antibody Modification and Conjugation

protocol follows these general guidelines and works the fraction collection process may be performed
well especially in the preparation of HRP–antibody visually—just pooling the main colored HRP peak as
conjugates. it comes off the column.
6. Pool the fractions containing protein. Adjust
Activation of Enzymes with Sodium Periodate the enzyme concentration to 10 mg/ml for the
Enzymes that are glycosylated (i.e., HRP and glucose conjugation step (see next section). The periodate-
oxidase) may be oxidized according to the following activated enzyme may be stored frozen or freeze-
method to produce aldehyde groups for reductive ami- dried for extended periods without loss of activity.
nation coupling to an antibody molecule. Do not store the preparation in solution at room
temperature or 4°C, since precipitation will occur
Protocol over time due to self-polymerization.
1. Dissolve the enzyme to be oxidized in water or
0.01-M sodium phosphate, 0.15-M NaCl, pH 7.2, at a Activation of Antibodies with Sodium Periodate
concentration of 10 to 20 mg/ml. Many immunoglobulin molecules are glycopro-
2. Dissolve sodium periodate in water at a teins that can be periodate-oxidized to contain reac-
concentration of 0.088 M. Protect from light. tive aldehyde residues. Polyclonal IgG molecules often
3. Immediately add 100 μl of the sodium periodate contain carbohydrate in the Fc portion of the molecule.
solution to each milliliter of the enzyme solution. This is sufficiently removed from the antigen bind-
This ratio of addition results in an 8-mM periodate ing sites to allow conjugation to take place through the
concentration in the reaction mixture. Mix to polysaccharide chains without compromising activity.
dissolve. Protect from light. Occasionally, however, some antibodies may contain
4. React in the dark for 15 to 20 min at room sites of glycosylation near the antigen binding regions,
temperature. If HRP is the enzyme being oxidized, and in this situation conjugation through these sites
a color change will be apparent as the reaction may affect binding activity. Although antibody–enzyme
proceeds—changing the brownish/gold color of conjugation by reductive amination is typically done
concentrated HRP to green. Limiting the time of by oxidation of the enzyme with subsequent crosslink-
oxidation will help to preserve enzyme activity. ing to an amine-containing antibody, oxidation of the
5. Immediately quench the reaction by the addition antibody with subsequent conjugation to an amine-
of 0.1 ml of glycerol per milliliter of reaction or hydrazide-containing molecule is also possible. It
solution. Instead of glycerol, N-acetylmethionine should be noted, however, that many monoclonal anti-
may be added to quench the reaction, because the bodies are not glycosylated and therefore can not be
thioether of the methionine side chain will react used in this protocol. Recombinant antibodies also do
with periodate to form sulfoxide or sulfone products not contain carbohydrate. A given monoclonal should
(Geoghegan and Stroh, 1992). In addition, sodium be checked to verify the presence of carbohydrate
sulfite (Na2SO3) was used by Stolowitz et al. (2001) to before attempting to use a periodate-mediated conjuga-
quench the periodate oxidation of HRP in solution. tion protocol.
This may be the simplest route to stopping the
reaction, as sulfite is inexpensive and the reduction Protocol
does not form reactive byproducts. Add quenching 1. Dissolve the antibody to be periodate-oxidized
reagent to provide at least a 2-times molar excess at a concentration of 10 mg/ml in 0.01-M sodium
over the amount of periodate initially added to the phosphate, 0.15-M NaCl, pH 7.2.
reaction. Alternatively, the reaction may be stopped 2. Dissolve sodium periodate in water to a final
by immediate gel filtration on a desalting resin. If a concentration of 0.1-M. Protect from light.
dextran-based resin is used for the chromatography, 3. Immediately add 100 μl of the sodium periodate
the support itself will react with sodium periodate to solution to each milliliter of the antibody solution.
quench excess reagent. Purify the oxidized enzyme Mix to dissolve. Protect from light.
by gel filtration using 0.01-M sodium phosphate, 4. React in the dark for 15 to 20 min at room
0.15-M NaCl, pH 7.2. To obtain efficient separation temperature.
between the oxidized enzyme and excess periodate 5. Immediately quench the reaction by the addition of
(or quenching agent), the sample size applied to sodium sulfite (Na2SO3) to provide a 2-times molar
the column should be at a ratio of no more than 5% excess over the initial amount of periodate added.
sample volume to the total column volume. Collect Purify the oxidized antibody by gel filtration using
0.5-ml fractions and monitor for protein at 280 nm. a desalting resin. The chromatography buffer is
HRP also may be detected by its absorbance at 0.1-M sodium phosphate, 0.15-M NaCl, pH 7.2. To
403 nm. When oxidizing large quantities of HRP, obtain efficient separation between the oxidized

BIOCONJUGATE TECHNIQUES
1.  Preparation of Antibody–Enzyme Conjugates 883
antibody and excess periodate, the sample size 6. React for 30 min at room temperature with gentle
applied to the column should be at a ratio of no mixing (in a fume hood).
more than 5% sample volume to the total column 7. Block unreacted aldehyde sites by addition of
volume. Collect 0.5-ml fractions and monitor for 50 μl of 1-M ethanolamine, pH 9.6, per milliliter
protein at 280 nm. of conjugation solution. Approximately a 1-M
6. Pool the fractions containing protein. Adjust ethanolamine solution may be prepared by addition
the antibody concentration to 10 mg/ml for the of 300 μl ethanolamine to 5 ml of deionized water.
conjugation step. The oxidized antibody should be Adjust the pH of the ethanolamine solution by
used immediately. addition of concentrated HCl, keeping the solution
cool on ice.
Conjugation of Periodate-Oxidized HRP to 8. React for 30 min at room temperature.
Antibodies by Reductive Amination 9. Purify the conjugate from excess reactants by dialysis
The following protocol assumes that HRP has or gel filtration using a desalting resin. Use 0.01-M
already been periodate-oxidized by the method given sodium phosphate, 0.15-M NaCl, pH 7.0, as the
in Section 1.3, above. buffer for either operation. The conjugate may be
further purified by removal of unconjugated enzyme
Protocol using one of the methods described in Section 1.5.
1. Dissolve the IgG to be conjugated at a concentration
of 10 mg/ml in 0.2-M sodium bicarbonate, pH 9.6, at Conjugation of Periodate-Oxidized Antibodies with
room temperature. The high pH buffer will result in Amine or Hydrazide Derivatives
very efficient conjugation with the highest possible
The following protocol assumes that the antibody
incorporation of enzyme molecules per antibody
has already been periodate-oxidized by the method of
molecule. To produce lower molecular weight
Section 1.3 (above) to create reactive aldehyde groups
conjugates, dissolve the IgG at a concentration of
suitable for coupling with amine-containing or hydra-
10 mg/ml in 0.1-M sodium phosphate, 0.15-M NaCl,
zide-containing molecules. This is an excellent method
pH 7.2.
for directing the antibody modification reaction away
2. The periodate-oxidized enzyme (HRP) prepared in
from the antigen binding sites, if the antibody glycosyl-
Section 1.3 was finally purified using 0.01-M sodium
ation points are solely in the Fc region of the molecule.
phosphate, 0.15-M NaCl, pH 7.2. For conjugation
For instance, biotinylation of intact antibodies can be
using the lower-pH-buffered environment, this
performed after mild periodate treatment using biotin-
HRP preparation can be used directly at 10 mg/ml
hydrazide (Chapter  11, Section 6.4) (Figure 20.10). It
concentration. For conjugation using the higher pH
should be noted, however, that periodate-oxidized anti-
carbonate buffer, dialyze the HRP solution against
bodies can self-conjugate through their own amines if
0.2-M sodium carbonate, pH 9.6 for 2 h at room
high-pH reductive amination is used. Conjugation with
temperature prior to use. A volume of HRP solution
periodate-oxidized antibodies works best if the receiv-
equal to the volume of antibody solution will be
ing molecule is modified to contain hydrazide groups
required.
and the reaction is carried out at more moderate pH
3. Mix the antibody solution with the enzyme solution
values (e.g., slightly acidic to neutral pH).
at a ratio of 1:1 (v/v). Since an equal mass of antibody
and enzyme is present in the final solution, this will 1. For conjugation to hydrazide-containing proteins,
result in a 3.75 molar excess of HRP over the amount dissolve the periodate-oxidized antibody at
of IgG. For conjugates consisting of greater enzyme- a concentration of 10 mg/ml in 0.1-M sodium
to-antibody ratios, proportionally increase the phosphate, 0.15-M NaCl, pH 6.0 to 7.2. For
amount of enzyme solution as required. Typically, conjugation to amine-containing molecules and
molar ratios of 4:1 to 15:1 (enzyme:antibody) give proteins, dissolve the oxidized antibody at 10 mg/ml
acceptable conjugates useful in a variety of ELISA in 0.2-M sodium carbonate, pH 9.6.
techniques. 2. Dissolve a hydrazide-containing enzyme or other
4. React for 2 h at room temperature. protein at a concentration of 10 mg/ml in 0.1-M
5. In a fume hood, add 10 μl of 5-M sodium sodium phosphate, 0.15-M NaCl, pH 6.0 to 7.2. For
cyanoborohydride (Sigma) per milliliter of reaction the preparation of a hydrazide-activated enzyme,
solution. Caution: Cyanoborohydride is extremely see Chapter 22, Section 2.4. For modification with
toxic. All operations should be carried out with a hydrazide-containing probe, such as biotin–
care in a fume hood. Also, avoid any contact with hydrazide, use a concentration of 5-mM in the
the reagent, as the 5-M solution is prepared in 1-N phosphate buffer. For conjugation through the amine
NaOH. groups of a secondary molecule, dissolve the

BIOCONJUGATE TECHNIQUES
884 20.  Antibody Modification and Conjugation

HO NaIO4
HO
O
O
O O
O O

HO OH O O
Antibody molecule Oxidation producing
containing polysaccharide chains reactive aldehyde groups

HN NH
H
N
H2N

O S
Biotin hydrazide
HO
O O
O O
HN NH
H
O N N

O S
Biotinylation through
hydrazone bond formation

FIGURE 20.10  Polysaccharide groups on antibody molecules may be oxidized with periodate to create aldehydes. Modification with bio-
tin–hydrazide results in hydrazone linkages. The sites of modification using this technique are often away from the antibody–antigen binding
regions, thus preserving antibody activity.

amine-containing protein at 10 mg/ml in 0.2-M 6. React for 30 min at room temperature (in a fume
sodium carbonate, pH 9.6. hood).
3. Mix the antibody solution from step 1 with the 7. Block unreacted aldehyde sites by addition of
protein solution from step 2 in amounts necessary to 50 μl of 1-M ethanolamine, pH 9.6, per milliliter
obtain the desired molar ratio for conjugation. Often, of conjugation solution. Approximately a 1-M
the secondary molecule is reacted in approximately ethanolamine solution may be prepared by addition
a 4- to 15-fold molar excess over the amount of of 300 μl ethanolamine to 5 ml of deionized water.
antibody present. Adjust the pH of the ethanolamine solution by
4. React for 2 h at room temperature. addition of concentrated HCl, while keeping the
5. In a fume hood, add 10 μl of 5-M sodium solution cool on ice.
cyanoborohydride (Sigma) per milliliter of reaction 8. React for 30 min at room temperature.
solution. Caution: Cyanoborohydride is extremely 9. Purify the conjugate from excess reactants by dialysis
toxic. All operations should be carried out with or gel filtration using desalting resin. Use 0.01-M
care in a fume hood. Also, avoid any contact with sodium phosphate, 0.15-M NaCl, pH 7.0, as the
the reagent, as the 5-M solution is prepared in 1-N buffer for either operation. The conjugate may be
NaOH. The addition of a reductant is necessary for further purified by removal of unconjugated enzyme
stabilization of the Schiff bases formed between an by one of the methods given in Section 1.5.
amine-containing protein and the aldehydes on the
antibody. For coupling to a hydrazide-activated
1.4.  Conjugation Using Antibody Fragments
protein, however, most protocols do not include a
reduction step. Even so, hydrazone linkages may be It is often advantageous to use antibody fragments
further stabilized by cyanoborohydride reduction. in the preparation of antibody–enzyme conjugates.
The addition of a reductant during hydrazide/ Selected fragmentation carried out by enzymatic diges-
aldehyde reactions also increases the efficiency and tion of intact immunoglobulins can yield lower-molec-
yield of the reaction. ular-weight molecules still able to recognize and bind

BIOCONJUGATE TECHNIQUES
1.  Preparation of Antibody–Enzyme Conjugates 885

SS
SS

SS F(ab’)2 Fragment
Pepsin
SS
+

IgG Molecule
Digested Fc Fragment

FIGURE 20.11  Digestion of IgG-class antibodies with pepsin results in heavy-chain cleavage below the disulfide groups in the hinge
region. The bivalent fragments that are formed are called F(ab′)2. The remaining Fc region normally is severely degraded into smaller peptide
fragments.

antigen. Conjugation of these fragments with enzyme et al. (1992). The following protocol describes the use of
molecules can result in ELISA reagents that possess pepsin to cleave IgG molecules at the C-terminal side of
better characteristics than corresponding conjugates the inter-heavy-chain disulfides in the hinge region, pro-
prepared with intact antibody. Such antibody fragment ducing a bivalent antigen binding fragment, F(ab′)2, with
conjugates display less interference with various Fc a molecular weight of about 105,000 (Figure 20.11). Using
binding proteins and also less immunogenicity (due to this enzyme, most of the Fc fragments undergo extensive
lack of the Fc region), more facile membrane penetra- degradation and cannot be recovered intact.
tion for immunohistochemical staining techniques (due
to lower overall conjugate molecular weight) (Wilson Protocol for Preparation of F(ab′)2 Fragments
and Nakane, 1978; Farr and Nakane, 1981), and lower Using Pepsin
nonspecific binding to surfaces or membranes (result- 1. Equilibrate by washing 0.25 ml of immobilized
ing in increased signal-to-noise ratios) (Hamaguchi pepsin (Thermo Fisher) with 4 × 1 ml of 20-mM
et al., 1979; Ishikawa et al., 1981a,b). sodium acetate, pH 4.5 (digestion buffer). Finally,
Enzymatic digests of IgG can result in two particularly suspend the gel in 1 ml of digestion buffer.
useful fragments called Fab and F(ab′)2, prepared by the 2. Dissolve 1 to 10 mg of IgG in 1 ml digestion buffer
action of papain and pepsin, respectively. Most specific and add it to the gel suspension.
enzymatic cleavages of IgG occur in relatively unfolded 3. Mix the reaction slurry in a shaker at 37°C for 2 to
regions between the major domains. Papain and pep- 48 h. The optimal time for complete digestion varies
sin, and similar enzymes including bromelain, ficin, and depending on the IgG subclass and species of origin.
trypsin, cleave immunoglobulin molecules in the hinge Mouse IgG1 antibodies are usually digested within
region of the heavy-chain pairs. Depending on the loca- 24 h, human antibodies are fragmented in 12 h,
tion of cleavage, the disulfide groups holding the heavy whereas some minor subclasses (e.g., mouse IgG2a)
chains together may or may not remain attached to the require a full 48-h digestion period.
antigen-binding fragments that result. If the disulfide 4. After the digestion is complete, add 3 ml of 10-mM
bonded region does remain with the antigen binding frag- Tris–HCl, pH 8.0, to the gel suspension. Separate the
ment, as in pepsin digestion, then a divalent molecule is gel from the antibody solution using filtration or by
produced (F(ab′)2) which differs from the intact antibody centrifugation.
by lack of an extended Fc portion. If the disulfide region 5. Apply the fragmented IgG solution to an
is below the point of digestion, then the two heavy-/light- immobilized protein A column containing 2 ml of gel
chain complexes that form the two antigen binding sites of (Thermo Fisher) that was previously equilibrated
an antibody are cleaved and released, forming individual with 10-mM Tris–HCl, pH 8.0.
dimeric fragments (Fab) containing one antigen binding 6. After the sample has entered the gel, wash the
site each (see Figure 20.4, discussed previously). column with 10-mM Tris–HCl, pH 8.0, while
Methods for producing immobilized papain or pepsin collecting 2-ml fractions. The fractions may be
for antibody fragmentation can be found in Hermanson monitored for protein by measuring absorbance at

BIOCONJUGATE TECHNIQUES
886 20.  Antibody Modification and Conjugation

Fab Fragments

+
SS
Papain
SS
SS
SS

IgG Molecule
Intact Fc Fragment

FIGURE 20.12  Papain digestion of IgG antibodies primarily results in cleavage in the hinge region above the interchain disulfides. This pro-
duces two heavy–light chain pairs, called Fab fragments, each containing one antigen binding site. The Fc region can normally be recovered intact.

280 nm. The protein peak eluting unretarded from 4. After the required time of digestion, add 3.0 ml
the column is F(ab′)2. 10-mM Tris–HCl buffer, pH 8.0, to the gel
7. Bound Fc or Fc fragments and any undigested IgG suspension, mix, and then separate the digest
may be eluted from the column with 0.1-M glycine, solution from the gel by filtration or centrifugation at
pH 2.8. 2000 g for 5 min.
5. Apply the supernatant liquid to an immobilized
Similarly, immobilized papain may be used to gen-
protein A column (2 ml gel) (Thermo Fisher) which
erate Fab fragments from immunoglobulin molecules.
was previously equilibrated by washing with 20 ml
Papain is a sulfhydryl protease that is activated by the
of 10-mM Tris–HCl buffer, pH 8.0.
presence of a reducing agent. Cleavage of IgG occurs
6. After the sample has entered the gel bed, wash the
above the disulfides in the hinge region, creating two
column with 15 ml of 10-mM Tris–HCl buffer, pH
types of fragments, two identical Fab portions and one
8.0, while 2.0-ml fractions are collected. Monitor the
intact Fc fragment (Figure 20.12). For preparation of the
fractions for protein by their absorbance at 280 nm.
immobilized papain gel used in the following protocol,
The protein eluted unretarded from the column is
see Hermanson et  al. (1992). The gel is also commer-
purified Fab.
cially available from Thermo Fisher.
7. Elute Fc and undigested IgG bound to the
Protocol for Preparation of Fab Fragments using immobilized protein A column with 0.1-M glycine–
Papain HCl buffer, pH 2.8.
1. Wash 0.5 ml of immobilized papain (Thermo Fisher) Conjugation of these fragments with enzymes is car-
with 4 × 2 ml of 20-mM sodium phosphate, 20-mM ried out using similar methods to those previously dis-
cysteine-HCl, 10-mM EDTA, pH 6.2 (digestion cussed for intact antibody molecules. F(ab′)2 fragments
buffer), and finally suspend the gel in 1.0 ml of may be selectively reduced in the hinge region with
digestion buffer. DTT, TCEP, or MEA using the identical protocols out-
2. Dissolve 10 mg of human IgG solution in 1.0 ml lined for whole antibody molecules (Chapter 2, Section
of digestion buffer and add it to the immobilized 4.1, and Section 1.1, this chapter). Mild reduction results
papain gel suspension. in cleaving the disulfides holding the heavy-chain pairs
3. Mix the gel suspension in a shaker at 37°C for 4 to together at the central portion of the fragment, thus cre-
48 h. Maintain the gel in suspension during mixing. ating two F(ab′) fragments each containing one antigen
The optimal time for complete digestion varies binding site (Figure 20.13).
depending on the IgG subclass and species of origin. The amine groups on these fragments also may be
Mouse IgG1 antibodies are usually digested within modified with thiolating agents, such as SATA or 2-imi-
27 h, whereas other mouse subclasses require only nothiolane, to create sulfhydryl residues suitable for
4 h; human antibodies are fragmented in 4 h (IgG1 coupling to maleimide-activated enzymes (Section 1.1)
and IgG3), 24 h (IgG4), or 48 h (IgG2); and bovine, (Figure 20.14). Amine groups may be further utilized
sheep, and horse antibodies are somewhat resistant in reductive amination coupling to periodate-oxidized
to digestion and require a full 48 h. glycoproteins, such as in the protocol outlined for HRP

BIOCONJUGATE TECHNIQUES
1.  Preparation of Antibody–Enzyme Conjugates 887

SS 2-MEA SH HS
SS SH
+ HS

Bivalent F(ab’)2 fragment Reduction to monovalent Fab’


containing disulfides fragments with free sulfhydryls

O
H
N N E
O
O

Maleimide-activated enzyme

O
S H
N N E
SH
O
O

Antibody–enzyme conjugate
formation through thioether bond

FIGURE 20.13  F(ab′)2 fragments produced by pepsin digestion of IgG can be reduced at their heavy-chain disulfides using a reducing
agent, such as 2-mercaptoethylamine (MEA), DTT, or TCEP. Conjugation then can be carried out with a maleimide-activated enzyme to produce
relatively low-molecular-weight complexes linked by thioether bonds.

conjugation, previously (Section 1.3, this chapter) (Figure In the preparation of such conjugates, a molar excess of
20.15). Successful periodate oxidation of the fragments enzyme is typically crosslinked to a specific antibody
themselves, however, may not be possible unless they to obtain a complex of high activity. As a result of this
contain carbohydrate in the antigen binding region, ratio, there is typically excess enzyme left unconjugated
which is true for some polyclonal antibodies. Finally, after completion of the reaction. The unconjugated
glutaraldehyde-mediated conjugation techniques will enzyme confers nothing to the utility of the final prod-
work with antibody fragments, but are not recom- uct and can be detrimental if it contributes to increased
mended due to the reasons discussed in Section 1.2. backgrounds in assay procedures. The removal of
The primary goal of any of these conjugation strate- this free enzyme component may be advantageous to
gies using antibody fragments is to maintain the activ- improving the resultant signal-to-noise ratio in some
ity of the antigen binding site while limiting the size of immunoassays. Commercial preparations of antibody–
the final complex with a second molecule. The use of enzyme conjugates usually are not purified to remove
heterobifunctional crosslinkers such as SMCC or reduc- unconjugated enzyme. Frequently, the major protein-
tive amination techniques allows sufficient control over aceous part of these products is not active conjugate,
the process to realize these goals. but leftover enzyme that contributes nothing to the
immunochemical activity of what was purchased.
1.5.  Removal of Unconjugated Enzyme from Boorsma and Kalsbeek (1976) state that unconju-
gated HRP must be removed from antibody–enzyme
Antibody–Enzyme Conjugates
conjugates to obtain optimal staining in immunoas-
Conjugates of antibodies and enzymes are essential say procedures. This is especially true in blotting tech-
components in immunoassay and detection systems. niques and cytochemical staining where free enzyme

BIOCONJUGATE TECHNIQUES
888 20.  Antibody Modification and Conjugation

Antigen binding
region

O O O
CH3 CH3
S H S
NH2 N O N
+
O O O
O
Fab fragment SATA Modification producing
containing amine groups N OH protected sulfhydryl groups
(from papain digestion of IgG)
O
NHS
NH2OH

O
H
N N E
+ H SH
N
O
O
O

Maleimide-activated enzyme Deprotection of sulfhydryl

H
N
O E
N O
H O
N S

Fab–enzyme conjugate
formation through thioetherbond

FIGURE 20.14  The thiolation reagent SATA can be used to create sulfhydryl groups on Fab fragments. After deprotection of the acetylated
thiol of SATA with hydroxylamine, conjugation with a maleimide-activated enzyme can take place, producing thioether linkages.

may become entrapped nonspecifically within the efficient way of removing free enzyme. However, gel
membrane or cellular structures. The presence of this filtration procedures can be time consuming and of
unconjugated enzyme leads to diffuse substrate noise relatively low capacity for the amount of gel required.
that can obscure the immunospecific signal. In addition, separation of higher-molecular-weight
Several methods may be used to purify an antibody– enzymes from antibody conjugates, such as in the case
enzyme conjugate and remove unconjugated enzyme. of AP (MW 140,000), is considerably less efficient or
For instances where the enzyme molecular weight is impossible. Gel filtration separation also becomes a
significantly different than the conjugate molecular problem if the conjugate itself consists of a broad range
weight, separation may be achieved by gel filtration of molecular weights, as is often true when glutaralde-
chromatography. Using the proper support with an hyde is used as the crosslinking agent.
exclusion limit and separation range able to accommo- The most effective methods of removing unconju-
date all the proteins in the sample, the conjugate peak gated enzyme all make use of affinity chromatography
will elute before the enzyme peak, thus providing an systems using specific ligands that can interact with the

BIOCONJUGATE TECHNIQUES
1.  Preparation of Antibody–Enzyme Conjugates 889

E E
HO HO
O O
O
NaIO4
O O O

HO OH O O

HRP containing Oxidation producing


polysaccharide chains reactive aldehyde groups
NaCNBH3

O
NH2
NH E

O Fab fragment
O

containing amine groups


O

(from papain digestion of IgG)


H

Reductive amination coupling


forming secondary amine linkage

FIGURE 20.15  Periodate oxidation of HRP creates aldehyde groups on the carbohydrate chains of the enzyme. Reaction with a Fab frag-
ment may then be done using reductive amination to produce a lower-molecular-weight complex than would be obtained using intact IgG
antibodies.

antibody portion of the conjugate. Thus, the supports preparation of an antibody–enzyme conjugate, the
retain any unconjugated antibody (usually in very low antibody binding capability of the crosslinked com-
percentage when the enzyme is reacted in excess) as plex toward its complementary antigen ideally remains
well as the antibody–enzyme conjugate produced from intact. This highly specific interaction can be used to
the crosslinking reaction. The unconjugated enzyme, purify the conjugate from excess enzyme if the anti-
however, passes through such affinity columns unre- body and enzyme can survive the conditions necessary
tarded. Two main methods are discussed below: (1) for binding and elution from such a column. Binding
affinity chromatography, which makes use of immobi- conditions are typically mild physiological pH condi-
lized immunoglobulin binding proteins or immobilized tions which cause no difficulty. However, many elu-
antigen molecules having specificity for the antibody tion conditions require acidic or basic conditions or the
used in the conjugate; and (2) nickel-chelate affinity presence of a chaotropic agent to deform the antigen
chromatography, which binds the Fc region of antibody binding site. Sometimes these conditions can irrevers-
molecules. ibly damage the antigen binding recognition capabil-
The use of immunoaffinity techniques (whether anti- ity of the antibody or denature the active site of the
gen specific or immunoglobulin binding proteins such enzyme, thus diminishing enzymatic activity. Activity
as protein A) allows strong binding of the antibody con- losses for both the antibody and enzyme can be severe
jugate, but they have the significant disadvantage of under such circumstances.
requiring elution conditions that are often too severe for Another potential disadvantage of an immunoaffin-
maintaining activity of the antibody or enzyme com- ity separation is the assumed abundance of the puri-
ponents. By contrast, nickel-chelate affinity techniques fied antigen in sufficient quantities to immobilize on a
give excellent binding of the conjugate while allowing chromatography support (see Chapter  15 for common
free enzyme to pass through the gel unretarded. It also immobilization techniques). Protein antigens should be
has the significant advantage of having mild elution immobilized at densities of at least 2 to 3 mg/ml of affin-
conditions which preserve the activity of the conjugate. ity gel to produce supports of acceptable capacity for
binding antibody. Often, the antigen is too expensive or
Immunoaffinity Chromatography scarce to obtain in the amounts needed.
Immunoaffinity chromatography makes use of However, if the antigen is abundant and inexpen-
immobilized antigen molecules to bind and separate sive and the antibody–enzyme complex will survive
specific antibody from a complex mixture. After the the associated elution conditions, then immunoaffinity

BIOCONJUGATE TECHNIQUES
890 20.  Antibody Modification and Conjugation

chromatography can provide a very efficient method of


purifying a conjugate from excess enzyme. This method
also ensures that the recovered antibody still retains its
ability to bind specific target molecules (i.e., the antigen
binding site was not blocked during conjugation). The
preparation of immunoaffinity supports can be found O
in Hermanson et  al. (1992) and in Chapter  15. A sug- O
O
O E
gested method for performing immunoaffinity chroma- NH
O
N Ni
tography follows: O
O HO
Protocol O O
O
1. Equilibrate the immunoaffinity column with 50-mM O E
O
Tris, 0.15-M NaCl, pH 8.0 (binding buffer). Wash
O
with at least 5 column volumes of buffer. The amount HO
of gel used should be based on the total binding O
O
capacity of the support. A determination of binding O E
O
capacity can be achieved by overloading a small- O
scale column, eluting, and measuring the amount of HO
conjugate that bound. Such an experiment may be
coupled with a determination of conjugate viability
FIGURE 20.16  An affinity chromatography support contain-
for using immunoaffinity as the purification method. ing iminodiacetic acid groups chelated with nickel may be used to
The final column size should represent an amount remove excess enzyme after reactions to produce antibody–enzyme
of gel capable of binding at least 1.5-times more than conjugates. The nickel chelate binds to the antibody in the Fc region,
the amount of conjugate that will be applied. retaining the conjugate while allowing free enzyme to pass through
the gel unretarded.
2. Apply the conjugate to the column in the binding
buffer while taking 2-ml fractions.
3. Wash with binding buffer until the absorbance at and exchangeable coordination sites. These exchange-
280 nm decreases back to baseline. The unbound able sites can then form complexes with coordination
protein flowing through the column will consist of sites on proteins or other molecules. Substances that
mainly unconjugated enzyme. Some conjugate may are able to interact with the immobilized metals will
also flow through if some of the conjugate is inactive bind and be retained on the column. Elution is typi-
or the column is overloaded. cally accomplished by one or a combination of the fol-
4. Elute the bound conjugate with 0.1-M glycine, lowing options: (1) lowering the pH; (2) raising the salt
0.15-M NaCl, pH 2.8, or another suitable elution strength; and/or (3) inclusion of a competing chelating
buffer. A neutral pH alternative to this buffer is the agent such as EDTA or imidazole in the buffer.
Gentle Elution Buffer from Thermo Fisher. If acid Sorensen (1993) reported that a nickel-chelate affinity
pH conditions are used, immediately neutralize the column will specifically bind IgG class immunoglobulins
fractions eluting from the column by the addition of while allowing certain enzymes to pass through the gel
0.5 ml of 1-M Tris, pH 8.0, per fraction. unretarded (Thermo Fisher). This phenomenon allows
the separation of antibody–enzyme complexes containing,
in particular, HRP or alkaline phosphatase conjugated to
Nickel-Chelate Affinity Chromatography common polyclonal or monoclonal antibodies. The nickel
Immobilized metal-chelate affinity chromatog- chelate column binds the conjugate through the Fc region
raphy (IMAC) is a powerful purification technique of the associated antibody, even if enzyme molecules are
whereby proteins or other molecules can be separated covalently attached. Any unconjugated enzyme will pass
based upon their ability to form coordination com- through the affinity column unretarded (Figure 20.16).
plexes with immobilized metal ions (Porath et al., 1975; Elution of the bound antibody–enzyme conjugate
Lonnerdal and Keen, 1982; Porath and Belew, 1983; occurs by only a slight shift in pH to acidic conditions or
Porath and Olin, 1983; Sulkowski, 1985; Kagedal, 1989). through the inclusion of a metal chelating agent like EDTA
The metal ions are stabilized on a matrix through the or imidazole in the binding buffer. Either method of elution
use of organic chelating compounds which usually is mild compared to most immunoaffinity separation tech-
have multivalent points of interaction with the metal niques (discussed in the previous section). Thus, purifica-
atoms. To form useful affinity supports, these metal ion tion of the antibody–enzyme complex can be performed
complexes must have some free or weakly associated without damage to the activity of either component.

BIOCONJUGATE TECHNIQUES
2.  Preparation of Labeled Antibodies 891
One limitation to this method should be noted. If the 2.  PREPARATION OF LABELED
antibody–enzyme conjugate is prepared using antibody ANTIBODIES
fragments such as Fab or F(ab′)2, then nickel chelate
affinity chromatography will not work, since the requi- In addition to labeling immunoglobulins with
site Fc portion of the antibody necessary for complexing enzymes to provide detectability through their cata-
with the metal is not present. lytic action on a substrate, antibody molecules can
The preparation of a metal-chelate affinity support also be labeled or tagged with small compounds that
containing iminodiacetic acid functionalities may be can provide detectable properties, either directly or
found in Hermanson et  al. (1992), or purchased from indirectly. The specificity of the antibody component
a commercial source (see also Chapter  15 for a dis- of the conjugate can then be used to bind unique anti-
cussion on the preparation of immobilized affinity genic determinants, while the attached tag supplies the
ligands, including metal chelate resins). Any metal properties necessary for detection. Such small chemi-
chelate support that is designed to bind His-tagged cal labels are typically one of several types: intense
fusion proteins also will work well in this procedure. fluorescent molecules, contrast agents, affinity tags, or
The following protocol is adapted from the instruc- unstable radioactive isotopes.
tions accompanying the nickel-chelate support. Radiolabeling antibodies with 125I forms the basis
Thermo Fisher offers a kit based on this technology for for most of the original radioimmunoassays (RIA) that
the purpose of removing unconjugated enzyme from were first developed in the early days of immuno-
antibody–enzyme conjugates (called the FreeZyme™ globulin-mediated testing. The use of radioisotopes in
Conjugate Purification Kit). tagging antibodies is used less often today for in vitro
immunoassays due to the hazards associated with
Protocol handling and disposal of radioactive compounds and
1. Pack a column containing an immobilized the better sensitivity offered by using enzyme labels.
iminodiacetic acid support (or another chelating However, radioisotopes are becoming very important
agent designed to bind His-tagged proteins) as monoclonal antibody labels for in vivo diagnostic or
(Thermo Fisher). The column size should be no less therapeutic conjugates for cancer therapy or detection.
than 1.5 times that required to bind the anticipated Radioactive isotopes either directly attached to antibod-
amount of conjugate to be applied. The maximal ies or attached through metal-chelating groups have
capacity of such a column for binding antibody become a significant tactic for the targeting and detec-
can be up to 50 mg/ml gel; however, best results tion of tumor cells in vivo (Chapter  1 and Chapter  12)
are obtained if no more than 10 to 20 mg/ml of (van Dongen et  al., 2012). In addition, a radiolabel
conjugate is applied. may have a distinct advantage over other chemical
2. Dissolve 50 mg of nickel ammonium sulfate per ml tags, because it is not influenced by conformational
of deionized water. Apply 1 ml of nickel solution changes within the antibody molecule or by changes
per ml of gel to the column. Note: The metal salt in its chemical environment, as enzymes or labels with
and all solutions containing it should be considered unique spectral characteristics are often affected. Thus,
hazardous waste and disposed of according to in certain circumstances, radiolabels can still provide an
relevant environmental regulations. important means of detection that approaches the most
3. Wash the column with 10 volumes of water, then sensitive and reliable tags now available, particularly
equilibrate the support with 2 volumes of 10-mM for in vivo imaging purposes.
sodium phosphate, 0.15-M NaCl, pH 7.0 (binding Another form of label often used to tag antibody
buffer). molecules is chemical modification with a biotin group.
4. Dissolve or dialyze the conjugate into binding buffer. Biotinylation (see Chapter 11, Section 6) creates an affin-
Apply the conjugate solution to the column while ity handle on an immunoglobulin with the ability to
collecting 2-ml fractions. bind strongly either avidin or streptavidin in two of the
5. Continue to wash the gel with 0.15-M NaCl (saline most tightly held noncovalent interactions known. With
solution) until the absorbance at 280 nm is down to a dissociation constant (Kd) on the order of 1.3 × 10−15,
baseline. The protein eluting from the column at this the avidin–biotin interaction can be used to detect bio-
point is unconjugated enzyme. tinylated molecules with extreme sensitivity. In this
6. Elute the bound conjugate with 0.1-M sodium type of system, instead of the antibody being directly
acetate, 0.5-M NaCl, pH 5.0. Pool the fractions labeled with a detectable probe, the avidin (or strep-
containing protein, and dialyze the conjugate into tavidin) molecules are modified to contain the detec-
10-mM sodium phosphate, 0.15-M NaCl, pH 7.0, or tion ­complex—consisting of an enzyme, fluorescent
other suitable storage buffers. probe, contrast agent, or radiolabel. Interaction of the

BIOCONJUGATE TECHNIQUES
892 20.  Antibody Modification and Conjugation

NH2

S
NH C
Antibody containing
available amine groups NH

+

O O O O–

O
– O
O O
O–

Thiourea bond formation


(green fluorescent probe)
N
C
S

FITC

FIGURE 20.17  FITC may be used to label amine groups on antibody molecules, forming isothiourea bonds.

biotinylated antibody with its targeted antigen is then In addition to the wide range of commercial probes,
amplified and detected by the addition of such labeled many other fluorescent molecules have been synthe-
avidin or streptavidin reagents. sized and described in the literature. Only a handful,
The following three sections describe the prepara- however, are generally used to label antibody mole-
tion and properties of fluorescent, radiolabeled, and cules. Perhaps the most common fluorescent tags with
biotinylated antibodies. See also Chapter 1 for a review application to immunoglobulin assays are reflected in
of labeled antibody reagents currently being used in a the main derivatives produced by the prominent anti-
broad range of applications in research, diagnostics, body manufacturing companies. These include deriva-
and therapeutics as well as Chapters 10, 11, and 12 for tives of cyanine dyes, fluorescein, rhodamine, Texas
discussions on the labels themselves. red, aminomethylcoumarin (AMCA), and phycobilip-
roteins. Figure 20.17 shows the reaction of fluorescein
isothiocyanate (FITC), one of the most common fluores-
2.1.  Fluorescently Labeled Antibodies
cent probes, with an antibody molecule.
Antibody molecules can be labeled with any one To a large degree, standardization has occurred
of more than a dozen different fluorescent probes cur- in the use of these fluorescent probes due to the large
rently available from commercial sources. Each probe body of published literature available on their success-
option has its own characteristic spectral signals of exci- ful application to antibody-based assays. As a result of
tation (or absorption) and emission (or fluorescence). this history, instrumentation has become widely avail-
Many derivatives of these fluorescent probes possess able for measuring the fluorescence signals, includ-
reactive functionalities convenient for covalently link- ing standard lasers and filter sets which match the
ing them to antibodies and other targeting molecules. common excitation and emission wavelengths. Such
Each of the main fluorophore families contains at least fluorescently labeled antibodies are used in immuno-
a few main choices in coupling chemistry to direct the histochemical staining (IHC) (Osborn and Weber, 1982),
modification reaction to selected functional groups on in flow cytometry or cell sorting techniques (Ormerod,
the molecule to be labeled. The most popular deriva- 1990; Watson, 1991), for tracking and localization of
tives include amine-reactive, sulfhydryl-reactive, and antigens, and in various double-staining methods
carbonyl-reactive. Examples of some of the more use- (Kawamura, 1977). Extensive use of fluorescent anti-
ful varieties of fluorescent probes can be found in bodies in microscopy, pathology, and high content
Chapter 10. screening for drug discovery has made fluorescently

BIOCONJUGATE TECHNIQUES
2.  Preparation of Labeled Antibodies 893
labeled antibodies the most common antibody deriva- cancers. Originally, radiolabeling of antibodies merely
tives used in immunochemical detection techniques meant modifying tyrosine residues with 125I. Now, a
(Lichtman and Conchello, 2005; Luo et al., 2011). number of different radioactive elements are being
In choosing a fluorescent tag, the most important attached, both covalently and through specialized
factors to consider are good absorption (high extinc- chelating compounds to provide imaging capabilities
tion coefficient), stable excitation without photobleach- for the detection of primary tumors and metastases
ing, and efficient, high quantum yield of fluorescence. (Order, 1989).
Some fluorophores, such as fluorescein, exhibit rapid Radioiodination can be carried out using any one
photobleaching and undergo fluorescent quenching of a number of techniques. Most of the procedures uti-
due to dye–dye interactions at even moderate dye sub- lize 125I as the unstable isotope of choice for in vitro use
stitution levels, which lowers the quantum yield and due to its easy availability, comparably long 60-day
fluorescence signal. Up to 50% of the fluorescent inten- half-life, and relatively low-energy photon emissions.
sity observed on a fluorescein-stained slide can be lost Radioactive 125I usually is supplied as its sodium salt
within one month in storage. AMCA and some cyanine and must be oxidized to create an electrophilic spe-
dyes have much better stability, but all fluorophores cies capable of modifying molecules. Commonly used
lose some intensity upon exposure to light or upon stor- oxidizing agents include chloramine-T, Iodogen, and
age. Exceptions to this rule are the use of fluorescent Iodo-beads (Chapter 12, Section 2). When used in direct
nanoparticles, such as dye-doped silica (Chapter  14, labeling techniques with antibodies and other proteins,
Section 5) and quantum dots (Chapter 10, Section 10). these oxidants cause an iodination reaction to occur at
In some cases, the preparation of a fluorescently available tyrosine or histidine residues within the poly-
labeled antibody is not even necessary. Particularly, peptide chain. If either of these amino acids is impor-
if indirect methods are used to detect antibody bind- tant to antibody activity and cannot be labeled, then
ing to antigens, then preparing a fluorescently labeled certain crosslinking or modification reagents contain-
primary antibody is not needed. Instead, the selec- ing an activated aromatic ring may be used to radiola-
tion from a commercial source of a labeled secondary bel the molecule at other functional sites, particularly
antibody having specificity for the species and class at lysine amines. An example of this technique is to use
of primary antibody to be used is all that is required. the Bolton–Hunter reagent (Chapter  12, Section 2.5)
However, if the primary antibody needs to be labeled labeled with radioactive iodine to modify the primary
and it is not manufactured commercially, then a custom amines within the antibody. This reagent also can be
labeling procedure will have to be performed. used to add an iodinatable site to molecules containing
Generalized protocols for the attachment of these flu- no tyrosine residues (Figure 20.18).
orophores to protein molecules, including antibodies, Reagent options and protocols for the radioiodin-
can be found in Chapter  10 and Chapter  14, Section 5. ation of antibodies and other molecules may be found
The main consideration for the modification of immu- in Chapter 12.
noglobulins is to couple these probes at an optimal level Another method of adding a radioactive tag to anti-
to allow good detectability without high backgrounds. bodies is to use a chelating compound capable of com-
Too low a substitution level and the response of the flu- plexing metal isotopes. One of the most frequently used
orophore will yield low signal strength and poor sensi- chelating reagents is diethylenetriamine pentaacetic acid
tivity. Too high a substitution level and the fluorophore (DTPA) (Chapter 12, Section 1.1). The reagent contains 2
may self-quench through energy transfer, decrease the anhydride groups that can be used to modify primary
antibody’s ability to bind target molecules by blocking amines in proteins and other molecules. The reaction
the antigen binding sites, or cause nonspecific interac- process involves ring opening and the formation of an
tions resulting in high background or noise levels. In amide bond. Ring opening also creates up to four free
some cases, trial and error will be required to optimize carboxylate groups which, combined with the three
this process. nitrogen atoms in the chelator, are able to form strong
For other examples of antibody labeling protocols coordination complexes with metals such as indium-111
see Goding (1976) and Harlow and Lane (1988). (Figure 20.19). Monoclonal antibodies labeled with
bifunctional chelating agents containing radioisotopes
can be used in targeting tumor cells in vivo. The detec-
2.2.  Radiolabeled Antibodies
tion sensitivity of radiolabeled antibodies has led to
The attachment of a radioactive label onto an anti- effective diagnostic procedures to monitor primary and
body molecule provides a powerful means of detec- secondary cancer growths. In addition, the intensity of
tion in immunoassay procedures, tracking of analytes, radioactivity at the tumor site when labeled monoclo-
for in vivo diagnostic procedures, and, more recently, nals are used therapeutically can be great enough to
for the detection or therapy of numerous types of cause tumor cell death and remission.

BIOCONJUGATE TECHNIQUES
894 20.  Antibody Modification and Conjugation

O O
O O
N 125 I N
O I O
O Iodinating O
HO HO
Reagent
I Mono- or di-iodinated
Bolton–Hunter reagent
Bolton–Hunter reagent

N
HO
O
NHS

H2N

O
I Antibody containing
N available amine groups
H
HO

I
Iodinated antibody

FIGURE 20.18  Bolton–Hunter reagent may be used to add radioactive iodine labels to antibody molecules by modification of amines.

O O

O O
N H N
O N+ O
O +
O H2N

DTPA

Antibody containing
111 In available amine groups

O O
O O
NH O
In
O N N O

N
111 In-labeled antibody
O

FIGURE 20.19  The bifunctional chelating reagent DTPA may be used to modify amine groups on antibody molecules, forming amide bond
linkages. The isotope indium-111 may then be complexed to the chelator group to create a radiolabeled targeting reagent.

BIOCONJUGATE TECHNIQUES
2.  Preparation of Labeled Antibodies 895

2.3.  Biotinylated Antibodies Several assay designs that use the enhanced sensitiv-
ity afforded through biotinylated antibodies have been
Another popular tag for use with immunoglobulins developed. Most of these systems use conjugates of avi-
is biotin. Modification reagents that can covalently add din or streptavidin with enzymes (such as HRP or AP),
a functional biotin group to proteins, nucleic acids, and although other labels (such as fluorophores) can be used
other molecules now come in many shapes and reactivi- as well. In the simplest assay design, called the labeled
ties (Chapter 11, Section 6, and Chapter 18, Section 1.3). avidin–biotin (LAB) system, a biotinylated antibody
Depending on the reactive group present on the bioti- is allowed to incubate and bind with its target antigen.
nylation reagent, precise functional groups on antibod- Next, an avidin–enzyme conjugate is introduced and
ies may be modified to create an affinity tag capable allowed to interact with the available biotinylation sites
of binding avidin or streptavidin derivatives. Amines, on the bound antibody. Substrate development then pro-
carboxylates, sulfhydryls, and carbohydrate groups vides the detectability necessary to quantify the antigen.
can be specifically targeted for biotinylation through In a slightly more complex design, the bridged
the appropriate choice of reactive biotin compound and avidin–biotin system (BRAB) uses (strept)avidin’s mul-
modification procedure. Figure 20.20 shows the bioti- tiple biotin-binding sites to create an assay potentially
nylation of an antibody with NHS–LC-biotin, one of the of higher sensitivity than that of the LAB assay. Again,
most common biotinylation reagents. the biotinylated antibody is allowed to bind its tar-
Streptavidin conjugates are now used extensively to get, but next unmodified (strept)avidin is introduced
detect biotinylated antibodies after they have bound an to bind with the biotin binding sites on the antibody.
antigen. A streptavidin probe labeled with an enzyme, Finally, a biotinylated enzyme is added to provide a
fluorophore, high-contrast agent, or radiolabel can pro- detection vehicle. Since the bound (strept)avidin still
vide a very sensitive detection system to locate or assay has additional biotin binding sites available, the poten-
antigens in many different applications. In addition, the tial exists for more than one biotinylated enzyme to
potential for more than one labeled (strept)avidin con- interact with each bound (strept)avidin molecule. In
jugate to become attached to each biotinylated antibody some cases, sensitivity can be increased over that of the
through multiple biotin groups provides an increase LAB technique by using the bridging ability of avidin
in detectability over antibodies directly labeled with a or streptavidin (Chapter 11, Section 2).
detectable tag.

O
O
O
N
O HN HN
O H
N
+
O S NH2

NHS–LC-biotin
Antibody containing
available amine groups
O

N
HO
O
NHS

O
O
H
N HN HN
H
N

O S

Biotinylated antibody

FIGURE 20.20  Biotinylated antibodies can be formed by reacting NHS–LC-biotin with available amine groups to create amide bonds.

BIOCONJUGATE TECHNIQUES
896 20.  Antibody Modification and Conjugation

Antigen FIGURE 20.21  The basic design of an immunotoxin conju-


binding sites gate consists of an antibody-targeting component crosslinked
to a toxin molecule. The complexation typically includes a
Crosslinker
disulfide bond between the antibody portion and the cyto-
toxic component of the conjugate to allow release of the toxin
intracellularly. In this illustration, an intact A–B toxin protein
B chain
provides the requisite disulfide, but the linkage may also be
designed into the crosslinker itself.
S S

Disulfide-linked
subunits
A chain
Antibody component
directed against tumor antigen Protein toxin possessing
cytotoxic properties in vivo

A modification on this theme can be used to pro- The design of such cytotoxic antibodies is conceptu-
duce one of the most sensitive enzyme-linked assay ally simple: attach a toxic substance or a mediator of
systems known. The ABC system (for avidin–biotin toxicity to the appropriate monoclonal and you have a
complex) increases antigen detectability beyond that “magic bullet” that can find and eliminate the one-in-
possible with either the LAB or BRAB designs by form- a-billion cells that have the requisite marker (Figure
ing a polymer of biotinylated enzyme and (strept)avi- 20.21). The antibody provides the recognition and bind-
din before addition to an antigen-bound, biotinylated ing capacity, while the associated toxic component
antibody. When avidin and a biotinylated enzyme are effects cellular alterations leading to cell death (Pastan
mixed together in solution in the proper proportion, the et al., 2006).
multiple binding sites on (strept)avidin create a high- The approach to constructing antibody immunocon-
molecular-weight, multimeric complex. If the biotinyl- jugates for cancer has taken a number of forms (Vogel,
ated enzyme is not in large enough excess to block the 1987; Dosio et  al., 2011). One of the first designs used
binding sites on all the (strept)avidin molecules, then conjugates of monoclonal antibodies with toxins that
additional sites will still be available on this complex were able to block protein synthesis at the ribosome
to bind a biotinylated antibody bound to its comple- level inside the cell (Lord et  al., 1988). Other conjugate
mentary antigen. The large complex provides multiple forms used radioactive labels that killed cells by over-
enzyme molecules to dramatically enhance the sensi- exposure to radiation in proximity to where antibody
tivity of detecting antigen. Thus, the ABC procedure is docking occurred (Order, 1989). Drug conjugates were
currently among the highest-sensitivity methods avail- also constructed that combined the known benefits of
able for immunoassay work. chemotherapy with the targeting capability of monoclo-
More detailed discussions of (strept)avidin–biotin nals (Reisfeld et al., 1989; Willner et al., 1993; Bross et al.,
systems as well as the process of adding a biotin affinity 2001; Sievers and Senter, 2012). The expected result was
group to proteins, nucleic acids, and other biomolecules the effective concentration of the chemotherapeutic
can be found in Chapter 11. agent at the location of the tumor—hopefully eliminat-
ing or minimizing the side-effects of traditional chemo-
therapy. In addition, conjugates of monoclonals with
3.  IMMUNOTOXIN CONJUGATES certain biological modulators such as lymphokines or
growth factors were tried to affect malignant cell viabil-
Monoclonal antibodies directed against tumor anti- ity (Obrist et al., 1988; Choudhary et al., 2011).
gens may be used as targeting agents for conduct- Some immunoconjugates utilize intermediate car-
ing certain cytotoxic substances to malignant cells for rier systems consisting of polymeric molecules such as
selective killing. Numerous cell surface markers are polysaccharides, particularly dextran (Hurwitz et  al.,
known to proliferate in human solid tumors (Boyer 1978, 1980, 1983a,b, 1985; Manabe et  al., 1983; Sela and
et  al., 1988; Carter et  al., 2004; Aplin et  al., 2011; Kelly Hurwitz, 1987). The activated dextran is crosslinked to
et al., 2011; Yang et al., 2011). The ability to raise mono- both the monoclonal and the cytotoxic agent, providing
specific antibodies to these markers creates the capacity multivalent conjugation sites to create larger complexes
to discretely target the tumor, causing cell death while (Section 2.3, this chapter, and Chapter  18, Section 2).
leaving healthy cells alone (Salmon, 1989; McCarron Liposomes can be used in similar fashion by anchor-
et al., 2005). ing the antibody to its outer surface and charging the

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 897
vesicle with cytotoxic compounds (Gregoriadis, 1984; and assay applications continues to grow. The use
Ho et al., 1986; Matthay et al., 1986; Singhal and Gupta, of monoclonals as therapeutic agents in vivo, how-
1986; Kirpotin et al., 2012). See Chapter 21 for a survey ever, was complicated by the body’s natural immune
of liposome conjugation techniques. Also, dendrimer response designed to prevent the invasion of foreign
conjugates with antibody targeting molecules and cyto- substances.
toxic components have been used to create multivalent A major problem in the use of immunotoxins is that
immunotoxin conjugates (Chapter 8). injection of conjugates prepared from mouse (or ani-
Other systems have been designed to use a two- mal-based) monoclonals usually results in antibody
stage approach where an antibody is conjugated with production against the foreign protein. Antibodies of
an intermediate agent, which, when combined with animal origin quickly get removed from circulation
another factor, could elicit cytotoxicity. For instance, by the immune system, which prevents any therapeu-
enzyme conjugates with monoclonals have been tic benefit from the conjugate drug. Sometimes allergic
used that could transform an inactive pro-drug into reactions can further complicate the side-effects, mak-
a chemo-cytotoxic agent in vivo (Senter et  al., 1989). ing continued therapy infeasible. Most often, however,
Antibody-directed enzyme-prodrug therapy (ADEPT) induced host immunoglobulins will quickly bind the
has become an important option in the design of anti- immunoconjugate and remove it from the circulatory
body therapeutics (Tietze and Schmuck, 2011). See system. Instead of finding the targeted tumor cells,
Chapter  1, Section 3.5, for current applications of the immunotoxin conjugate ends up sequestered and
ADEPT conjugates. degraded in the liver or removed by the kidneys. Since
Monoclonals can also be tagged with a biotin label the common culprit in this scenario is often the mono-
and a secondary, avidin–toxin conjugate used to tar- clonal, the acronym HAMA, for human anti-mouse anti-
get the antibody once it bound the tumor cell. Two- body, is given to this response.
stage radiative treatment also has been tried through In an attempt to overcome the HAMA problem,
the use of boron neutron capture therapy (Holmberg “humanized” mouse monoclonals were designed
and Meurling, 1993). In this case, a carrier molecule is where large portions of the murine antibody are sub-
modified to contain 10B. After administration in vivo stituted for their human counterparts. For instance,
to target tumor cells, neutron bombardment yields replacing a mouse Fc portion with the corresponding
an unstable intermediate 11B which immediately human ones can significantly decrease the immune
undergoes a fission reaction to yield 7Li and 4He. The response against such conjugates. Replacing every-
induced radiation from this decomposition then kills thing but the hypervariable regions that code for anti-
the surrounding cells. gen binding has been accomplished, too. Unfortunately,
Tumor-targeting conjugates which use biospecific regardless of how much “humanization” is done, the
agents other than monoclonal antibodies have been remaining murine part still has the potential of caus-
developed as well. The targeting component in these ing immunological reactions. However, in the interven-
systems consists of any molecule that can function ing years since the beginning of monoclonal antibody
as a ligand having specific affinity for some recep- targeting in the late 1970s, the development of fully
tor molecule on the surface of the tumor cells. Such an human antibodies and antibody–drug conjugates
affinity molecule might be a hormone (typically called (ADCs) of many types has dramatically changed the
hormonotoxins) (Singh et al., 1989; Singh et al., 1993), a effectiveness of such agents in vivo (Jakobovits, 1995;
growth factor, an antigen specific for binding particular Kucherlapati et  al., 2012). The advent of recombinant
antibodies projecting from B cell surfaces, transferrin, antibody technology and transgenic mice producing
α2-macroglobulin, or anything else able to specifically antibodies of completely human origin has essentially
interact with the targeted tumor cells. See Chapter  1 overcome the problem of immune system response
for a review of current strategies in the use of antibody to the antibody component. As a result, there are now
conjugates and other conjugates in the treatment of hundreds of antibody therapeutic agents in develop-
cancer. ment and over a dozen antibody conjugates in late-
It became apparent very early in the development stage clinical trials with excellent prospects for approval
of such agents that their conception and design was (see Therapeutic Monoclonal Antibodies: World Market
much easier to imagine than to successfully imple- 2010–2025, Visiongain, 2010). ADCs employing fully
ment. Monoclonal antibodies used in vitro could eas- human antibodies attached to many different chemo-
ily detect antigen molecules in complex mixtures with therapeutic agents are now poised to revolutionize the
very little nonspecificity or cross-reactivity, even in cell- use of antibody conjugates for the treatment of disease
based assays. Very quickly following the invention of (Chari, 2008). See Chapter  1 for an extensive overview
hybridoma technology, monoclonals were employed in of antibody bioconjugates designed for therapeutic
numerous diagnostic assays and their use in detection purposes.

BIOCONJUGATE TECHNIQUES
898 20.  Antibody Modification and Conjugation

Modification of immunotoxin conjugates with syn- 2012). Toxins of many different types can be used to
thetic polymers also has been used to mask the complex create effective immunotoxin conjugates, including the
from the host immune response. Particularly, polyeth- proteins ricin from castor beans (Ricinus communis),
ylene glycol (PEG; see Chapter  18) has been found to abrin from Abrus precatorius, modeccin, gelonin from
be quite successful in reducing or eliminating immune Gelonium multiflorum seeds, diphtheria toxin produced
system reactions to modified proteins or antibodies by Corynebacterium diphtheriae, pokeweed antiviral pro-
(Roffler and Tseng, 1994). Modification of an antibody teins (PAPs; three types: PAP, PAP II, and PAP-S) from
conjugate with two to four PEG molecules increases the Phytolacca americana seeds, cobra venom factor (CVF),
serum half-life and improves tumor localization of the Pseudomonas exotoxin, restrictocin from Aspergillus
targeted reagent. restrictus, momordin from Momordica charantia seeds,
Other innovations in preparing targeted conjugates saporin from Saponaria officinalis seeds, as well as other
for cancer utilize recombinant DNA techniques to cre- ribosome-inactivating proteins (RIPs).
ate antibody molecules that are entirely of human ori- By far the most popular choices for the toxin com-
gin (Huse et  al., 1989; Orlandi et  al., 1989; Sastry et  al., ponent of protein-based immunotoxins are ricin,
1989; Ward et  al., 1989). A completely human antibody abrin, modeccin, and diphtheria toxin. The three plant
molecule eliminates the immunological problems asso- toxins have lectin binding activity toward terminal
ciated with mouse monoclonals. Intact antibodies, Fab β-galactosyl residues and they can be inhibited by the
fragments, small Fv fragments held together by syn- presence of simple sugars like galactose and lactose. The
thetically designed amino acid segments, and short toxin proteins bind to cell-surface polysaccharide recep-
peptides representing the antigen binding site have all tors with high affinity (Ka in the range of 107–108/M).
been developed by recombinant means. Although fre- Ricin, abrin, and modeccin consist of two subunits with
quently the word “antibody” is used to describe these remarkably similar structures and activities. The intact
engineered proteins, many of the molecules are far proteins have molecular weights of approximately
removed from the traditional picture of an antibody 63–65,000 with each subunit of about equal size. The
molecule. The terms “single-chain antibody” or “single- subunits are joined by disulfide linkages that are impor-
chain Fv protein” are commonly used and more closely tant reversible bonds in the mechanism of cytotoxicity.
describe these new targeting molecules. The A chain is called the effectomer and possesses ribo-
With the great diversity of targeted toxic agents somal-inactivating properties. The B chain contains the
being developed for cancer therapy, it would be dif- carbohydrate binding site and it is termed the haptomer.
ficult to characterize this section strictly as antibody While the intact toxin molecules have potent cytotoxic
conjugation. While many, if not most, studies utilize effects on cells, they exhibit no ribosomal inactivating
monoclonal antibodies of one form or another as the activity on ribosomes in a cell-free system. By contrast,
biospecific targeting component, the complete picture reduction of the toxins with a disulfide reducing agent
involves the crosslinking of a wide variety of molecules creates the opposite effects. Reduced, dissociated toxin
together to create the final conjugate. This section pres- subunits inhibit ribosomal activity in cell-free systems,
ents some of the most common methods of immuno- but they have no affect on intact cells.
toxin preparation. For the preparation of other unique The reason for these properties is due to the toxins’
targeted toxin conjugates, including. mode of action. Toxin molecules bind through sac-
ADCs, the methods found throughout this book for charide recognition sites on the B chain to particular
linking one particular functional group to another, can β-galactosyl-containing glycoprotein or glycolipid com-
be followed with an excellent probability for success. ponents on the surface of cell membranes. In animals
When preparing ADC conjugates, it is often impor- sensitive to these toxins, the necessary polysaccha-
tant to give great consideration to the cross-bridge and ride ligands are present in large quantities on virtu-
linkages formed between the antibody and the drug. ally all cell types (Cumber et  al., 1985). Upon binding
Bonds that are cleavable in vivo, such as disulfides and of the protein dimer to the cell, the A chain enters the
hydrazone linkages, have proven to be essential in the cell either by active transport into endocytic vesicles
successful design of effective ADCs (McCarron et  al., or through some mechanism of its own. Once inside
2005; Beck, 2010). the cell membrane, the A chain enters the cytoplasmic
space, binding to and enzymatically inactivating the
60S subunit of ribosomes (Olsnes and Pihl, 1976, 1982).
3.1.  Properties and Use of Immunotoxin
The result is cessation of protein synthesis and even-
Conjugates tual cell death. Because the A chain’s action is through
Conjugates of monoclonal antibodies and protein enzymatic means, as little as one active toxin molecule
toxins have been studied extensively for their useful- is enough to seriously disrupt protein synthesis opera-
ness in the treatment of cancer (Chandramohan et  al., tions and probably sufficient to kill a target cell (Eiklid

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 899

Disulfide
linkage

B chain
subunit
Sugar
binding site

B chain
subunit
A chain S S
subunit

Disulfide
linkage
Enzymatic
active site

A chain
subunit

FIGURE 20.22  Conceptualized construction of an A–B subunit protein toxin (left). The B chain contains a binding region for docking onto
cell surfaces, while the A chain contains a catalytic site that produces cytotoxic affects intracellularly. The two subunits are joined by a disulfide
bond that is reductively cleaved at the cellular level to allow the A subunit to cause cell death. A molecular model of the protein toxin ricin is on
the right.

et al., 1980). The turnover rate of one A chain molecule Due to the extraordinary toxicity of intact ribosome-
is about 1500 ribosomes inactivated per minute (Olsnes, inactivating toxins like ricin, abrin, and modeccin,
1978). purification and handling of these proteins must be
Diphtheria toxin also is a two-subunit protein, carried out with extreme care. Even dust from crude
but it is initially synthesized by certain strains of seed powders or lyophilized proteins should be con-
Corynebacterium diphtheriae as a single polypeptide sidered dangerous. During the height of the Cold War
chain of molecular weight 63,000. Proteolytic process- days, a Soviet KGB agent killed a man from the West
ing results in the formation of a “nicked toxin” which by injecting at most only milligram quantities of ricin
is enzymatically inactive, but consists of two subunits into his leg using a modified umbrella tip. There even
bonded together by an interchain disulfide. Upon have been instances of worker deaths at companies
reduction of the disulfide, the A chain (MW 24,000) is that routinely purify these proteins. For this reason, all
released and manifests enzymatic activity toward ribo- handling operations of intact toxin dimers and puri-
somal proteins (Collier and Cole, 1969; Sandvig and fied subunits should be performed in fume or laminar-
Olsnes, 1981). Its mode of action is different than that flow hoods. Avoid, also, the use of laboratory tools that
of the plant toxins. The A chain fragment of diphthe- could lead to puncture wounds causing contaminating
ria toxin catalyzes the ADP-ribosylation of eukaryotic toxin injection.
aminoacyl-transferase II (EF2) using NAD+ (Honjo Some potentially cytotoxic proteins contain only a
et  al., 1968; Gill et  al., 1969). The B chain, by contrast, single polypeptide chain, such as gelonin and PAPs.
possesses no enzymatic activity, but evidence points to Such toxins manifest similar enzymatic ribosome-inac-
the fact that a binding site on it recognizes certain cell tivating properties as the multi-subunit proteins like
surface receptors. As in the action of ricin, abrin, and ricin, but do not possess the cell-recognition capac-
modeccin, the B chain of diphtheria toxin is necessary ity that the B chain subunit contains. The result is the
for cytotoxicity (Colombatti et al., 1986). There also is a inability of these toxins to bind or affect intact cells.
role for the C-terminus cysteine residue of the B chain However, they do maintain the typical ribosome-inacti-
in cell penetration (Dell’Arciprete et al., 1988). vating properties in a cell-free system that the A chain
Figure 20.22 illustrates the basic structure of these of two-subunit toxins possess. If these proteins are con-
common two-subunit toxins, showing schematically jugated with a cell-targeting agent, such as the B chain
their major characteristics. The molecular model of ricin of ricin or a specific antibody that recognizes cell-sur-
is from Rutenber et al. (1991), RSCB structure No. 2aai. face epitopes, full cytotoxicity results.

BIOCONJUGATE TECHNIQUES
900 20.  Antibody Modification and Conjugation

Gelonin and PAPs are much more convenient to maintain the antigen binding character of the attached
work with than ricin and the other two-subunit tox- antibody and at the same time not block the ribosome-
ins. Most importantly, they are relatively nontoxic to inactivating activity of the toxin component.
cells unless conjugated with something that can facili- Studies have been carried out to investigate the
tate cell binding and internalization (Stirpe et  al., 1980; importance of using a cleavable linker between the anti-
Irvin, 1983). The pI of these toxins is in the basic range, body and the toxin. This configuration in the immuno-
and they each have a molecular weight of about 30,000 conjugate would mimic the natural state of two-subunit
(Barbieri and Stirpe, 1982). These single-subunit pro- toxins like ricin that are held together by disulfide
teins are very stable, especially to purification tech- bonds. There is evidence that disulfide reduction and
niques, but also to most modification and crosslinking cleavage of the A chain from the B chain is necessary
steps associated with preparing immunotoxins. Studies for cytotoxicity in native toxins (Olsnes, 1978). There is
have shown (Lambert et  al., 1985) that modification of similar evidence that the creation of cytotoxic immu-
gelonin or PAPs can be performed with 2-iminothiolane notoxins using only A chain subunits requires that the
(Traut’s reagent; Chapter  2, Section 4.1) to create sulf- conjugation be done with a monoclonal using a cross-
hydryl groups without loss of activity. Previous stud- linker that possesses a disulfide bond in its cross-bridge
ies, however, have determined that the use of SPDP to or creates a disulfide linkage upon coupling (Masuho
modify gelonin resulted in a 90% inactivation (Thorpe et  al., 1982). Using disulfide-cleavable crosslinkers in
et al., 1981). The difference in these results may be due the preparation of immunotoxins results in the anti-
to the retention of positive charge characteristics on the body taking on the role of the B chain in recognizing
modified amine when using Traut’s reagent, but neu- and binding to antigenic determinants on the surface
tralization of that charge when using SPDP. This is an of cells. After binding, some mechanism internalizes
example of how a slight difference in conjugation strat- the conjugate wherein the two components then are
egy can result in a dramatic difference in conjugate separated by disulfide reduction. The A chain subunit is
performance. then freed to enter the cytoplasmic space where enzy-
Immunotoxin conjugates consist of an antibody matic degradation of the ribosomal proteins occurs.
covalently crosslinked to a toxin molecule in a way that Other investigators, however, have demonstrated
maintains the unique properties of both proteins. The that conjugations of antibody with intact, two-sub-
antibody component consists of a monoclonal having unit toxins can be carried out using non-cleavable
specificity for an antigenic determinant on the surface crosslinkers such as NHS ester–maleimide heterobi-
of a particular cell type. Most often, the targeted cells functionals (Chapter  6, Section 1) (Myers et  al., 1989).
are tumors that express a unique cell surface marker Presumably, the toxin is still able to release the A chain
which can be recognized by the monoclonal. The role of after the antibody has bound to the cell, since the con-
the antibody, therefore, is to function as a passive taxi, jugation process does not permanently attach the two
carrying the toxin component to the targeted cells. Once toxin subunits together—only the toxin to the antibody.
at the tumor location, the toxin component effects its Thus, two main strategies can be used in making
intended ribosome-inhibiting action, ultimately causing immunotoxin conjugates (Figure 20.23). In the most
cell death and tumor destruction. often used method, the isolated A chain of two-subunit
Since immunotoxin conjugates are destined to be toxins (or the intact polypeptide of single-subunit tox-
used in vivo, their preparation involves more critical ins like gelonin) is conjugated to a monoclonal using a
consideration of crosslinking methods than most of the crosslinker that can introduce a disulfide bond. When
other conjugation protocols described in this book. The using only purified A chain, it is common (but not abso-
following sections discuss the issues associated with lutely required) to couple through the sulfhydryl that is
toxin conjugates and the main crosslinking methods for freed during A–B chain cleavage by disulfide reduction.
preparing them. The single-chain toxins like gelonin, however, have no
free sulfhydryls, so a thiolation agent such as 2-imino-
thiolane (Chapter  2, Section 4.1) may be used to create
3.2.  Preparation of Immunotoxin Conjugates them (Lambert et al., 1985).
It has become apparent that the method of crosslink- When using ricin A chains, it has been found that
ing can dramatically affect the activity of an immu- chemical deglycosylation of the subunit prevents its
notoxin in vivo. This is true not only with regard to nonspecific binding to receptors for mannose on cer-
possible direct blocking by the crosslinker of the enzy- tain cells of the reticuloendothelial system (Bitetta
matic active site which is responsible for inactivation of and Thorpe, 1985; Ghetie et  al., 1988, 1991; O’Hare
ribosomes, but the chemistry of conjugation is also an et  al., 1988). Thus, immunotoxin conjugates consist-
important factor in proper binding and entry of the con- ing of deglycosylated ricin A chain (dgA) have been
jugate into the cell. Preparation of the conjugate should shown to survive longer in vivo and are more efficient

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 901
Enzymatic active
site on A chain
A Chain
Intact IgG
Cleavable
disulfide
S S

Blocked B
chain binding site
B chain F(ab’)2
Conjugation using
intact A-B toxin

Fab
SH
A Chain SH
Half antibody
Fab’
HS
HS
S S

Conjugation using Engineered


Cleavable
A chain toxin subunit scFv fragments
disulfide
linkage

Conjugate Types Antibody Types

FIGURE 20.23  The strategies involved in creating an immunotoxin conjugate are numerous. Intact antibody molecules or enzymatic frag-
ments may be used as the targeting component. Even small recombinant Fv fragments that are genetically engineered to limit host immune
response can be employed. Conjugates can include two-subunit toxins or purified A chain components. If intact toxins are used, the B chain
binding site must be blocked to prevent nonspecific cell death. If A chain subunits are used, to induce cytotoxic effects in the conjugate the cross-
linking agent must generate a disulfide bond that can allow toxin release.

at reaching their intended target cells. In addition, if the one cell type (and thus prevent nonspecific cell death),
antibody component does not contain Fc region, but all cell binding capability within the toxin itself must be
consists of only F(ab′)2, Fab′, Fab, or smaller Fv frag- removed. Fortunately, a large proportion of the bind-
ments, then nonspecific binding of the immunotoxin in ing sites on the B chains are usually blocked during the
vivo will be reduced to a minimum. One study found conjugation process, and the galactose binding potential
that constructing immunotoxin conjugates with molar is significantly impaired. Further purification to remove
ratios of two dgA per antibody molecule resulted in a conjugates that have remaining galactose binding poten-
7-fold increase in cytotoxicity over a 1:1 conjugate ratio tial can be carried out on an acid-treated agarose chroma-
(Ghetie et al., 1993). tography column (which contains galactose residues) or
A chain immunotoxins, however, may not be quite on a column of asialofetuin bound to agarose (Cumber
as cytotoxic as conjugates formed from intact toxin mol- et  al., 1985). Conjugate fractions which do not bind to
ecules (Manske et  al., 1989). In an alternative approach both affinity gels contain no nonspecific binding poten-
to A chain use, the intact toxin of two-subunit proteins is tial toward non-targeted cells.
directly conjugated to a monoclonal without isolation of More elaborate methods of blocking or eliminating
the A chain. Conjugation of an antibody with intact A–B the B chain galactose binding site can also be done to
chain toxins can be performed without a cleavable linker, prevent nonspecific cytotoxicity. For instance, the cross-
as long as the A chain can still separate from the B chain linking agent may have a lactose molecule designed
once it is internalized. Therefore, it is important to avoid into it that can block B chain activity. The lactose por-
intramolecular crosslinking during the conjugation pro- tion possesses natural affinity for the B chain binding
cess which can prevent release of the A–B complex. In site, and thus it occupies that area while a nearby reac-
addition, since the B chain possesses a recognition site tive group covalently attaches the crosslinker to neigh-
for most cell surfaces, it still has the ability to nonselec- boring functional groups on the protein.
tively bind and kill non-tumor cells. To maintain anti- Moroney et  al. (1987) used this approach in creating
body specificity in intact toxin conjugates toward only a ricin conjugate. Lactose was modified at its reducing

BIOCONJUGATE TECHNIQUES
902 20.  Antibody Modification and Conjugation

FIGURE 20.24  In an elaborate strategy to block the B chain binding site in the construction of immunotoxins using intact A–B subunit tox-
ins, cystamine was first coupled to the reducing end of lactose by reductive amination. DTT was then used to reduce the cystamine disulfide
group, revealing the free thiol. A pyridyl disulfide-activated agarose gel then was used to couple the lactose derivative through its sulfhydryl.
Next, trichloro-s-triazine was reacted with the support to modify the secondary amine on the cystamine component, forming a reactive gel.
Finally, addition of an intact toxin to the affinity support caused binding with the lactose group at the B chain binding site. Since the B chain was
now in proximity to the chlorotriazine ring, covalent coupling occurred with available amines on the protein toxin, thus permanently blocking
the binding pocket. After removal of the modified toxin from the gel using a disulfide reducing agent, the free thiol of the cystamine group was
used to conjugate with an SMCC-activated immunoglobulin.

end with cystamine via reductive amination (Chapter 2, Regardless of their method of preparation, the
Section 5.3). The cystamine was reduced with dithioth- required and ideal characteristics of immunotoxin con-
reitol (DTT) and immobilized on an affinity gel which jugates can be summarized in the following points:
was activated with a pyridyl dithiol group (Chapter  3,
Section 2.6). The coupled lactose was then modified 1. The conjugation process must leave the antigen
with a chlorotriazine derivative through the second- binding sites on the antibody component free
ary amine that was created by the reductive amina- to interact with its intended target. Crosslinker
tion process. Next, the ricin molecule was immobilized modification or blockage of these binding sites by the
by the additional reactive group on the chlorotriazine attached toxin must be kept to a minimum.
ring. Since this reactive group was immediately adja- 2. The toxin component of the conjugate must be
cent to the lactose residue, the ricin bound the sugar able to elicit cytotoxicity by ribosomal damage as
at its B chain binding site and it was covalently cou- it could in its native state. This means that the cell
pled through a nearby amine. This process effectively penetration and enzymatic properties of the toxin
blocked and eliminated all nonspecific binding poten- remain unaltered, although an antibody molecule is
tial in the ricin dimer. After removal of the blocked ricin conjugated to it.
from the support by DTT, the free sulfhydryl group 3. The activity of toxin binding to cells through the
on the protein was conjugated to an SMCC-modified B chain must be eliminated in the conjugate to
antibody, forming the final conjugate (Figure 20.24). prevent nonspecific binding and nonselective cell
Although this process worked in preparing an effective death. This may be accomplished by using only the
immunotoxin conjugate, most conjugation schemes are A chain subunit or by blocking the B chain binding
less elaborate. site in the intact toxin conjugate.

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 903
4. Avoid covalently linking the A and B chains together SPDP
during the crosslinking process. This can be done by SPDP, N-succinimidyl 3-(2-pyridyldithio)propio-
using heterobifunctional crosslinkers that are more nate, is by far the most popular heterobifunctional
controllable in their reactivity than homobifunctional crosslinking agent used for immunotoxin conjugation
reagents. (Chapter 6, Section 1.1). The activated NHS ester end of
5. The crosslinking process must minimize SPDP reacts with amine groups in one of the two pro-
polymerization of either the antibody or the toxin. teins to form an amide linkage. The 2-pyridyldithiol
Low-molecular-weight, 1:1 or 1:2 conjugates of group at the other end reacts with sulfhydryl groups in
antibody-to-toxin are best. the other protein to form a disulfide linkage (Carlsson
6. The crosslinker used to form the bond between the et  al., 1978). The result is a crosslinked antibody–toxin
antibody and toxin must be able to survive in vivo conjugate containing cleavable disulfide bonds that
and not be cleaved by enzymatic or reductive means can emulate the activity of native two-subunit toxin
before reaching the targeted cells. molecules.
7. The conjugate must reach its intended target without LC–SPDP (Chapter  6, Section 1.1) is an analog of
being intercepted, bound and destroyed by the host SPDP containing a hexanoate spacer arm within its
immune system. internal cross-bridge. The increased length of the
8. Administration of the immunotoxin should result in extended crosslinker is important in some conjuga-
cell death and complete elimination of all target cells. tions to avoid steric problems associated with closely
The last two points are the most difficult to realize. linked macromolecules. However, for the preparation of
The following methods describing the conjugation strat- immunotoxins, no advantages were observed for LC–
egies used to prepare immunotoxins work well in cre- SPDP over SPDP (Singh et al., 1993).
ating complexes containing active antibody and toxin. SPDP is also useful in creating sulfhydryls in one of
The majority of research today is not so much concerned the two proteins being conjugated (Chapter  2, Section
with further optimization of the crosslinking process, 4.1). Once modified with SPDP, the protein can be
but primarily is directed at overcoming the host immune treated with DTT (or another disulfide reducing agent)
system and making the conjugates more effective in to release the pyridine-2-thione leaving group and form
accomplishing complete targeted tumor destruction. the free sulfhydryl. The terminal SH group then can be
used to conjugate with any crosslinking agents contain-
Preparation of Immunotoxin Conjugates via ing sulfhydryl-reactive groups, such as maleimide or
Disulfide Exchange Reactions iodoacetyl (for covalent conjugation) or 2-pyridyldithiol
Since the cytotoxic potential of most common tox- groups (for reversible conjugation).
ins relies on their subunit disulfide cleavability with In the preparation of immunotoxins, some proce-
subsequent release of associated A chains, most suc- dures call for the modification of the antibody with
cessful conjugation techniques for preparing immuno- SPDP to introduce reactive thiols (Cumber et al., 1985).
toxins involve the use of disulfide exchange reactions. The NHS ester end of the crosslinker is reacted at
Heterobifunctional crosslinking agents containing an slightly alkaline pH with the primary amines on the
amine-reactive group at one end and a disulfide bond antibody. After removal of excess reagent by gel filtra-
with a good leaving group on the other end are com- tion, the pyridyl disulfide groups are reduced by DTT.
mon choices for making these conjugates (Chapter  6, The reductant causes the removal of pyridine-2-thione
Section 1, and Chapter  18, Section 1.2). The leaving groups and the creation of sulfhydryl groups on the
group on the disulfide portion of the crosslinker per- immunoglobulin. The reason the antibody is thiolated
mits efficient disulfide interchange with a free sulfhy- in this manner and not the toxin is to avoid exposing
dryl on the antibody or toxin. The resultant covalent the intact toxin to reducing conditions that could disas-
bond thus is a cleavable disulfide which mimics the sociate the A and B subunits.
native cleavability inherent in the toxin dimer. To activate the toxin, SPDP again can be used to
modify the intact A–B component. After purification of
PYRIDYL DISULFIDE REAGENTS the modified toxin from excess crosslinker, the SPDP–
The most common reactive group for initiating disul- toxin is mixed with the thiolated antibody to effect the
fide interchange reactions is a pyridyl disulfide. Attack final conjugate (Figure 20.25).
of a nucleophilic thiolate anion dissociates the pyridine- This multi-step crosslinking method employing
2-thione leaving group and forms a new disulfide bond SPDP on both molecules has been used to prepare a
with the incoming sulfhydryl compound. Several cross- number of immunotoxin conjugates (Edwards et  al.,
linking reagents containing these groups are popular 1982; Thorpe et  al., 1982; Colombatti et  al., 1983; Wiels
choices for producing antibody–toxin conjugates. et  al., 1983; Vogel, 1987; Reiter and Fishelson, 1989).

BIOCONJUGATE TECHNIQUES
904 20.  Antibody Modification and Conjugation

N O
SPDP-activated
+ O O S N H antibody
NH 2 N
S
O

Antibody containing O S N
SPDP N OH
amine groups S

O
NHS DTT HN
S
A Chain

S S
SPDP-activated
A–B toxin HN
Thiolated antibody
O B chain H
N S N
S
O SH

S S

HN
H
N
S O
O S

Immunotoxin formation
via disulfide bond

FIGURE 20.25  SPDP can be used to modify both an antibody and a toxin molecule for conjugation purposes. In this case, the antibody is
thiolated to contain a sulfhydryl group by modification with SPDP followed by reduction with DTT. A toxin molecule is then activated with
SPDP and reacted with the thiolated antibody to form the final conjugate through a disulfide bond.

While this method has worked well for many differ- chains and therefore does not require crosslinker thiola-
ent toxins, its main potential disadvantage is exposure tion of one of the proteins.
of the antibody to reducing conditions that potentially Another way of utilizing SPDP is to again activate
could cleave the disulfide bonds holding its heavy and the antibody to create the pyridyl disulfide derivative,
light chains together. Alternative methods using SPDP but this time thiolate the toxin component using 2-imi-
in a non-reducing environment may result in better nothiolane (Chapter  2, Section 4.1). 2-Iminothiolane
conjugates. reacts with primary amines in a ring-opening reaction
For instance, if toxin A chain–antibody conjugates that creates a terminal sulfhydryl group without reduc-
are to be prepared, the antibody can be similarly acti- tion. Intact A–B toxins and toxins containing only one
vated with SPDP, but in this case not treated with subunit, like gelonin, PAPs, and Pseudomonas toxin
reductant. After removal of excess crosslinker, the acti- A, can be coupled to antibodies using this procedure
vated antibody can be directly mixed with isolated A (Lambert et al., 1985; Bjorn et al., 1986; Scott et al., 1987;
chain to create the conjugate (Figure 20.26). This proce- Lambert et  al., 1988; Ozawa et  al., 1989). Mixing the
dure makes use of the indigenous sulfhydryl residues SPDP-activated antibody with the thiolated toxin effec-
produced during reductive separation of the A and B tively forms the final conjugation (Figure 20.27).

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 905

N O
SPDP-activated
+ O O S N antibody
H
NH2 N
S
O

Antibody containing O S N
SPDP N OH
amine groups S
NHS
O

Remove B chain
by binding to
immobilized
lactose
B Chain
B Chain
+ HS
DTT SH
S S

A Chain
A Chain A Chain

Toxinmolecule Reduction creating


containing A–B subunits free sulfhydryl groups

HN
S

Pyridine-2-thione

H
N A Chain

O S
S

Immunotoxin formation
via disulfidelinkage

FIGURE 20.26  SPDP can be used to activate an antibody molecule through its available amine groups to form a sulfhydryl-reactive deriva-
tive. Toxin molecules containing disulfide-linked A–B chains may be reduced with DTT to isolate the A chain component containing a free thiol.
The SPDP-activated antibody is then mixed with the reduced A chain to effect the final conjugate by disulfide bond formation.

SPDP can also be used to conjugate other target- these conjugation techniques. The appropriate opti-
ing molecules to toxins, such as transferrin, epidermal mization for a particular toxin conjugate should
growth factor, α2-macroglobulin, and human chorionic be done.
gonadotropin (Fizgerald et al., 1980; Helenius et al., 1980; Caution! Toxins are highly toxic even in very low
Keen et  al., 1982; Oeltmann, 1985). To create these con- amounts. Handle all toxin molecules and their isolated sub-
jugates, one of the two components must be activated units with extreme care.
with SPDP to generate the sulfhydryl-reactive pyridyl
disulfide groups, while the other component must be Protocol for Thiolation of Antibody with
modified to contain the SH functionality. Mixing these SPDP and Conjugation to an SPDP-Activated
modified molecules together forms the toxin conjugate. Toxin
The following methods are generalized to pro- Caution: Toxin molecules are dangerously toxic even in
vide an overview of how SPDP can be used in small amounts. Use extreme care in handling.

BIOCONJUGATE TECHNIQUES
906 20.  Antibody Modification and Conjugation

+
NH2Cl
S S
S S
+ S +
Cl H2 N
HS
N
H
Toxin containing 2-Iminothiolane Thiolated toxin
A–B subunit structure

SPDP-activated
antibody

H S S
N +
Cl H2N
S
O S N
H

Immunotoxin conjugation
via disulfide linkage

FIGURE 20.27  An intact A–B subunit toxin molecule may be activated with 2-iminothiolane with good retention of cytotoxic activity. The
thiolated toxin may then be conjugated with SPDP-activated antibody to generate the immunotoxin conjugate through a disulfide bond.

Note: In this protocol, for every milligram of toxin 10,000. Retain this solution for the conjugation
employed, 2.5 mg of antibody are required to obtain the reaction.
correct molar ratios in the final conjugate.
(B) THIOLATION OF ANTIBODY WITH SPDP 
(A) ACTIVATION OF TOXIN WITH SPDP  1. Dissolve the antibody to be conjugated in 0.1-M
1. Dissolve the toxin to be conjugated in 0.1-M sodium sodium phosphate, 0.15-M NaCl, pH 7.5, at a
phosphate, 0.15-M NaCl, pH 7.5, at a concentration concentration 10 mg/ml. Note: Some protocols use
of 10 mg/ml. Some protocols use as an SPDP reaction the borate buffer system described in step A. Use
buffer, 50-mM sodium borate, 0.3-M NaCl, 0.5% 2.5 mg of antibody per mg of toxin to be conjugated.
n-butanol, pH 9.0. Both buffer systems work well for 2. Dissolve SPDP in DMF at a concentration of 3 mg/
the NHS ester modification reaction, although the pH ml. Add 24 μl of this solution to each milliliter of
9 buffer is at the higher end of effective derivatization the antibody solution with gentle mixing to effect
with active esters, since the hydrolysis rate is complete dissolution.
dramatically increased at this level of alkalinity. 3. React for 30 min at room temperature.
2. Dissolve SPDP (Thermo Fisher) in DMF at a 4. Remove excess crosslinker by gel filtration using
concentration of 3 mg/ml. Add 20 μl of this solution a desalting resin. Perform the chromatography
to each milliliter of the toxin solution. Gently mix to using as the buffer 0.1-M sodium phosphate, 0.15-M
effect dissolution. Retain the SPDP stock solution for NaCl, 10-mM EDTA, pH 7.5. The solution should
use in the antibody modification step, below. be degassed under vacuum and nitrogen bubbled
3. React for 30 min at room temperature. through it to remove oxygen. The presence of
4. Purify the SPDP-activated toxin from excess reagents EDTA stabilizes the free sulfhydryls formed in the
and reaction byproducts by gel filtration using a following steps against metal-catalyzed oxidation.
desalting resin. For the chromatography separation, 5. Concentrate the fractions containing protein from
use as a buffer 0.1-M sodium phosphate, 0.15-M the gel filtration step to 10 mg/ml using a centrifugal
NaCl, pH 7.5, containing 10-mM EDTA. concentrator (MW cutoff of 10,000).
5. Concentrate the toxin to 10 mg/ml using a centrifugal 6. To reduce the pyridyl dithiol groups and create
concentrator with a molecular weight cutoff of reactive sulfhydryls, dissolve DTT in water at a

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 907
concentration of 17.2 mg/ml and immediately add agents left over from the disassociation of the toxin
500 μl of this solution to each ml of concentrated subunits during the A subunit isolation. Reductants
antibody solution. Mix to dissolve and react for will compete for the SPDP activation sites on the
30 min at room temperature. antibody molecule.
7. Remove excess DTT by gel filtration using the same 7. React for 18 h at room temperature. Isolation of the
buffer as in step 4. Pool the fractions containing 1:1 or 1:2 antibody–toxin conjugate can be achieved
protein and concentrate to 10 mg/ml. through a gel filtration separation using a column of
Sephacryl S-200. Isolation of conjugates containing
molar ratios of 1:2 antibody:dgA have resulted in
(C) CONJUGATION OF SPDP-ACTIVATED TOXIN
greater cytotoxicity behavior in vivo (Ghetie et al., 1993).
WITH THIOLATED ANTIBODY 
1. Immediately mix the concentrated, thiolated
(E) CONJUGATION OF SPDP-ACTIVATED ANTIBODIES
antibody solution from part B with the SPDP-
WITH 2-IMINOTHIOLANE-MODIFIED TOXINS Caution:
activated toxin from part A.
Toxin molecules are dangerously toxic even in small amounts. Use
2. React for 18 h at room temperature to form the final
extreme care in handling.
conjugate. Isolation of the ideal 1:1 or 1:2 antibody–
A third option for immunotoxin preparation is to
toxin conjugate can be achieved through gel filtration
again activate the antibody with SPDP, while this time
separation using a column of Sephacryl S-300 or the
thiolating a single-chain toxin molecule to conjugate
equivalent.
with it. This method works especially well using 2-imi-
nothiolane (Chapter 2, Section 4.1) to create sulfhydryls
(D) ACTIVATION OF ANTIBODY WITH SPDP AND
on gelonin or PAPs. Gelonin is a single-polypeptide
CONJUGATION TO A TOXIN A CHAIN  Caution: Toxin mol-
toxin containing no free sulfhydryls. A number of
ecules are dangerously toxic even in small amounts. Use extreme
options are available for thiolation; however, the use
care in handling.
of SPDP to add sulfhydryl groups inactivates the toxin,
Since the A chain of toxin molecules contains a free
while 2-iminothiolane preserves its activity, perhaps by
sulfhydryl group, there is no need in this conjugation
maintaining the positive charge on the amines that are
strategy to thiolate one of the molecules. The follow-
being modified (Lambert et al., 1985).
ing protocol calls for 1.73 mg of antibody per milligram
of toxin A chain to produce a conjugate possessing the
(F) ACTIVATION OF ANTIBODY WITH SPDP 
correct molar ratio of components. Best results for creat-
1. Dissolve the antibody to be conjugated in 0.1-M
ing a highly cytotoxic immunotoxin will be obtained if
sodium phosphate, 0.15-M NaCl, pH 7.5, at a
deglycosylated ricin A chain is used.
concentration of 10 mg/ml.
1. Dissolve the antibody to be conjugated in 0.1-M 2. Dissolve SPDP (Thermo Fisher) in DMF at a
sodium phosphate, 0.15-M NaCl, pH 7.5, at a concentration of 3.0 mg/ml. Add 30 μl of this solution
concentration 10 mg/ml. to each milliliter of the antibody solution with gentle
2. Dissolve SPDP (Thermo Fisher) in DMF at a mixing to effect dissolution.
concentration of 3.0 mg/ml. Add 30 μl of this solution 3. React for 30 min at room temperature.
to each milliliter of the antibody solution with gentle 4. Remove excess crosslinker by gel filtration using a
mixing to effect complete dissolution. desalting resin. Perform the chromatography using
3. React for 30 min at room temperature. 0.1-M sodium phosphate, 0.15-M NaCl, 10-mM
4. Remove excess crosslinker by gel filtration using a EDTA, pH 7.5.
desalting resin. Perform the chromatography using 5. Concentrate the fractions containing SPDP-activated
0.1-M sodium phosphate, 0.15-M NaCl, 10-mM antibody from the gel filtration step to 10 mg/ml
EDTA, pH 7.5. using a centrifugal concentrator (MW cutoff of
5. Concentrate the fractions containing SPDP-activated 10,000).
antibody from the gel filtration step to 10 mg/ml using
a centrifugal concentrator (MW cutoff of 10,000). (G) THIOLATION OF GELONIN (OR OTHER SINGLE-
6. Mix the activated antibody solution with a solution POLYPEPTIDE TOXINS) WITH 2-IMINOTHIOLANE
of deglycosylated toxin A chain (dgA) dissolved in (TRAUT’S REAGENT) 
0.1-M sodium phosphate, 0.15-M NaCl, pH 7.5, 1. Dissolve gelonin at a concentration of 10 mg/ml
containing 10-mM EDTA. The ratio of mixing should in 50-mM triethanolamine hydrochloride, pH 8.0,
equal 1.73 mg of antibody per milligram of A chain containing 10-mM EDTA. The buffer should be
or 580 μl of A chain solution at 10 mg/ml per milliliter de-gassed under vacuum and bubbled with nitrogen
of activated antibody solution at 10 mg/ml. The to remove oxygen that may cause sulfhydryl
A chain solution must not contain any reducing oxidation after thiolation.

BIOCONJUGATE TECHNIQUES
908 20.  Antibody Modification and Conjugation

2. Dissolve 2-iminothiolane (Thermo Fisher) in SMPT


degassed, nitrogen-bubbled deionized water at a Succinimidyloxycarbonyl-α-methyl-α-(2-
concentration of 20 mg/ml (makes a 0.14-M stock pyridyldithio)toluene (SMPT) is a heterobifunctional
solution). The solution should be used immediately. crosslinking agent similar to SPDP that contains an
Add 70 μl of the 2-iminothiolane solution to each ml amine-reactive NHS ester on one end and a sulfhy-
of the gelonin solution (final concentration is about dryl-reactive pyridyl disulfide group on the other
10-mM). (Chapter 6, Section 1.2). Reaction with a sulfhydryl-con-
3. React for 1 h at 0°C (or on ice) under a nitrogen taining protein results in a cleavable disulfide linkage,
blanket. important for immunotoxin activity. SMPT is an analog
4. Purify the thiolated protein from unreacted Traut’s of SPDP that differs only in its cross-bridge, which con-
reagent by gel filtration on a desalting resin using tains an aromatic ring and a hindered disulfide group
0.1-M sodium phosphate, 0.15-M NaCl, pH 7.5, (Thorpe et al., 1987; Ghetie et al., 1990). The spacer arm
containing 10-mM EDTA. The presence of EDTA of SMPT is slightly longer than SPDP, but the presence
in this buffer helps to prevent oxidation of the of the benzene ring and α-methyl group adjacent to the
sulfhydryl groups and resultant disulfide formation. disulfide sterically hinders the structure sufficiently to
The degree of SH modification in the purified provide increased half-life of immunotoxin conjugates
protein may be determined using the Ellman’s assay in vivo.
(Chapter 2, Section 4.1). SMPT is often used in place of SPDP for the prepara-
5. Concentrate the thiolated toxin to 10 mg/ml tion of immunotoxin conjugates. The hindered disulfide
using centrifugal concentrators. Immediately use the of SMPT has distinct advantages in this regard. Thorpe
modified protein in the conjugation reaction et al. (1987) showed that SMPT conjugates had approxi-
to prevent inactivation of 2-iminothiolane- mately twice the half-life in vivo as SPDP conjugates.
modified molecules by recyclization (Chapter 2, Antibody–toxin conjugates prepared with SMPT pos-
Section 4.1). sess a half-life in vivo of up to 22 h, presumably due to
the decreased susceptibility of the hindered disulfide
(H) CONJUGATION OF SPDP-ACTIVATED ANTIBODY toward reductive cleavage.
WITH THIOLATED GELONIN  Ghetie et  al. (1991) developed a large-scale prepara-
1. Mix the SPDP-activated antibody with the thiolated tion procedure for antibody-deglycosylated ricin A
gelonin in equal mass quantities (or equal volumes if chain (dgA) conjugates utilizing this crosslinker. The
they are at the same concentration). This ratio results following procedure describes a generalized method for
in about a 5-fold molar excess of toxin over the using SMPT to prepare dgA–antibody conjugates. It is
amount of antibody. based on the Ghetie protocol, but using smaller quan-
2. React for 20 h at 4°C under a nitrogen blanket. tities of reagents. Figure 20.28 illustrates the reactions
3. To block unreacted sulfhydryl groups, add involved in using SMPT.
iodoacetamide to the solution to a final concentration
of 2-mM. Protocol
4. React for an additional 1 h at room temperature.
Caution: Toxin molecules are dangerously toxic even in
5. Remove unconjugated gelonin by passage of the
small amounts. Use extreme care in handling.
conjugate solution over a column of immobilized
The following method calls for mixing activated anti-
protein A (Thermo Fisher). Use 2 ml of the protein
body with ricin A chain at a ratio of 2 mg antibody per
A column for each 10 mg of conjugate to be
milligram of A chain. Adjustments to the amount of
purified. Equilibrate the column with 50-mM
antibody and A chain initially dissolved in the reaction
sodium phosphate, 0.15-M NaCl, pH 7.5. Apply
buffers should be done to anticipate this ratio.
the conjugate sample and allow it to enter the gel.
Continue to wash the column with equilibration 1. Dissolve the antibody to be conjugated in 0.1-M
buffer while taking 2-ml fractions until baseline is sodium phosphate, 0.15-M NaCl, pH 7.5, at a
reached (monitored at an absorbance of 280 nm). concentration of 10 mg/ml. If the antibody contains
Unconjugated gelonin will pass through the column oligomers (as evidenced by nondenaturing
unretarded. Elute bound conjugate with 0.1-M acetic electrophoresis or HPLC gel filtration analysis),
acid, 0.15-M NaCl. Immediately add 0.1 ml of 1-M then the monomeric IgG form should be isolated
potassium phosphate, pH 7.5 to each bound fraction by gel filtration using a column of Sephacryl
for neutralization. Alternatively, gel filtration may be S-200HR. If no oligomers are present, then omit the
used to isolate the conjugate from lower-molecular- chromatographic purification.
weight antibody and gelonin. A column of Sephacryl 2. Dissolve SMPT (Thermo Fisher) in DMF at a
S-200 works well for this purpose. concentration of 4.8 mg/ml. Add 27 μl of this solution

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 909

O
N
N O S S SMPT-activated
+ N
antibody
NH2
O O HN S S
CH3
O

Antibody containing O CH3


SMPT N OH
amine groups
O
NHS

Remove B chain
by binding to
immobilized
lactose
B chain
B Chain
SH
+ HS
DTT
S S

A chain A Chain
A Chain

Toxinmolecule Reduction creating


containing A–B subunits free sulfhydryl groups

HN
S

Pyridine-2-thione

HN S S A Chain

O CH3

Immunotoxin formation
via disulfide linkage

FIGURE 20.28  SMPT may be used to form immunotoxin conjugates by activation of the antibody component to form a thiol-reactive deriv-
ative. Reduction of an A–B toxin molecule with DTT can facilitate subsequent isolation of the A chain containing a free thiol. Mixing the A chain
containing a sulfhydryl group with the SMPT-activated antibody causes immunotoxin formation through disulfide bond linkage. The hindered
disulfide of an SMPT crosslink has been found to survive in vivo for longer periods than conjugates formed with SPDP.

to each ml of the antibody solution. Mix gently. The 10 mg/ml using centrifugal concentrators with a
final concentration of SMPT in the reaction mixture molecular weight cut-off of 10,000.
is 0.13 mg/ml, which translates into about a 4.8- 5. Dissolve deglycosylated ricin A chain (dgA) in 0.1-M
fold molar excess of crosslinker over the amount of sodium phosphate, 0.15-M NaCl, 10-mM EDTA,
antibody present. pH 7.5, at a concentration of 10 mg/ml. The buffer
3. React for 1 h at room temperature. should be degassed under vacuum and nitrogen
4. Remove unreacted SMPT and reaction byproducts bubbled through it to remove oxygen. Prepare half
by gel filtration on a desalting resin. Pool fractions the amount of A chain solution as the amount of
containing SMPT-activated antibody (the first peak antibody prepared in step 1. If the A chain
eluting from the column) and concentrate them to preparation is done in bulk quantities or if the

BIOCONJUGATE TECHNIQUES
910 20.  Antibody Modification and Conjugation

protein has been stored for lengthy periods, it may propionate (PDTP), the acid precursor of SPDP contain-
be necessary to reduce the sulfhydryls with DTT ing no NHS ester group on the carboxylate. Sulfhydryl
prior to proceeding with the crosslinking reaction. interchange reaction at the pyridyl dithiol end results in
If A chain sulfhydryl oxidation is suspected, add the formation of a disulfide linkage with SH containing
2.5 mg of DTT per milliliter of A chain solution. React molecules. The carboxylate end is not further deriva-
for 1 h at room temperature. Purify the reduced ricin tized to contain a reactive species, but may be coupled
A chain by gel filtration on a desalting resin using to amines by the carbodiimide reaction (Chapter  4,
the PBS–EDTA buffer. Apply no greater volume of Section 1). Reaction of PDTP with an antibody molecule
sample to the gel than is represented by 5% of the in the presence of 1-ethyl-3-(3-dimethylaminopropyl)
column volume to ensure good removal of excess carbodiimide (EDC) results in the formation of amide
DTT. Collect the protein and concentrate to 10 mg/ml linkages with the active pyridyl disulfide groups still
using centrifugal concentrators. available for coupling to sulfhydryl-containing toxins
6. Mix the reduced A chain solution with activated (Figure 20.29). Mixing the PDTP–antibody with puri-
antibody solution at a ratio of 2 mg of antibody fied ricin A chain results in disulfide crosslinks identical
per milligram of A chain. Sterile filter the solution to those obtained using SPDP as the crosslinker (Jansen
through a 0.22-μm membrane, and react at room et  al., 1980; Gros et  al., 1985). PDTP also has been used
temperature under nitrogen for 18 h. to activate transferrin to contain reactive pyridyl dithiol
7. To block excess pyridyl disulfide active sites on groups for conjugation to ricin A chain molecules (Raso
the antibody, add cysteine to a final concentration and Basala, 1984, 1985).
of 25 μg/ml. React for an additional 6 h at room Since activated molecules and crosslinks formed
temperature. between two species are identical to those formed using
8. To isolate the conjugate, apply the immunotoxin SPDP, it is of little advantage to use PDTP. Furthermore,
solution to a column of Sephacryl S-200HR. Collect an EDC-mediated reaction of the carboxylate end of the
the peaks with molecular weights between 150,000 crosslinker with amine groups on proteins can cause
and 210,000. Further purification to remove excess concomitant zero-length crosslinking and polymeriza-
unconjugated antibody can be done on a column of tion of protein molecules. For these reasons, SPDP is the
immobilized Cibacron Blue (available commercially better choice for preparing immunotoxin conjugates.
from Thermo Fisher or for column preparation, see
Hermanson et al., 1992). Equilibration of the column USE OF CYSTAMINE, ELLMAN’S REAGENT, OR
with 50-mM sodium borate, 1-mM EDTA, pH 9.0, S-SULFONATES
will cause binding of the conjugate, but not the free Other reagent systems can be used to form disul-
antibody. Elution of purified immunotoxin conjugate fide linkages between antibody and toxin molecules in
can be done with 50-mM sodium borate, 1-mM immunotoxin conjugates. Cystamine can be incorpo-
EDTA, 0.5-M NaCl, pH 9.0 (see Ghetie et al., 1991). rated into proteins by reaction of its terminal amines
with the carboxylates on the proteins via the car-
bodiimide reaction (Chapter  4, Section 1). The resul-
3-(2-PYRIDYLDITHIO)PROPIONATE tant modifications contain disulfide linkages that can
A lesser-used reagent to introduce sulfhydryl-reac- undergo disulfide interchange reactions with other
tive pyridyl disulfide groups is 3-(2-pyridyldithio) sulfhydryl-containing molecules (Chapter  2, Section

O S N EDC H
+ S
N
NH2
O S N
S

PDTP;
Antibody containing PDTP-activated antibody
3-(2-pyridyldithio)
amine groups (same as SPDP activation)
proprionate

FIGURE 20.29  PDTP may be used to modify antibody molecules using a carbodiimide reaction with EDC. The derivative is the same as
that obtained using SPDP activation and is highly reactive toward sulfhydryls.

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 911
4.1). For instance, a cystamine-modified targeting com- molecule of the highly chromogenic 5-sulfido-2-nitro-
ponent, such as an antibody, can be mixed with the benzoate, also called 5-thio-2-nitrobenzoic acid (TNB).
reduced A chain of a toxin molecule to cause conju- The intense yellow color produced by the TNB anion
gate formation through the creation of a disulfide bond can be measured by its absorbance at 412 nm. Thus, the
(Figure 20.30) (Oeltmann and Forbes, 1981). Epidermal efficiency of conjugation can be determined spectro-
growth factor was modified with cystamine and cou- photometrically using this procedure (Pirker et al., 1986;
pled with reduced diphtheria toxin using this approach Fitzgerald et al., 1988). A method for the large-scale con-
(Shimisu et al., 1980). jugation of Fab′ fragments containing an available sulf-
Similarly, Ellman’s reagent [5,5′-dithiobis(2-nitroben- hydryl group and deglycosylated ricin A chain (also
zoic acid)] can be used to activate one thiol-containing containing an SH group) were developed using Ellman’s
molecule by disulfide exchange and subsequently used reagent as the crosslinker (Ghetie et al., 1988).
to couple to a second sulfhydryl-containing molecule A final method of forming disulfide crosslinks
by the same mechanism (Chapter 2, Section 5.2) (Figure between toxins and targeting molecules is the use of
20.31). The disulfide of Elman’s reagent readily under- S-sulfonate formation using sodium sulfite (Na2SO3)
goes disulfide exchange with a free sulfhydryl to form in the presence of sodium tetrathionate (Na2S4O6).
a mixed disulfide with simultaneous release of one Tetrathionate reacts with sulfhydryls to form

S
+ H2N S
NH 2
O
COOH C S NH 2
N S
H
Antibody containing
Cystamine Cystamine-activated antibody
carboxylate groups

Remove B chain
by binding to
immobilized
lactose
B chain
B Chain
+ HS
DTT SH
S S

A Chain
A Chain
A Chain

Toxinmolecule Reduction creating


containing A–B subunits free sulfhydryl groups

NH2
HS

2-Mercaptoethylamine

O
C S
N S
H
Immunotoxin formation
via disulfide linkage

FIGURE 20.30  Cystamine may be used to make immunotoxin conjugates by a disulfide interchange reaction. Modification of antibody mol-
ecules using an EDC-mediated reaction creates a sulfhydryl-reactive derivative. A chain toxin subunits containing a free thiol can be coupled to
the cystamine-modified antibody to form disulfide crosslinks.

BIOCONJUGATE TECHNIQUES
912 20.  Antibody Modification and Conjugation

OH
O O
O
OH
O OH
+ O
N S O
SH + S S N+
SH O S N+
SH
O
O
Fab’ fragment containing
Ellman’s reagent OH TNB-activated Fab’ fragment
sulfhydryl groups
O
O
+
N SH
O
TNB

Remove B chain
by binding to
immobilized
lactose
B chain
B Chain
SH
+ HS
DTT
S S

A Chain A Chain
A Chain

Toxinmolecule Reduction creating


containing A–B subunits free sulfhydryl groups

OH
O
O
+
N SH
O
TNB

S S
SH

Immunotoxin formation
via disulfide linkage

FIGURE 20.31  Fab′ antibody fragments containing free thiols can be activated with Ellman’s reagent to form a sulfhydryl-reactive deriva-
tive. A chain toxin subunits that contain a free thiol group may be coupled to the activated Fab′ molecule to produce an immunotoxin complex.

sulfenylthiosulfate intermediates (Section 1.1). These


derivatives are reactive toward other thiols to create Preparation of Immunotoxin Conjugates via
disulfide linkages rapidly. Sulfite ions react with disul- Amine- and Sulfhydryl-Reactive Heterobifunctional
fides to form S-substituted thiosulfates, also known Crosslinkers
as S-sulfonates, and a thiol. The combination of these Other forms of heterobifunctional crosslinkers that
reagents results in the transformation of available thi- can be used for this purpose are the amine- and sulf-
ols and disulfides into reactive S-sulfonates that can be hydryl-reactive agents that produce a thioether bond
used to crosslink with sulfhydryl-containing molecules. with SH containing molecules (Chapter  6, Section 1).
S-sulfonate conjugation can be used to conjugate the The amine-reactive end of these crosslinkers is usually
A chain of toxin molecules with sulfhydryl-containing an NHS ester group that can form a stable amide bond
Fab′ fragments with good efficiency (Masuho et al., 1979). with amine-containing proteins. One of two main reac-
Although the use of these alternative disulfide-gen- tive groups are usually used on the sulfhydryl-reactive
erating agents has proven successful in some appli- end: an iodoacetyl group which couples to sulfhydryls
cations, pyridyl disulfide-containing crosslinkers, as with loss of HI or a maleimide group which undergoes
discussed previously, are more common for producing a double-bond addition reaction with SH groups (see
immunotoxin conjugates. Chapter 3, Sections 2.1 and 2.2).

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 913
Since this type of crosslinker forms non-cleavable potentially creating stable thioether bonds (Chapter  3,
thioether bonds between toxin molecules and the tar- Section 2.1).
geting component of the conjugates, they are not appro- Conjugations using SIAB to create immunotoxins
priate for use with A chain or single-chain toxins. This can be performed by first reacting the NHS ester end
is because the crosslinker will not allow the conjugated of the crosslinker with available amine groups on the
A chain to break free of the antibody by disulfide reduc- antibody and then coupling to a thiolated toxin dimer—
tion after docking at the cellular target. Since release of or by first reacting it with the toxin and coupling to a
the A chain is a prerequisite to ribosomal inactivation, thiolated antibody (Figure 20.32). Thiolation of the sec-
such conjugates will prove to be ineffective cytotoxic ondary component is usually done with SPDP or 2-imi-
agents. One report found a 1000-fold increase in cyto- nothiolane. Other crosslinkers containing an iodoacetyl
toxicity when an immunotoxin containing PAP or gelo- group can be used in a similar fashion.
nin was prepared using a cleavable disulfide linker as Conjugations with iodoacetyl crosslinkers have been
opposed to a non-cleavable thioether linkage (Lambert done using ricin and cobra venom factor (Thorpe et al.,
et al., 1985). 1984; Vogel, 1987; Myers et al., 1989). The following gen-
To make effective immunotoxin conjugates using eralized protocol for using SIAB is based on the method
the following crosslinkers, it is necessary to crosslink of Cumber et al. (1985).
intact A–B toxins to antibodies, not single-chain or
A chain toxins. Using intact two-subunit toxins allows Protocol
the A chain to break free of the complex and perform its Caution: Toxin molecules are dangerously toxic even in
cytotoxic duties upon entering the target cell. Two main small amounts. Use extreme care in handling.
criteria are especially important when using A–B toxin To prepare an antibody–ricin conjugate using this
conjugates: the B chain binding site must be blocked or protocol, 2.25 mg of antibody is needed for every milli-
inoperative in the final immunotoxin complex to pre- gram of toxin.
vent nonspecific cell death, and, second, the two sub-
units of the toxin must not be covalently crosslinked by (A) ACTIVATION OF TOXIN WITH SIAB 
the conjugation procedure, precluding them from being 1. Dissolve intact ricin in 0.1-M sodium phosphate,
separated in vivo. 0.15-M NaCl, pH 7.5, at a concentration of 10 mg/ml.
Fortunately, satisfying these criteria is not difficult. 2. Dissolve SIAB (Thermo Fisher) in DMSO at a
The heterobifunctional crosslinkers described in this concentration of 1.4 mg/ml. Prepare fresh and protect
section are sufficiently controllable as to prevent A–B from light to avoid breakdown of the active halogen
chain crosslinking. In addition, during the conjugation group.
process, the B chain binding site often becomes inacti- 3. Add 160 μl (225 μg) of the SIAB solution to each
vated or physically blocked by the attached antibody milliliter of the ricin solution.
molecule. Subsequent cleanup of the conjugate using 4. React for 30 min at room temperature in the dark.
affinity chromatography over a column containing an 5. Remove excess crosslinker from the activated ricin
immobilized sugar can completely eliminate any poten- by gel filtration using a desalting resin.
tial nonspecificity contributed by the B chain in the final 6. Concentrate the purified, SIAB-activated toxin
preparation. to 10 mg/ml using centrifugal concentrators with
It should be noted that the use of the following cross- a molecular weight cut-off of 10,000. Protect the
linkers to create other forms of toxic conjugates for activated toxin from light to prevent degradation of
cancer therapy is not restricted by the disulfide bridge the iodoacetyl reactive group.
requirement (Trail et  al., 1993; Willner et  al., 1993).
Drug–toxin conjugates, hormono-toxins, lymphokine– (B) THIOLATION OF SPECIFIC ANTIBODY MOLECULE
or growth factor–toxin conjugates all can be made using WITH SPDP 
nonreversible thioether linkages without difficulty. 1. Dissolve the antibody to be conjugated in 0.1-M
sodium phosphate, 0.15-M NaCl, pH 7.5, at a
SIAB concentration 10 mg/ml. Use 2.25 mg of antibody per
N-Succinimidyl(4-iodoacetyl)aminobenzoate (SIAB) milligram of toxin to be conjugated.
is a heterobifunctional crosslinker containing amine- 2. Dissolve SPDP (Thermo Fisher) in DMF at a
reactive and sulfhydryl-reactive ends (Chapter  6, concentration of 3 mg/ml. Add 24 μl of this solution
Section 1.5). The NHS ester on one end of the reagent to each milliliter of the antibody solution with gentle
can be used to couple with primary amine-containing mixing to effect complete dissolution.
molecules, forming stable amide linkages (Chapter  3, 3. React for 30 min at room temperature.
Section 1.4). The other end contains an iodoacetyl group 4. Remove excess crosslinker by gel filtration
that is specific for coupling to sulfhydryl residues, using a column of desalting resin. Perform the

BIOCONJUGATE TECHNIQUES
914 20.  Antibody Modification and Conjugation

B Chain

O I
H
N S S
O I
S S + N O H
N
O NH O
O
O
A Chain O
SIAB N OH
Toxin molecule
containing A & B subunits SIAB-activated
NHS O toxin

H
N

HS O

H
N

S S S O
H
N
NH O Immunotoxin created via
thioether bond formation
O

FIGURE 20.32  SIAB can be used to activate toxin molecules for coupling with sulfhydryl-containing antibodies. In this case, the antibody
molecule is thiolated using SATA and deprotected to reveal the free sulfhydryl. Reaction with the SIAB-activated toxin forms the final conjugate
by thioether bond formation.

chromatography using 0.1-M sodium phosphate, per milligram of toxin. Protect the solution from
0.15-M NaCl, 10-mM EDTA, pH 7.5. The buffer light.
should be degassed under vacuum and nitrogen 2. React for 18 h at room temperature in the dark.
bubbled through it to remove oxygen. The presence 3. To block unreacted sulfhydryl groups, add
of EDTA stabilizes the free sulfhydryls formed in the iodoacetamide to the solution to a final concentration
following steps against metal-catalyzed oxidation. of 2 mM. React for an additional 1 h at room
5. Concentrate the fractions containing protein from temperature.
the gel filtration step to 10 mg/ml using a centrifugal 4. Isolation of the ideal 1:1 or 1:2 antibody–toxin
concentrator (MW cut-off of 10,000). conjugate can be done by gel filtration separation
6. To reduce the pyridyl dithiol groups and create using a column of Sephacryl S-300.
reactive sulfhydryls, dissolve DTT (Thermo Fisher)
in water at a concentration of 17.2 mg/ml and SMCC
immediately add 500 μl of this solution to each Succinimidyl-4-(N-maleimidomethyl)cyclohexane-
milliliter of concentrated antibody solution. Mix to 1-carboxylate (SMCC) is a crosslinker with significant
dissolve and react for 30 min at room temperature. utility in protein conjugation (Chapter 6, Section 1.3). It
7. Remove excess DTT by gel filtration using the same is a popular choice among heterobifunctional reagents,
buffer as in step 4. Pool the fractions containing especially for the preparation of antibody–enzyme and
protein and concentrate to 10 mg/ml. hapten–carrier conjugates (Hashida and Ishikawa, 1985;
Dewey et  al., 1987). The NHS ester end of the reagent
(C) CONJUGATION OF SIAB-ACTIVATED TOXIN WITH can react with primary amine groups on proteins to
THIOLATED ANTIBODY  form stable amide bonds. The maleimide end of SMCC
1. Mix activated toxin from part A with thiolated is specific for coupling to sulfhydryls when the reaction
antibody from part B at a ratio of 2.25 mg of antibody pH is in the range of 6.5 to 7.5 (Smyth et al., 1964). The

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 915

O
SO 3 O

+ N O N O
NH2 H
O O N
O N
O
O
Antibody containing O
Sulfo-SMCC N OH
amine groups
Sulfo-SMCC-
O
activated antibody
NHS

Thiolated toxin
(via Traut’s reagent) S S
+
H2N
HS
N
H

S S
+
O H2N
H S
N N N
H
O
O

Immunotoxin conjugation via


thioether bond formation

FIGURE 20.33  Sulfo-SMCC may be used to activate antibody molecules for coupling to thiolated toxin components. An intact A–B toxin
molecule can be modified to contain sulfhydryls by treatment with 2-iminothiolane. Thiolation with this reagent retains the cytotoxic proper-
ties of the toxin while generating a sulfhydryl for conjugation. Reaction of the thiolated toxin with the maleimide-activated antibody creates the
immunotoxin through thioether bond formation.

nature of the reactive groups of SMCC allow for highly The following protocol is a suggested method for the
controlled crosslinking procedures to be performed conjugation of SMCC-activated antibodies with 2-imino-
wherein the resulting products can be closely limited thiolane-modified, intact toxin molecules (Figure 20.33).
to a 1:1 ratio in the final complex. Thus, low-molecu- It utilizes the water soluble analog of SMCC, sulfo-
lar-weight conjugates can be made which make ideal SMCC, which contains a negatively charged sulfonate
reagents for in vivo purposes. group on its NHS ring.
However, since SMCC forms nonreversible thioether
linkages with sulfhydryl groups, it can only be used Protocol
in the preparation of immunotoxins if intact A–B tox- Caution: Toxin molecules are dangerously toxic even in
ins are employed in the conjugate. In such conjugates, small amounts. Use extreme care in handling.
the A chain still has the potential for reductive release Note: This protocol requires mixing activated anti-
from the B chain subunit after cellular docking and body with thiolated toxin at a ratio of 2.25 mg of anti-
internalization. Immunotoxins prepared with A chain body per milligram of toxin. This ratio should be taken
or single-subunit toxins will not display cytotoxicity if into account before starting the reactions.
crosslinked with SMCC, since the crosslinker does not
create cleavable disulfide bonds upon conjugation. (A) ACTIVATION OF ANTIBODY WITH SULFO-SMCC 
SMCC has been used to prepare immunotoxins with 1. Dissolve 10 mg of specific antibody in 1 ml of 0.1-M
cobra venom factor (Vogel, 1987) and was compared to sodium phosphate, 0.15-M NaCl, pH 7.2.
other crosslinkers in the preparation of gelonin and PAP 2. Weigh out 2 mg of sulfo-SMCC (Thermo Fisher) and
conjugates (Lambert et al., 1985). add it to the above solution. Mix gently to dissolve.

BIOCONJUGATE TECHNIQUES
916 20.  Antibody Modification and Conjugation

To aid in measuring the exact quantity of crosslinker, milligram of toxin. Protect the solution from
a concentrated stock solution may be made in light.
water and an aliquot equal to 2 mg transferred to 2. React for 18 h at room temperature.
the reaction solution. If a stock solution is made, it 3. To block unreacted sulfhydryl groups, add
should be dissolved rapidly and used immediately iodoacetamide to the solution to a final concentration
to prevent extensive hydrolysis of the active ester. of 2 mM. React for an additional 1 h at room
Alternatively, a stock solution of sulfo-SMCC may temperature.
be prepared in DMSO and an aliquot added to the 4. Isolation of the ideal 1:1 antibody–toxin conjugate
aqueous reaction. can be done by gel filtration separation using a
3. React for 1 h at room temperature. column of Sephacryl S-300.
4. Immediately purify the maleimide-activated protein 5. Removal of nonspecific binding potential in the
by applying the reaction mixture to a desalting B chain must be done before using an A–B intact
column. Do not dialyze the solution, since the toxin conjugate in vivo. A large proportion of the
maleimide activity will be lost over the time course binding sites on the B chains are usually blocked
required to complete the operation. To obtain good during the above conjugation process, and the
separation between the protein peak (eluting first) galactose binding potential is significantly impaired.
and the peak representing excess reagent and Further purification to remove conjugates that have
reaction byproducts (eluting second), the applied galactose binding potential can be performed on an
sample size should be no more than 5 to 8% of the acid-treated agarose chromatography column (which
column bed volume. contains galactose residues) or on a column of
5. Collect the peak containing the activated antibody asialofetuin bound to agarose (Cumber et al., 1985).
(eluting first) and concentrate to 10 mg/ml using Conjugate fractions that do not bind to both affinity
centrifugal concentrators. Use immediately for gels contain no nonspecific binding potential toward
conjugating to a thiolated toxin. non-targeted cells.

(B) THIOLATION OF INTACT A–B TOXIN 


1. Dissolve the toxin (e.g., intact ricin) at a MBS
concentration of 10 mg/ml in 50-mM triethanolamine m-Maleimidobenzoyl-N-hydroxysuccinimide ester
hydrochloride, pH 8.0, containing 10-mM EDTA. (MBS) is a heterobifunctional crosslinking agent con-
The buffer should be degassed under vacuum and taining an NHS ester and a maleimide group. The NHS
bubbled with nitrogen to remove oxygen that may ester can react with primary amines in proteins and
cause sulfhydryl oxidation after thiolation. other molecules to form stable amide bonds, while the
2. Dissolve 2-iminothiolane in degassed, nitrogen- maleimide end reacts with sulfhydryl groups to create
bubbled deionized water at a concentration of 20 mg/ stable thioether linkages (Chapter  6, Section 1.4). The
ml (makes a 0.14-M stock solution). The solution reagent can be used in many different conjugation pro-
should be used immediately. Add 70 μl of the tocols to crosslink amine-containing proteins with sulf-
2-iminothiolane solution to each milliliter of the toxin hydryl-containing proteins. Since the thioether bond
solution (final concentration is about 10-mM). formed at the maleimide end is nonreversible, MBS
3. React for 1 h at 0°C (on ice) under a nitrogen blanket. can be used for immunotoxin preparation only if the
4. Purify the thiolated toxin from unreacted Traut’s conjugate involves crosslinking intact A–B toxins with
reagent by gel filtration using 0.1-M sodium antibody molecules. Using intact toxins (as opposed to
phosphate, 0.15-M NaCl, pH 7.5, containing 10-mM single-chain or A chain isolates), the A chain still is able
EDTA. The presence of EDTA in this buffer helps to release from the complex after cellular docking and
to prevent oxidation of the sulfhydryl groups inactivate ribosomal activity (Youle and Nevelle, 1980;
with resultant disulfide formation. The degree of Dell’Arciprete et al., 1988; Myers et al., 1989).
SH modification in the purified protein may be MBS contains a benzoic acid derivative as its
determined using the Ellman’s assay (Chapter 2, cross-bridge. In many applications involving NHS–
Section 4.1). maleimide crosslinkers, non-aromatic cross-bridges are
5. Concentrate the thiolated toxin to 10 mg/ml using considered superior to aromatic ones. This is reflected
centrifugal concentrators. in the stability of the maleimide group to hydro-
lysis prior to conjugating with a sulfhydryl group.
(C) CONJUGATION OF SMCC-ACTIVATED ANTIBODY For immunotoxin preparation, however, aromatic
WITH THIOLATED TOXIN  maleimides resulted in better conjugate yield and
1. Mix the thiolated toxin with SMCC-activated more potent cytotoxic effects when compared to ali-
antibody at a ratio of 2.25 mg of antibody per phatic ones (Myers et al., 1989). MBS, therefore, may be

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 917

B Chain O O
N S S
O O
S S + N O
N
NH O
O O O
A chain O
MBS N OH
Toxin molecule
containing A & B subunits MBS-activated
NHS O toxin

HS
HS

Half antibody

S S O S
N HS

NH O

Immunotoxin created via


thioether bond formation

FIGURE 20.34  Activation of an intact A–B toxin molecule with MBS with subsequent conjugation with a reduced antibody fragment to pro-
duce an immunotoxin.

a crosslinker of choice when making conjugates with 4. React for 30 min at room temperature.
intact toxin molecules. 5. Immediately purify the MBS-activated toxin by gel
The following protocol is adapted from Myers et  al. filtration using a column of desalting resin. Apply
(1989). It involves activation of ricin with MBS and con- no more sample than represents 5 to 8% of the gel
jugation with a partially reduced antibody (Figure 20.34). volume. Isolate the protein peak by its absorbance at
280 nm and concentrate to 10 mg/ml using centrifugal
Protocol concentrators with a molecular weight cut-off of 10,000.
Caution: Ricin molecules are dangerously toxic even in
small amounts. Use extreme care in handling. (B) PARTIAL REDUCTION OF ANTIBODY WITH DTT 
This method uses a molar ratio of 15:1 for 1. Dissolve the antibody in 0.1-M sodium phosphate,
ricin:antibody. This requires 6.24 mg of ricin per milligram 0.15-M NaCl, 10-mM EDTA, pH 7.5, at a
of antibody. This ratio should be considered when deter- concentration of 10 mg/ml.
mining how much starting materials to use for each step. 2. Add DTT to a final concentration of 50 mM. Note:
Lower concentrations of DTT may be used to avoid
(A) ACTIVATION OF RICIN WITH MBS  extensive reduction that may dissociate the antibody
1. Dissolve ricin at a concentration of 10 mg/ml in 0.1-M subunits. Concentrations in the range of 3- to 5-mM
sodium phosphate, 0.15-M NaCl, pH 7.5. DTT have been shown to be appropriate for the
2. Dissolve MBS (Thermo Fisher) in DMF at a reduction of just a few disulfides within the antibody
concentration of 2 mg/ml. structure (Sun et al., 2005). Some optimization of the
3. Add 76 μl of the MBS solution to each ml of the reductant concentration may have to be performed
ricin solution. This represents a 3:1 molar ratio of to obtain the best results for a particular antibody
crosslinker to protein. molecule.

BIOCONJUGATE TECHNIQUES
918 20.  Antibody Modification and Conjugation

3. Reduce for 30 min at room temperature. base formation with subsequent reduction using
4. Purify the reduced antibody using gel filtration on a sodium cyanoborohydride to form stable secondary
column of Sephadex G-25. Concentrate the protein to amine linkages (Chapter 3, Section 5.3). Carbohydrates,
10 mg/ml using centrifugal concentrators. glycoproteins, and other polysaccharide-containing
molecules can be oxidized to contain aldehyde residues
(C) CONJUGATION OF MBS-ACTIVATED RICIN WITH by sodium periodate or specific oxidases (Chapter  2,
PARTIALLY REDUCED ANTIBODY  Section 4.4). Some antibodies and toxin molecules are
1. Mix the MBS-activated ricin with the partially glycoproteins and contain sufficient carbohydrate to be
reduced antibody in a molar ratio of 15:1 (or 6.24 mg utilized for reductive amination crosslinking.
activated ricin per mg of reduced antibody). This A second method of immunotoxin preparation by
represents a volume ratio (at 10 mg/ml for both reductive amination involves the use of a polysaccha-
proteins) of 1 ml ricin solution mixed with 160 μl ride spacer. Soluble dextran may be oxidized with peri-
antibody solution. odate to form a multifunctional crosslinking polymer.
2. React for 18 h at room temperature. Reaction with antibodies and cytotoxic molecules in the
3. Purification of the immunotoxin conjugate from presence of a reducing agent forms multivalent immu-
unconjugated ricin can be performed using a column notoxin conjugates. The following sections discuss
of TSK3000 SW (Toya Soda, Japan) according to the these options.
method of Myers et al. (1989).
4. Removal of nonspecific binding potential in the B Periodate Oxidation of Glycoproteins Followed by
chain must be done before using an A–B intact toxin Reductive Conjugation
conjugate in vivo. See step 5 of the MBS conjugation Antibody molecules usually contain carbohydrate
protocol discussed previous to this section. in their Fc regions. Similarly, many toxins, such as
ricin and abrin, are glycoproteins that contain abun-
SMPB dant polysaccharide. These carbohydrate residues can
Succinimidyl-4-(p-maleimidophenyl)butyrate be oxidized with 10-mM sodium periodate to form
(SMPB) is a heterobifunctional analog of MBS contain- reactive aldehyde groups capable of being conjugated
ing an extended cross-bridge (Chapter  6, Section 1.6). with primary amines (Chapter  2, Section 4.4). Mixing
The crosslinker has an amine-reactive NHS ester on one an aldehyde-containing glycoprotein with another
end and a sulfhydryl-reactive maleimide group on the amine-containing molecule in the presence of sodium
other. Conjugates formed using SMPB thus are linked borohydride or sodium cyanoborohydride reduces the
by stable amide and thioether bonds. intermediate Schiff bases that are formed to stable sec-
As in the case of MBS, discussed previously, SMPB ondary amine bonds. Since functional groups on the
was found to be more effective than aliphatic cross- antibody and the toxin components are the only ones
linkers in producing immunotoxin conjugates with ricin necessary for this type of conjugation strategy, it is
that have high yields of cytotoxicity (Myers et al., 1989). often referred to as a zero-length crosslinking procedure
This was attributed to the reagent’s aromatic ring struc- (Chapter  4). In other words, no additional crosslinking
ture. A comparison with SPDP produced immunotoxin reagents are introduced into the site of the crosslink.
conjugates concluded that SMPB formed more stable This method of conjugation is used with great success
complexes that survive in serum for longer periods in the formation of antibody–enzyme conjugates, espe-
(Martin and Papahadjopoulos, 1982). cially using the glycosylated enzyme, horseradish per-
The method for the preparation of immunotoxins oxidase (HRP) (Chapter 22, Section 1.1).
with SMPB is identical to that used for MBS (above). The disadvantage of this type of conjugation
Since the thioether bonds formed with sulfhydryl-con- approach for producing immunotoxins is that many
taining molecules are non-cleavable, A chain isolated of the monoclonal antibodies or antibody fragments
fractions or single-chain toxin molecules cannot be con- used for immunotoxin conjugation are devoid of car-
jugated with antibodies with retention of cytotoxicity. bohydrate. Especially when using small Fv fragments
Only intact A–B toxin molecules may be used with this or single-chain antibodies produced by recombinant
crosslinker, since the A chain is still capable of being techniques, there are typically no polysaccharide por-
reductively released from the complex. tions attached to them. In this case, creation of alde-
hydes on the targeting component is not possible. In
3.3.  Preparation of Immunotoxin Conjugates via addition, not all toxin molecules contain carbohydrate.
Ricin, abrin, and cobra venom factor are glycoproteins
Reductive Amination
and can be oxidized and coupled to antibodies without
Conjugations involving aldehyde groups and amine- difficulty (Olsnes and Pihl, 1982a,b; Vogel and Muller-
containing molecules can be performed through Schiff Eberhard, 1984). However, it is not well known whether

BIOCONJUGATE TECHNIQUES
3. Immunotoxin Conjugates 919

O O
HO O HO O

O O O O
O O O O
O
HO O O HO O
O
HO O O HO O O
O O O
O O
O O O

HO O HO O
Periodate-oxidized O
O
dextran polymer

B Chain

S S

H2N
A Chain

Toxin molecule Antibody containing


containing A & B subunits primary amine groups

NaCNBH3

S
S

O NH
HO O HO O
NH O O O
O O O O
O
HO O O HO O
O
HO O O HO O O
O O O
O O
O O O

HO O HO O
NH HN
Multivalent complex
S
S

FIGURE 20.35  A periodate-oxidized dextran polymer may be reacted with both an antibody and an intact toxin component using reductive
amination to form a multivalent immunotoxin complex.

immunotoxin conjugates formed by this procedure repeating unit is an isomaltose group. Most prepa-
retain their ability to inhibit ribosomal activity. rations of dextran contain some branching, mainly
Suggested procedures for using reductive amination incorporating 1,2, 1,3, and 1,4 linkages. The degree of
techniques may be found in Chapter 2, Section 4.4 and branching is characteristic of its source—the strain and
Chapter 4, Section 4. species of yeast or bacteria from which the dextran orig-
inated. The terminating monosaccharide in a dextran
Periodate Oxidized Dextran as Crosslinking Agent polymer is often a fructose group. Dextran polymers of
Dextran polymers consist of glucose residues bound molecular weight 10,000 to 40,000 provide long, hydro-
together predominantly in α-1,6 linkages. The main philic arms that can accommodate multiple attachment

BIOCONJUGATE TECHNIQUES
920 20.  Antibody Modification and Conjugation

points for macromolecules along their length. Soluble the carrier, the antibody is added along with a reducing
dextrans can be oxidized in aqueous solution to create agent to create the final secondary amine linkages (Sela
numerous aldehyde residues suitable for use in reduc- and Hurwitz, 1987). The dextran backbone provides
tive amination techniques (Hurwitz et  al., 1978, 1985; the ability to couple many more drug molecules associ-
Manabe et al., 1983; Sela and Hurwitz, 1987). Periodate ated with each antibody than could be accomplished by
oxidation results in the cleavage of the carbon–carbon direct conjugation to the antibody itself.
bonds between the Nos. 2 and 3 carbons within each Although dextran can be a versatile crosslinking
monosaccharide unit of the chain, transforming the agent for the preparation of many forms of macromo-
associated hydroxyl groups into aldehydes (Chapter  2, lecular conjugates, immunotoxin conjugation may be
Section 4.4). impeded by the non-reversibility of the multiple amide
Periodate oxidized dextran can be used as a protein bond linkages formed during reductive amination.
modification or crosslinking agent (Chapter 18, Section Certainly, only intact A–B toxins have a chance of suc-
2.2). Conjugation of antibody molecules to toxins can be ceeding with this method, since A chain or single-sub-
performed with dextran to produce immunotoxins suit- unit toxins would not be capable of release from the
able for in vivo administration. Mixing of the antibody complex after cellular docking. Even intact two-subunit
and toxin together with the oxidized dextran under toxins, however, may not be capable of releasing an A
alkaline conditions will result in the formation of Schiff chain unit, due to the multivalent nature of the oxidized
base interactions with the amines on both proteins. dextran linker. For this reason, activated dextran may
Reduction of these Schiff base linkages with sodium be more useful for constructing antibody conjugates
borohydride or sodium cyanoborohydride results in consisting of cytotoxic organic compounds, not protein
stable secondary amine bonds, covalently attaching toxins—for example, chemotherapeutic drugs, hor-
multiple antibody and toxin molecules along the length mones, or radioactive complexes.
of the polysaccharide chain (Figure 20.35). Methods for using oxidized dextran and reduc-
Chemoimmunoconjugates or antibody–drug conju- tive amination techniques can be found in Chapter  3,
gates (ADCs) consisting of anticancer agents attached to Section 4.4; Chapter  3; Section 5; and especially
antibody targeting molecules also can be formed using Chapter  18, Section 2.2. Reference should also be
oxidized dextran carriers. Cancer therapeutic agents made to the use of dendrimers as carriers for making
such as adriamycin, bleomycin, and daunomycin can cytotoxic targeting complexes (Chapter  8) as well as
be coupled to the oxidized dextran through their amine Chapter  1, Section 3.5, which provides a broad over-
groups (also see Chapter  1, Section 3.5). After forma- view of the types and applications of therapeutic bio-
tion of Schiff base linkages between these drugs and conjugates used today.

BIOCONJUGATE TECHNIQUES

You might also like