1267 FTP PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Eur. J. Immunol. 2006.

36: 1267–1274 Molecular immunology 1267

Induction of IgG antibodies against the GD2 carbohydrate


tumor antigen by vaccination with peptide mimotopes
Angelika B. Riemer1,2, Elisabeth Frster-Waldl3, Kira H. Brmswig1,3,
Arnold Pollak3, Christoph C. Zielinski4, Hubert Pehamberger2,
Holger N. Lode5, Otto Scheiner1 and Erika Jensen-Jarolim1
1
Department of Pathophysiology, Medical University of Vienna, Vienna, Austria
2
Department of Dermatology, Medical University of Vienna, Vienna, Austria
3
Department of Pediatrics and Juvenile Medicine, Medical University of Vienna,
Vienna, Austria
4
Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
5
Charit-Universittsmedizin Berlin, Pediatrics, Campus Virchow, Berlin, Germany

The disialoganglioside GD2, a carbohydrate antigen, is expressed on all tumors of Received 19/7/05
Revised 25/1/06
neuroectodermal origin, including melanoma, neuroblastoma, sarcoma and small cell
Accepted 27/2/06
lung cancer. Due to its specific expression on tumor surfaces, GD2 is an attractive target
for immunotherapies. The mouse/human chimeric anti-GD2 mAb ch14.18 is already [DOI 10.1002/eji.200535279]
applied in melanoma and neuroblastoma trials as a passive immunotherapy. To
establish an active immunotherapy alternative, we aimed to replace the poorly
immunogenic ganglioside with immunogenic peptides. Previously, we used the ch14.18
antibody to select GD2 peptide mimics from a phage display library. In the present
study, two mimics of the ch14.18 epitope were coupled to keyhole limpet hemocyanin
and used for immunizing BALB/c mice. Induction of a specific humoral immune
response towards the original antigen GD2, both purified and expressed on
neuroblastoma and melanoma cells, could be demonstrated in ELISA, Western blot, Key words:
and immunofluorohistochemistry. As the elicited antibodies were of the IgG isotype, the Cancer vaccine
mimotope conjugates were capable of recruiting T cell help and inducing memory  ch14.18
phenomena. In conclusion, we show that an epitope of the carbohydrate antigen GD2  Disialoganglioside
can successfully be translated into immunogenic peptide mimotopes. Our immunization GD2  Epitope mimic
experiments indicate that GD2 mimotopes are suitable for active immunotherapy of  Tumor-associated
GD2-expressing tumors. carbohydrate antigen

Introduction metastasis and progression rates [1]. Well-documented


examples of such TACA are GM2, GD2, and GD3
Tumors expressing high levels of certain tumor-asso- gangliosides in neuroectodermal tumors [2, 3]. Their
ciated carbohydrate antigens (TACA) exhibit greater surface localization makes these gangliosides effective
targets for active and passive antibody therapies. The
disialoganglioside GD2 is considered especially inter-
Correspondence: Erika Jensen-Jarolim, Department of Patho-
physiology, Center of Physiology and Pathophysiology, Medical
esting, as it is expressed on all tumors of neuroecto-
University of Vienna, Whringer Grtel 18–20, 1090 Vienna, dermal origin, including melanoma, neuroblastoma,
Austria sarcoma and small cell lung cancer [4, 5], but is absent
Fax: +43-1-40400-5130 in normal tissues and only minimally expressed in brain
e-mail: erika.jensen-jarolim@meduniwien.ac.at [3] and peripheral nerves [5, 6]. Due to its localization
Abbreviations: DGG: synthetic C-DGGWLSKGSW-C 
in focal adhesion plaques at the interface of tumor cells
GRL: synthetic C-GRLKMVPDLE-C  HAMA: human anti-mouse
antibodies  KLH: keyhole limpet hemocyanin  TACA: tumor- and their substratum, it is thought to play a role in cell
associated carbohydrate antigen attachment and metastasis [7].

f 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji.de


1268 Angelika B. Riemer et al. Eur. J. Immunol. 2006. 36: 1267–1274

Several mAb against GD2 have been raised, with peptides were again checked for specific recognition
14.18 [8] and 3F8 [9] being the most prominent by the selecting antibody. Indeed, synthetic C-
examples. mAb 3F8 has been given to neuroblastoma GRLKMVPDLE-C and C-DGGWLSKGSW-C (subse-
patients in its murine form [10], and from these studies quently referred to as “GRL” and “DGG”), circularized
it was seen that the antigen GD2 is rarely lost following by disulfide bridging between the flanking cysteines,
mAb treatment [11]. mAb 14.18 has been further were recognized by ch14.18, but not by the isotype
developed into a human/mouse chimeric derivative, control antibody (Fig. 1, left panel). To ensure
ch14.18. Both 14.18 and ch14.18 trigger tumor cell lysis immunogenicity and availability of sufficient T helper
via antibody-dependent cellular cytotoxicity and com- epitopes, the mimotopes were coupled to the immuno-
plement-dependent cytotoxicity [12, 13]. In preclinical genic carrier, keyhole limpet hemocyanin (KLH).
models, the 14.18 antibodies have been shown to Specific mimicry of the ch14.18 epitope through
prevent the outgrowth of experimental melanoma mimotope conformation was also preserved in the
[14] and neuroblastoma [15] tumors. Consequently, conjugated form. KLH itself was only nonspecifically
ch14.18 was applied in several clinical trials against recognized by ch14.18 (Fig. 1, right panel).
melanoma [16], neuroblastoma [17, 18] and also
osteosarcoma [18] with an acceptable safety profile. Selected GD2 mimotopes elicit cross-reactive
The effects of these passive antibody therapies were antibodies
thought to be enhanced by a combination with systemic
cytokines, GM-CSF [19, 20] and IL-2 [21]. In the next Three groups of mice were immunized four times with
step, ch14.18 was expressed as a fusion protein with either GRL-KLH, DGG-KLH, or KLH alone. Subsequently,
IL-2, to target the cytokine directly to the tumor blood was drawn and IgG serum levels were determined.
site [22]. This compound is already being tested in To enable comparison between the groups, in a first step,
clinical trials [23]. antibodies directed against the carrier molecule, KLH,
As an alternative to these passive antibody applica- were measured in the ELISA format. All three groups
tions, active immunizations against GD2 have been showed comparable anti-KLH values (OD 450–570 nm
investigated. However, the antigen itself cannot be used 0.388  0.004, 0.396  0.005, and 0.418  0.012 for
as an effective immunogen due to its glycolipid nature. mice immunized with GRL-KLH, DGG-KLH, and KLH,
Carbohydrates are mostly T cell-independent antigens, respectively), indicating successful immunization in all
and as no T cell help is involved, relatively low antibody three groups. We then proceeded to determine the anti-
titers and no memory responses are induced. Therefore, mimotope titers. Uncoupled mimotope peptides, a
GD2 has to be antigenically enhanced for immunization control peptide, and the carrier protein were dotted
purposes. One possibility is coupling of the ganglioside onto a nitrocellulose membrane, and incubated with
to an immunogenic carrier [24]; another is the serial dilutions of sera from mimotope-KLH-immunized
translation of the carbohydrate antigen into a mimicking mice. As seen in the ELISA assay, anti-KLH titers were
peptide [25] or protein [26–28] structure. Our group comparable for both groups (Fig. 2, KLH panels).
has generated circular decapeptide mimics of the 14.18 Mimotope-induced antibodies were found to be cross-
epitope on the GD2 antigen using the phage display reactive for both peptides, i.e., GRL-KLH-immunized
technique described in a previous study [25]. They were mice recognized GRL as well as DGG, and DGG-KLH-
proven to be true epitope mimics (so called mimotopes)
by mimicry tests in the ELISA format and by 3-D
computer modeling. The aim of the present study was to
evaluate the mimotopes for their capability of inducing a
GD2-specific humoral immune response.

Results
Figure 1. Synthetic peptide mimics of the ch14.18 epitope on
disialogangliside GD2 are specifically recognized by ch14.18.
The vaccine constructs Left panel: Cyclic peptides GRL and DGG, dotted in triplicates,
are recognized by ch14.18, but not by the isotype control
Out of the 13 mimotopes generated for the ch14.18 antibody cetuximab, or detecting anti-human IgG antibody
epitope, the two cyclic peptides showing the best (buffer control). Right panel: After conjugation to the im-
munogenic carrier molecule KLH, cyclic GRL and DGG
mimicry results [25], C-GRLKMVPDLE-C and C-
mimotope conformation is preserved. ch14.18 recognized the
DGGWLSKGSW-C, were selected for immunization mimotope conjugates (GRL-KLH and DGG-KLH), but only
studies. For this, they were produced synthetically. nonspecifically interacts with the carrier KLH. Controls are
Before proceeding to immunizations, the synthetic again clear.

f 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji.de


Eur. J. Immunol. 2006. 36: 1267–1274 Molecular immunology 1269

Figure 2. Sera of mice immunized with mimotope conjugates


are cross-reactive with both peptides mimicking the ch14.18
epitope on GD2. Mice were immunized with GRL-KLH or DGG- Figure 3. Mimotope-induced antibodies recognize the original
KLH. Total Ig titer determinations by DotBlot against the antigen, disialoganglioside GD2. Mice immunized with GRL-
peptides GRL and DGG, against the carrier KLH, and against a KLH or DGG-KLH developed IgG antibodies specifically reacting
control peptide are shown. Serum dilutions are given in the with GD2, but not with the control ganglioside GM1, in ELISA.
figure. KLH-immunized mice show no reactivity with either ganglio-
side. Box plots show median, 25th and 75th percentiles of IgG
levels of respective groups. Both differences of mimotope
immunized mice recognized DGG as well as GRL (Fig. 2, groups in GD2/GM1 recognition, as well as differences between
GRL and DGG panels). As both peptides, although mimotope and control groups in GD2 recognition, are
different in sequence, were characterized to be 3-D statistically significant (p<0.05) by two-tailed Student's t-test.
mimics of the same epitope, this finding is not
surprising, but supports their structural equivalence. carrier groups again was found to be significant
Interestingly, both GRL-KLH- and DGG-KLH-immunized (p<0.001 for GRL-KLH, and p<0.01 for DGG-KLH).
mice reacted more strongly with the GRL peptide. We thus demonstrated that immunization with the
Neither group recognized a cyclic decamer control selected GD2 mimotopes indeed induces a specific anti-
peptide (Fig. 2, bottom panels), so the induced GD2 immune response. Ig subclasses and exact amounts
antibodies are demonstrated to be specific for the of anti-GD2 antibodies (shown in Table 1) elicited by
GD2 mimotopes. mimotope vaccination were also determined by gang-
lioside ELISA. In serum dilution series, IgM could be
Mimotope-induced antibodies recognize the detected up to dilutions of 1:3000 in both mimotope
original antigen GD2

Table 1. Anti-GD2 antibody quantities (lg/mL) in mouse sera


To assess whether the induced antibodies not only react
after mimotope immunization
with the actual immunogen, but also with the original
antigen, GD2, a ganglioside ELISA was performed. Both Mimotope used for immunization
mimotope-KLH conjugates induced antibodies that Antibody subclass
C-GRLKMVPDLE-C C-DGGWLSKGSW-C
reacted specifically with GD2, but not with the control
IgM 210.00 50.00
ganglioside GM1 (Fig. 3). This difference in recognition
was found to be statistically significant (p<0.001 for IgG1 2.30 2.20
GRL-KLH-immunized mice, and p<0.05 for DGG-KLH- IgG2a 0.55 1.78
immunized mice). Mice immunized with the carrier KLH IgG2b 0.47 0.20
alone did not recognize either ganglioside, and the
IgG3 – –
difference in GD2 recognition between mimotope and

f 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji.de


1270 Angelika B. Riemer et al. Eur. J. Immunol. 2006. 36: 1267–1274

tope-immunized mice recognized GD2 at a band at


55 kDa in Western blots. Binding intensity was found to
be dose dependent. Serum from mice immunized with
the carrier KLH alone did not recognize the antigen
(Fig. 4, results for the SK-NA-S cell extract). After
deglycosylation of the blots, the 55-kDa band was no
longer detectable with ch14.18 or the immune sera,
indicating that the antibodies indeed recognize the
carbohydrate moiety of GD2. These data show that the
mimotope-induced antibodies recognize GD2 not only
in the purified preparation, which had also been used in
Figure 4. Immunodetection of GD2 by anti-mimotope sera in a
characterizing the mimotopes [25], but also in its native
Western blot of a SK-N-AS cell lysate. Blotted GD2 is detected by
ch14.18 as a band at 55 kDa. Sera from mice immunized with form.
the GRL-KLH and DGG-KLH conjugates also recognize GD2 To demonstrate whether anti-mimotope antibodies
(serum dilutions given in figure, total Ig detected), whereas sera also react with GD2 on cell surfaces, we performed
from mice immunized with the carrier KLH alone show no immunofluorescence staining of M21 and SK-NA-S cells.
reactivity with the antigen.
mAb ch14.18 was used as a positive control (Fig. 5A,
results of the M21 staining). Sera of mice immunized
groups, IgG1 up to 1:300 in the GRL-KLH and up to with GRL-KLH (Fig. 5B) or DGG-KLH (Fig. 5C) showed
1:1000 in the GDD-KLH group, IgG2a up to 1:1000 in marked membranous staining of GD2-expressing cells.
both groups, and IgG2b up to 1:300 and 1:100, As seen in the GD2 ELISA, anti-GRL-KLH antibodies
respectively. No IgG3 or IgA could be detected in the reacted slightly more strongly with the target cells. Only
sera. background staining was observed when serum anti-
bodies from mice immunized with KLH alone were
Mimotope-induced antibodies recognize GD2 in tested (Fig. 5D). SW480 control cells did not stain with
melanoma and neuroblastoma cell extracts and any antibody (data not shown).
on the cell surface

To further confirm the relevance of the mimotope- Discussion


induced antibodies, we sought to show that they also
recognize GD2 on tumor cells. Cell extracts of GD2- In the present study, we show that an epitope of the
expressing M21 melanoma cells and SK-NA-S neuro- carbohydrate antigen GD2 can be successfully trans-
blastoma cells were prepared and separated electro- lated into immunogenic peptide epitope mimics. Dis-
phoretically. In both extracts, ch14.18, which was ialoganglioside GD2 belongs to the class of TACA, and is
applied as a positive control, and sera from mimo- expressed on malignancies of neuroectodermal origin.

Figure 5. Immunofluorescence staining of GD2 on M21 melanoma cells. Anti-GD2 IgG antibodies were detected by FITC-
conjugated secondary antibodies. Nuclei were stained with Hoechst dye. Cells were viewed with a Zeiss Axioplan 2 fluorescence
microscope. (A) Anti-GD2 mAb ch14.18 (positive control). (B) Sera from mice immunized with GRL-KLH. (C) Sera from mice
immunized with DGG-KLH. (D) Sera from mice immunized with KLH alone do not specifically recognize GD2.

f 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji.de


Eur. J. Immunol. 2006. 36: 1267–1274 Molecular immunology 1271

For these tumors, such as melanoma and neuroblasto- recombinant Fab without the Fc portion. This limits their
ma, therapeutic options in advanced disease are limited. practicability for vaccine formulation.
Therefore, novel treatment modalities are being sought. Another possibility to generate mimics of any
GD2 has so far been exploited as a passive immunother- structure recognized by an antibody is the phage display
apy target with the chimeric mAb ch14.18 in neuro- technique [33]. It is a technique to define peptides
blastoma and melanoma trials. However, passive anti- mimicking natural epitopes, including carbohydrate
body applications have several drawbacks. First, these structures [34–36]. Phage display peptide libraries
antibodies need to be engineered to be human/mouse consist of filamentous phage particles displaying
chimeric or preferably fully humanized, to prevent random peptides of defined length on their surface.
formation of human anti-mouse antibodies (HAMA) in By biopanning [37] such phage display libraries with an
the patient. Also with ch14.18, which is a human/mouse antibody of interest, epitope mimics, so-called mimo-
chimeric antibody, the development of human anti- topes, can be selected. These peptides mimic the original
chimeric antibodies has been seen [20]. Second, antigen by virtue of their 3-D structure, in which the
'artificial' antibodies have to be repeatedly administered amino acids of the peptide adopt the same shape as the
to achieve continuous serum levels that are needed for epitope recognized by the selecting antibody [38, 39].
anti-tumor efficacy. This dependency on weekly infu- Thus, the poorly immunogenic carbohydrate antigens
sions is extremely demanding for cancer patients. Third, can be converted into peptide epitope mimics, which
the production costs of the amounts of chimeric or have a much greater potential of both antigenicity and
humanized antibodies needed for passive immunother- memory induction. Another strength of vaccinations
apy are enormous. For all these reasons, an active with these epitope mimics is that by virtue of their not
immunotherapy that would induce anti-GD2 antibodies being identical to the original antigen, immunological
would be an attractive solution, both circumventing tolerance, which exists for self antigens, can be
multiple infusions, as well as the danger of inducing an circumvented. Additionally, in the vaccine formulation,
immune response against the non-human parts of the a highly immunogenic carrier molecule is included,
artificial antibodies. The continuous availability of providing epitopes for eliciting T cell help. Moreover, the
'natural', polyclonal antibodies induced by active vaccine is administered with an immunological
immunization is expected to show all the beneficial adjuvant. Taking these three strategies together,
properties of passive immunotherapy, with the addi- immunological tolerance to GD2 can be overcome.
tional advantage that the induced antibodies are not of The mimotope strategy has already been successfully
a single given antibody subclass. By being both pursued for a number of carbohydrate antigens, including
polyclonal and of different isotypes, these antibodies several TACA [35, 40–42], with the resulting mimotopes
are expected to be capable of triggering the whole array being used for immunizations either coupled to KLH [43],
of antibody-mediated immune functions against the or encoded as minigenes in a DNA vaccine approach [44].
tumor. In a previous study, our group defined such mimotopes of
Indeed, active immunizations against GD2 have been the epitope recognized by mAb ch14.18 on GD2 [25].
pursued for a long time. In initial studies, gangliosides These were, to the best of our knowledge, the first peptide
were used directly as immunogens [29, 30], a strategy mimics generated for this ganglioside antigen. Early last
that encountered problems as carbohydrates are in- year, another group described the definition of a GD2
trinsically T cell-independent antigens, which only elicit mimotope [45], which they encoded in a minigene for a
weak immune responses and mostly antibodies of the DNA vaccination approach. In the present study, we
IgM subclass. To overcome these limitations, the weakly aimed to assess whether 'our' cyclic mimotopes were
immunogenic gangliosides were coupled to the im- immunogenic and whether the induced antibodies
munogenic carrier molecule KLH [24]. Currently, a recognize the original antigen GD2. To ensure correct
GD2-KLH conjugate vaccine is being tested in patients cyclization and thus peptide conformation, we chose to
with malignant melanoma [31]. have them produced synthetically, and coupled to KLH.
Yet other approaches seek to replace the carbohy- We were able to show that two selected GD2 epitope
drate moiety with its immunological drawbacks alto- mimics elicited a humoral immune response. The induced
gether. The first possibility to mimic carbohydrates with antibodies were cross-reactive for the two peptides, a
a protein structure was the development of anti- finding that strengthens the mimotope hypothesis.
idiotypic antibodies (anti-Id). Indeed, several anti-Id Obviously, both peptides, although different in sequence,
mimicking GD2 have been generated [26–28], and one share the same 3-D characteristics, which mimic the
of them has already been used as a vaccine in advanced ch14.18 epitope. Importantly, the anti-mimotope anti-
melanoma patients [32]. However, anti-Id are still mAb, bodies confirmed this mimicry by specifically recognizing
and, when applied as antigens, can give rise to HAMA. the original antigen, GD2 in ELISA, Western Blot, and on
So either they need to be humanized, or produced as the cell surface. We thus demonstrated that the GD2

f 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji.de


1272 Angelika B. Riemer et al. Eur. J. Immunol. 2006. 36: 1267–1274

ganglioside antigen can be translated into peptide (EMD-Lexigen, Lexington, MA). Cetuximab, also a mouse/
mimotopes for vaccination purposes. human chimeric IgG1 mAb, directed against EGFR, was used as
Peptide vaccines that can be synthesized custom- an isotype control.
made are cost-effective, simple to produce, and can
easily be quality-controlled during the manufacturing Synthesis of vaccine constructs and DotBlot assay
process. They are chemically stable and contain no
oncogenic, toxic, or infectious material. As active The 1,12-cyclic peptides C-GRLKMVPDLE-C (GRL) and C-
immunogens, synthetic mimotopes can continuously DGGWLSKGSW-C (DCC) were manufactured synthetically
induce available tumor-targeting antibodies. These (piChem, Graz, Austria). Conformational accuracy was
characteristics answer the points raised as disadvantages verified with ch14.18 in a DotBlot assay. In brief, the peptides
of passive antibody therapies, i.e., high costs and were solubilized in PBS/20% dimethylformamide, and dotted
onto a nitrocellulose membrane at 1 mg/mL. Blot strips were
multiple necessary infusions. Additionally, the induction
incubated with ch14.18 or cetuximab, respectively, and bound
of undesired antibodies (e.g., HAMA) is ruled out, as not
antibody was detected by peroxidase-conjugated anti-human
only a pure antigen, but a specific epitope is applied. IgG (Jackson ImmunoResearch, West grove, PA) using the
Moreover, the resulting 'natural' humoral and cellular ECL+ chemiluminescence detection protocol (Amersham
immune responses against GD2, compared to passive Pharmacia Biotech, Little Chalfont, UK). Subsequently, the
anti-GD2 immunotherapy, may even mediate enhanced peptides were coupled via a linker (GPGPG) and S-acetyl-thio-
anti-tumor effects. acetate on their C terminus to a succinimidyl-4-(N-maleini-
In conclusion, we have shown that an epitope of a midomethyl)cyclohexan-1-carboxylate-activated immuno-
carbohydrate antigen can successfully be translated into genic carrier, KLH. The conjugate was again checked for
immunogenic peptide mimotopes. Furthermore, immu- ch14.18 binding capability in a DotBlot assay (as above).
nizations with these epitope mimics induced antibodies
again recognizing the original carbohydrate, in our case Immunization of BALB/c mice
the disialoganglioside GD2. As the elicited antibodies
were of the IgG isotype, we could show that with Three groups (n=8) of BALB/c mice (Charles River Labora-
repeated vaccinations the mimotope conjugates are tories, Sulzfeld, Germany) were immunized i.p. with 10 lg of
capable of recruiting T cell help and inducing memory the mimotope conjugates, GRL-KLH and DGG-KLH, or the
phenomena. We thus provide evidence that GD2 carrier protein KLH alone, respectively, on days 1, 15, 36 and
mimotopes are suitable candidates for active immu- 57. Aluminum hydroxide was used as an adjuvant in all groups.
Blood was taken from the tail vein on days 0 (preimmune
notherapy of GD2-expressing tumors, such as melanoma
serum), 22, 43 and 64. Mice were treated according to
and neuroblastoma.
European Union Rules of Animal Care, with permission no.
66.009/35-BrGT/P2004 from the Austrian Federal Ministry of
Materials and methods Education, Science and Culture.

Cell lines and cell lysates Titer determination

Two GD2-positive cell lines were used in this study, the human All titers were determined using the third immune sera
melanoma cell line M21 and the human neuroblastoma cell (collected on day 64). Anti-carrier titers were assessed by
line SK-NA-S. The human colon adenocarcinoma cell line ELISA. For anti-carrier titers, ELISA plates (Nunc, Roskilde,
SW480, which is GD2 negative, was employed as a negative Denmark) were coated with KLH, 1 lg/mL in bicarbonate
control. All cells were grown in RPMI medium (Gibco BRL, buffer, pH 9.6, by overnight incubation at 4 C. Plates were
Inchinnan, UK) supplemented with 10% fetal calf serum, 1% then washed with PBS/0.05% Tween 20, and nonspecific
glutamine, and 1% penicillin/streptomycin. binding was blocked by incubation with PBS/1% dry milk
Total cell lysates were prepared as described previously powder. Sera were added at a dilution of 1:100 in PBS/0.01%
[38], with a lysis buffer containing 20 mM Tris, 150 mM NaCl, dry milk. Bound antibodies were detected with a peroxidase-
1 mM EDTA, 1 mM EGTA, 1% Triton X-100 and a protease conjugated rat-anti-mouse IgG antibody (Jackson Immuno
inhibitor cocktail (Complete, Roche, Basel, Switzerland), at Research Laboratories, Inc., West Grove, PA). The reaction was
pH 7.5. The protein concentration was determined photome- developed with TMB substrate (BD Biosciences, San Diego,
trically, using bicinchoninic acid (BCA Protein Assay Kit, CA). OD was measured in an ELISA reader (Dynatech,
Pierce, Rockford, IL). Extracts were aliquoted and stored Denkendorf, Germany) at 450–630 nm.
at –80 C. Anti-peptide titers were determined by incubation of serial
dilutions of pooled sera (1:5000; 1:10 000; 1:50 000;
Monoclonal antibodies 1:100 000; 1:1 000 000) with dotted mimotope peptides, a
control peptide, and KLH. Bound antibodies were detected
ch14.18, a mouse/human IgG1 mAb, directed against with a peroxidase-conjugated sheep-anti-mouse Ig antibody
disialogangliside GD2, was kindly provided by Dr. S. D. Gillies (Amersham) as in the DotBlot assay described above.

f 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji.de


Eur. J. Immunol. 2006. 36: 1267–1274 Molecular immunology 1273

Ganglioside ELISA Hans & Blanca Moser Fund scholarships. The work was
also supported by DFG (Lo 635–2) and Frdergesellschaft
Purified gangliosides GD2 and GM1 (Sigma, St. Louis, MO) Kinderkrebs-Neuroblastomforschung to H.N. Lode. We
were coated at a concentration of 1 lg/mL to ELISA plates thank Harald Kurz and Silke Gruber for excellent
(Nunc) by dilution in ethanol and subsequent evaporation of technical assistance.
the diluent. Plates were washed and blocked as described
above. Third immune sera were added in a dilution series of
1:100–1:3000 in PBS/0.01% dry milk, and bound antibodies
were detected with a peroxidase-conjugated rat-anti-mouse References
IgG antibody (Jackson). For subclass determinations, mouse Ig
1 Hakomori, S., Tumor-associated carbohydrate antigens defining tumor
standard dilution series were coated onto the same ELISA malignancy: basis for development of anti-cancer vaccines. Adv. Exp. Med.
plates. Third immune sera were added to the ganglioside- Biol. 2001. 491: 369–402.
coated wells as described above, while standards were 2 Ritter, G. and Livingston, P. O., Ganglioside antigens expressed by human
incubated with PBS/0.01% dry milk. Rat anti-mouse Ig cancer cells. Semin. Cancer Biol. 1991. 2: 401–409.
subclass antibodies were then added to all wells at a dilution 3 Zhang, S., Cordon-Cardo, C., Zhang, H. S., Reuter, V. E., Adluri, S.,
of 1:500 in PBS/0.01% dry milk, and bound antibodies Hamilton, W. B., Lloyd, K. O. and Livingston, P. O., Selection of tumor
detected with a peroxidase-conjugated mouse anti-rat IgG antigens as targets for immune attack using immunohistochemistry: I. Focus
on gangliosides. Int. J. Cancer 1997. 73: 42–49.
antibody (Jackson). The peroxidase/substratum reactions
were developed and measured as described above. 4 Schulz, G., Cheresh, D. A., Varki, N. M., Yu, A., Staffileno, L. K. and
Reisfeld, R. A., Detection of ganglioside GD2 in tumor tissues and sera of
neuroblastoma patients. Cancer Res. 1984. 44: 5914–5920.
Cell lysate Western blots
5 Cheung, N. K., Saarinen, U. M., Neely, J. E., Landmeier, B., Donovan, D.
and Coccia, P. F., Monoclonal antibodies to a glycolipid antigen on human
M21 and SK-N-AS cell lysates (prepared as described above) neuroblastoma cells. Cancer Res. 1985. 45: 2642–2649.
were separated by SDS-PAGE, and blots incubated with 6 Xiao, W. H., Yu, A. L. and Sorkin, L. S., Electrophysiological characteristics
ch14.18 or serum pool dilutions as detecting antibodies. of primary afferent fibers after systemic administration of anti-GD2
Bound ch14.18 was detected by a peroxidase-conjugated goat ganglioside antibody. Pain 1997. 69: 145–151.

anti-human IgG antibody (Jackson), and bound mouse 7 Cheresh, D. A., Harper, J. R., Schulz, G. and Reisfeld, R. A., Localization of
antibodies by a peroxidase-conjugated sheep anti-mouse Ig the gangliosides GD2 and GD3 in adhesion plaques and on the surface of
human melanoma cells. Proc. Natl. Acad. Sci. USA 1984. 81: 5767–5771.
antibody (Amersham), using the ECL+ chemiluminescence
8 Cheresh, D. A., Rosenberg, J., Mujoo, K., Hirschowitz, L. and Reisfeld, R.
detection protocol (Amersham) and Biomax-MS films (Kodak).
A., Biosynthesis and expression of the disialoganglioside GD2, a relevant
target antigen on small cell lung carcinoma for monoclonal antibody-
Immunofluorohistochemistry mediated cytolysis. Cancer Res. 1986. 46: 5112–5118.

9 Saito, M., Yu, R. K. and Cheung, N. K., Ganglioside GD2 specificity of


M21 and SK-NA-S cells were plated at 5  104 cells/mL on monoclonal antibodies to human neuroblastoma cell. Biochem. Biophys. Res.
eight-well Lab-Tek tissue culture chamber slides (Miles Commun. 1985. 127: 1–7.

Laboratories Inc., Naperville, IL). SW480 cells were used as 10 Cheung, N. K., Kushner, B. H., Cheung, I. Y., Kramer, K., Canete, A.,
negative controls. Cells were grown overnight until half- Gerald, W., Bonilla, M. A. et al., Anti-G(D2) antibody treatment of minimal
residual stage 4 neuroblastoma diagnosed at more than 1 year of age. J. Clin.
confluent. Chamber slides were then cooled to 4 C, and Oncol. 1998. 16: 3053–3060.
washed with ice-cold PBS. Cells were fixed with 4%
11 Kramer, K., Gerald, W. L., Kushner, B. H., Larson, S. M., Hameed, M. and
paraformaldehyde in PBS for 30 min, and chamber slides Cheung, N. K., Disaloganglioside GD2 loss following monoclonal antibody
were incubated with 50 mM NH4Cl in PBS to quench fixation, therapy is rare in neuroblastoma. Med. Pediatr. Oncol. 2001. 36: 194–196.
and blocked with 1% BSA in PBS. Subsequently, cells were 12 Mujoo, K., Cheresh, D. A., Yang, H. M. and Reisfeld, R. A., Disialoganglio-
incubated with ch14.18 (positive control), or with pooled anti- side GD2 on human neuroblastoma cells: target antigen for monoclonal
antibody-mediated cytolysis and suppression of tumor growth. Cancer Res.
GRL-KLH and anti-DGG-KLH third immune sera, and bound 1987. 47: 1098–1104.
antibodies were detected by FITC-conjugated goat anti-mouse
13 Mueller, B. M., Romerdahl, C. A., Gillies, S. D. and Reisfeld, R. A.,
IgG (Caltag Laboratories, Burlingame, CA). Pooled third Enhancement of antibody-dependent cytotoxicity with a chimeric anti-GD2
immune sera from the KLH-immunized mice were used as antibody. J. Immunol. 1990. 144: 1382–1386.
control. Nuclei were stained with 0.1 lg/mL Hoechst dye 14 Kendra, K., Malkovska, V., Allen, M., Guzman, J. and Albertini, M., In vivo
(Sigma) in PBS for 10 min. Cells were mounted in Mowiol binding and antitumor activity of ch14.18. J. Immunother. 1999. 22:
mounting medium and viewed with a Zeiss Axioplan 2 (Carl 423–430.
Zeiss, Jena, Germany). 15 Raffaghello, L., Marimpietri, D., Pagnan, G., Pastorino, F., Cosimo, E.,
Brignole, C., Ponzoni, M. and Montaldo, P. G., Anti-GD2 monoclonal
antibody immunotherapy: a promising strategy in the prevention of
neuroblastoma relapse. Cancer Lett. 2003. 197: 205–209.
Acknowledgements: The work was supported by
16 Saleh, M. N., Khazaeli, M. B., Wheeler, R. H., Allen, L., Tilden, A. B.,
BioLife Science GmbH, Vienna, Austria; by project grant Grizzle, W., Reisfeld, R. A. et al., Phase I trial of the chimeric anti-GD2
no. 10965 of the Austrian National Bank Science Fund; monoclonal antibody ch14.18 in patients with malignant melanoma. Hum.
Antibodies Hybridomas 1992. 3: 19–24.
and by the Center of Excellence in Clinical and
Experimental Oncology (CLEXO), Austrian Federal Min- 17 Handgretinger, R., Anderson, K., Lang, P., Dopfer, R., Klingebiel, T.,
Schrappe, M., Reuland, P. et al., A phase I study of human/mouse chimeric
istry of Education, Science and Culture (GZ 200.062/2-VI/
antiganglioside GD2 antibody ch14.18 in patients with neuroblastoma. Eur.
1/2002). A.B. Riemer and K.H. Brmswig are recipients of J. Cancer 1995. 31A: 261–267.

f 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji.de


1274 Angelika B. Riemer et al. Eur. J. Immunol. 2006. 36: 1267–1274

18 Yu, A. L., Uttenreuther-Fischer, M. M., Huang, C. S., Tsui, C. C., Gillies, S. 31 Ragupathi, G., Livingston, P. O., Hood, C., Gathuru, J., Krown, S. E.,
D., Reisfeld, R. A. and Kung, F. H., Phase I trial of a human-mouse chimeric Chapman, P. B., Wolchok, J. D. et al., Consistent antibody response against
anti-disialoganglioside monoclonal antibody ch14.18 in patients with ganglioside GD2 induced in patients with melanoma by a GD2 lactone-
refractory neuroblastoma and osteosarcoma. J. Clin. Oncol. 1998. 16: keyhole limpet hemocyanin conjugate vaccine plus immunological adjuvant
2169–2180. QS-21. Clin. Cancer Res. 2003. 9: 5214–5220.

19 Murray, J. L., Kleinerman, E. S., Jia, S. F., Rosenblum, M. G., Eton, O., 32 Foon, K. A., Lutzky, J., Baral, R. N., Yannelli, J. R., Hutchins, L.,
Buzaid, A., Legha, S. et al., Phase Ia/Ib trial of anti-GD2 chimeric Teitelbaum, A., Kashala, O. L. et al., Clinical and immune responses in
monoclonal antibody 14.18 (ch14.18) and recombinant human granulocyte- advanced melanoma patients immunized with an anti-idiotype antibody
macrophage colony-stimulating factor (rhGM-CSF) in metastatic melanoma. mimicking disialoganglioside GD2. J. Clin. Oncol. 2000. 18: 376–384.
J. Immunother. Emphasis Tumor Immunol. 1996. 19: 206–217.
33 Scott, J. K. and Smith, G. P., Searching for peptide ligands with an epitope
20 Ozkaynak, M. F., Sondel, P. M., Krailo, M. D., Gan, J., Javorsky, B., library. Science 1990. 249: 386–390.
Reisfeld, R. A., Matthay, K. K. et al., Phase I study of chimeric human/
murine anti-ganglioside G(D2) monoclonal antibody (ch14.18) with 34 Oldenburg, K. R., Loganathan, D., Goldstein, I. J., Schultz, P. G. and
granulocyte-macrophage colony-stimulating factor in children with neuro- Gallop, M. A., Peptide ligands for a sugar-binding protein isolated from a
blastoma immediately after hematopoietic stem-cell transplantation: a random peptide library. Proc. Natl. Acad. Sci. USA 1992. 89: 5393–5397.
Children's Cancer Group Study. J. Clin. Oncol. 2000. 18: 4077–4085. 35 Hoess, R., Brinkmann, U., Handel, T. and Pastan, I., Identification of a
peptide which binds to the carbohydrate-specific monoclonal antibody B3.
21 Albertini, M. R., Hank, J. A., Schiller, J. H., Khorsand, M., Borchert, A. A.,
Gene 1993. 128: 43–49.
Gan, J., Bechhofer, R. et al., Phase IB trial of chimeric antidisialoganglio-
side antibody plus interleukin 2 for melanoma patients. Clin. Cancer Res. 36 Phalipon, A., Folgori, A., Arondel, J., Sgaramella, G., Fortugno, P.,
1997. 3: 1277–1288. Cortese, R., Sansonetti, P. J. and Felici, F., Induction of anti-carbohydrate
antibodies by phage library-selected peptide mimics. Eur. J. Immunol. 1997.
22 Lode, H. N., Xiang, R., Varki, N. M., Dolman, C. S., Gillies, S. D. and
27: 2620–2625.
Reisfeld, R. A., Targeted interleukin-2 therapy for spontaneous neuro-
blastoma metastases to bone marrow. J. Natl. Cancer Inst. 1997. 89: 37 Parmley, S. F. and Smith, G. P., Antibody-selectable filamentous fd phage
1586–1594. vectors: affinity purification of target genes. Gene 1988. 73: 305–318.
23 King, D. M., Albertini, M. R., Schalch, H., Hank, J. A., Gan, J., Surfus, J., 38 Riemer, A. B., Klinger, M., Wagner, S., Bernhaus, A., Mazzucchelli, L.,
Mahvi, D. et al., Phase I clinical trial of the immunocytokine EMD 273063 in Pehamberger, H., Scheiner, O. et al., Generation of peptide mimics of the
melanoma patients. J. Clin. Oncol. 2004. 22: 4463–4473. epitope recognized by trastuzumab on the oncogenic protein Her-2/neu. J.
Immunol. 2004. 173: 394–401.
24 Zhang, H., Zhang, S., Cheung, N. K., Ragupathi, G. and Livingston, P. O.,
Antibodies against GD2 ganglioside can eradicate syngeneic cancer 39 Riemer, A. B., Hantusch, B., Sponer, B., Kraml, G., Hafner, C., Zielinski,
micrometastases. Cancer Res. 1998. 58: 2844–2849. C. C., Scheiner, O. et al., High-molecular-weight melanoma-associated
antigen mimotope immunizations induce antibodies recognizing melanoma
25 Frster-Waldl, E., Riemer, A. B., Dehof, A. K., Neumann, D., Bramswig,
cells. Cancer Immunol. Immunother. 2005. 54: 677–684.
K., Boltz-Nitulescu, G., Pehamberger, H. et al., Isolation and structural
analysis of peptide mimotopes for the disialoganglioside GD2, a neuro- 40 Kieber-Emmons, T., Luo, P., Qiu, J., Agadjanyan, M., Carey, L., Hutchins,
blastoma tumor antigen. Mol. Immunol. 2005. 42: 319–325. W., Westerink, M. A. and Steplewski, Z., Peptide mimicry of adenocarci-
noma-associated carbohydrate antigens. Hybridoma 1997. 16: 3–10.
26 Saleh, M. N., Stapleton, J. D., Khazaeli, M. B. and LoBuglio, A. F.,
Generation of a human anti-idiotypic antibody that mimics the GD2 antigen. 41 Kieber-Emmons, T., Luo, P., Qiu, J., Chang, T. Y., O, I., Blaszczyk-Thurin,
J. Immunol. 1993. 151: 3390–3398. M. and Steplewski, Z., Vaccination with carbohydrate peptide mimotopes
promotes anti-tumor responses. Nat. Biotechnol. 1999. 17: 660–665.
27 Foon, K. A., Sen, G., Hutchins, L., Kashala, O. L., Baral, R., Banerjee, M.,
Chakraborty, M. et al., Antibody responses in melanoma patients 42 Monzavi-Karbassi, B., Cunto-Amesty, G., Luo, P. and Kieber-Emmons, T.,
immunized with an anti-idiotype antibody mimicking disialoganglioside Peptide mimotopes as surrogate antigens of carbohydrates in vaccine
GD2. Clin. Cancer Res. 1998. 4: 1117–1124. discovery. Trends Biotechnol. 2002. 20: 207–214.
28 Basak, S., Birebent, B., Purev, E., Somasundaram, R., Maruyama, H., 43 Hou, Y. and Gu, X. X., Development of peptide mimotopes of
Zaloudik, J., Swoboda, R. et al., Induction of cellular immunity by anti- lipooligosaccharide from nontypeable Haemophilus influenzae as vaccine
idiotypic antibodies mimicking GD2 ganglioside. Cancer. Immunol. Immun- candidates. J. Immunol. 2003. 170: 4373–4379.
other. 2003. 52: 145–154.
44 Kieber-Emmons, T., Monzavi-Karbassi, B., Wang, B., Luo, P. and Weiner,
29 Livingston, P. O., Ritter, G. and Calves, M. J., Antibody response after D. B., Cutting edge: DNA immunization with minigenes of carbohydrate
immunization with the gangliosides GM1, GM2, GM3, GD2 and GD3 in the mimotopes induce functional anti-carbohydrate antibody response. J.
mouse. Cancer Immunol. Immunother. 1989. 29: 179–184. Immunol. 2000. 165: 623–627.

30 Portoukalian, J., Carrel, S., Dore, J. F. and Rumke, P., Humoral immune 45 Bolesta, E., Kowalczyk, A., Wierzbicki, A., Rotkiewicz, P., Bambach, B.,
response in disease-free advanced melanoma patients after vaccination with Tsao, C. Y., Horwacik, I. et al., DNA vaccine expressing the mimotope of
melanoma-associated gangliosides. EORTC Cooperative Melanoma Group. GD2 ganglioside induces protective GD2 cross-reactive antibody responses.
Int. J. Cancer 1991. 49: 893–899. Cancer Res. 2005. 65: 3410–3418.

f 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji.de

You might also like