ArticlePDF Available

Development of a Human Monoclonal Antibody to Ganglioside GM2 with Potential for Cancer Treatment

Authors:

Abstract and Figures

A human B-lymphoblastoid cell clone, L55-81, that produces human monoclonal antibody (MAb) to ganglioside G(M2) was established from peripheral blood B lymphocytes of a melanoma patient. L55-81 secretes IgMkappa light chain antibody in a serum-free medium. G(M2) specificity of the antibody was tested by immune adherence assay, TLC immunostaining, and ELISA. Anti-G(M2) antibody was shown to have the ability to kill the G(M2)-rich human melanoma cell line M14 in the presence of human or rabbit complement. A purified L55-81 MAb (>99.5% purity in protein concentration) was biotinylated and tested for its reactivity to various histological-type biopsied tumor and normal tissues in an avidin-biotin detection system. L55-81 MAb (20 microg/ml) reacted with several types of tumor tissues such as melanoma (7 of 10), colon carcinoma (4 of 5), ovary carcinoma (4 of 5), breast carcinoma (1 of 5), kidney carcinoma (1 of 5), and prostate carcinoma (1 of 5). None of the normal tissues derived from 24 different organs and adjacent normal tissues surrounding the cancerous tissues were stained. Production of the antibody in a serum-free medium, the cytotoxic potential with human complement, the inability to react to normal tissues, and the ability to target antigen-specific target cells make L55-81 a potential therapeutic agent for the treatment of cancers expressing ganglioside G(M2).
Content may be subject to copyright.
(CANCERRESEARCH56. 5666-5671. December 15, 19961
ABSTRACT
A human B-lymphoblastoid cell clone, L55-81, that produces human
monoclonal antibody (MAb) to ganglioside G@ was established from
peripheral blood B lymphocytes of a melanoma patient. L55-81 secretes
IgMK light chain antibody in a serum-free medium. GM2 specificity of the
antibody was tested by immune adherence assay, TLC immunostaining,
and ELISA. Anti-GM2antibody was shown to have the ability to kill the
GM2-rich human melanoma cell line M14 in the presence of human or
rabbit complement. A purified L55-81 MAb (>99.5% purity in protein
concentration) was biotinylated and tested for its reactivity to various
histological-type biopsied tumor and normal tissues in an avidin-biotin
detection system. L55-81 MAb (20 tag/mI) reacted with several types of
tumor tissues such as melanoma (7 of 10), colon carcinoma (4 of 5), ovary
carcinoma (4 of 5), breast carcinoma (1 of 5), kidney carcinoma (1 of 5),
and prostate carcinoma (1 of 5). None of the normal tissues derived from
24 different organs and adjacent normal tissues surrounding the cancer
ous tissues were stained. Production of the antibody in a serum-free
medium, the cytotoxic potential with human complement, the inability to
react to normal tissues, and the ability to target antigen-specific target
cells make L55-81 a potential therapeutic agent for the treatment of
cancers expressing ganglioside GM2.
INTRODUCTION
Ganglioside GM2 [GalNAcj3l-4(NeuNAccs2-3)Galf3l-4Glcj3l-lce
ramide] is a major tumor-differentiation antigen in human melanoma
(1—3)and hepatoma (4). Ganglioside antigens have been one of the
prominent antigens used as targets for the study of immunotherapy of
cancer using MAbs3 (5, 6). Murine MAbs reactive to ganglioside
antigens have shown partial or complete responses in the treatment of
neuroblastoma and melanoma patients (7—15).However, the effec
tiveness is usually only transient, and higher doses or repeated anti
body injections were not attempted in these trials due to the toxicity
or human anti-mouse antibody responses in patients. Because human
MAbs do not usually recognize human normal tissues, they offer
potential advantages over murine MAbs and chimeric human-mouse
MAbs for cancer therapy. Human MAbs do not elicit human anti
mouse antibody responses and interact more efficiently with the
human effector system to activate potentially therapeutically useful
functions. In spite of these obvious advantages, the number of human
MAt, cell lines that stably secrete a high density of human MAb has
been few; thus, their therapeutic potential has not yet been deter
mined. In 1982 and 1983, our laboratory developed two human MAbs,
L72 and L55, that had specificity for ganglioside GD2 and GM2,
respectively (1, 16, 17), and, more recently, developed human MAb
L612 to ganglioside GM3 (18). The immunoglobulin class of these
antibodies is 1gM with a biologically functional Fc portion.
These cell lines were initially grown in a culture medium contain
Received 7/2/96; accepted 10/16/96.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance with
18 U.S.C. Section 1734 solely to indicate this fact.
I Supported by NIH/National Cancer Institute Grants CA30647 and CA 12582.
2 To whom requests for reprints should be addressed, at Department of Biotechnology
Sciences, John Wayne Cancer Institute, 2200 Santa Monica Boulevard, Santa Monica, CA
90404.Phone:(310)449-5218;Fax:(310)998-3915.
3 The abbreviations used are: MAb, monoclonal antibody; IA, immune adherence;
AJCC, American Joint Committee on Cancer.
ing FCS. After repeated recloning and testing of the cell lines in
several different commercially available serum-free media, some but
not all cell lines were successfully adapted to a serum-free medium. In
an effort to establish cell lines that could produce human MAb to GM2
in a serum-free medium, we cloned two new human B cell lines
(L55-8 1 and L55-637) and successfully adapted one (L55-81) of these
cell lines to AIM-V. The adaptation of human 1gM MAb-producing
cell lines to a serum-free medium is essential for clinical therapeutic
trials because it is nearly impossible to purify 1gM antibody to a
clinical grade from spent medium containing animal serum within a
reasonable cost.
In this investigation, we have assessed three major preclinical
criteria of the suitability of the antibody for passive immunotherapy of
cancer: (a) the epitope specificity; (b) antitumor cytotoxic potential;
and (c) the level of reactivity to cancer and normal tissues. The
L55-81 cell line not only produces human MAb in serum-free medium
but also secretes a greater amount of antibody (10—30 @g/ml)than our
original L55 cell line (1—4@tg/ml)in standard plastic flasks. When the
cell line is grown in a hollow fiber system (AcuSyst-Maximizer 1000;
Cellex, Minneapolis, MN), the antibody concentration increased to
200—400 @g/ml.Using a standard procedure of 1gM purification, we
were able to achieve over 99.5% purity of L55-8l antibody. The
present study was designed to investigate at a preclinical level the
therapeutic usefulness of human MAb to GM2. The antibody was
demonstrated to be highly specific to cancer tissues and killed anti
gen-positive cancer cells in the presence of complement. As suggested
in a previous report citing the original L55 antibody (17), the L55-81
antibody reacted not only to one particular histological type of cancer
but to a variety of cancer tissues. No normal tissues obtained from 24
different organs or normal parts of tissues surrounding cancer tissues
were positive for the antibody. This indicates that the antibody may be
applicable to the treatment of many different cancers without causing
toxicity.
MATERIALS AND METHODS
Establishment of Human MAb to Ganglioside G@. Human B cell lines
that produce human MAb specific for ganglioside GM2were established from
peripheral blood B lymphocytes of a melanoma patient using the EBV trans
formation technique as described previously (17). The transformed B-lympho
blastoid cells that produced human MAb to GM2 were viably frozen. The cells
were thawed, and additional clonings were performed to obtain two relatively
higher-producer L55-8l and L55-637 cell lines. These antibodies were of 1gM
class with Klight chains. After recloning three times in serum-free medium
(AIM-V; Life Technologies, Inc., Gaithersburg, MD), L55-8l was successfully
adapted to the medium, whereas L55-637 failed to grow. Human 1gMsecreted
in the spent tissue culture medium was measured by a double sandwich ELISA.
Functional antibodies were monitored by IA assay using GM2-rich human
melanoma cell line M14 (17) and ELISA using purified GM2(19, 20). The
epitope specificity of the antibodies was determined by ELISA (20) and TLC
immunostaining (21) using a variety of gangliosides and neutral glycolipids as
antigen sources.
Glycolipids. Glycosphingolipids from melanoma cell lines used in TLC
immunostaining assay to determine the specificity of human MAb were
extracted and purified according to methods reported previously (2). Commer
cially available human and bovine gangliosides and neutral glycolipids used in
ELISA were purchased from Sigma (St. Louis, MO), KG. Pullman (Munich,
5666
Development of a Human Monoclonal Antibody to Ganglioside G@ with Potential
for Cancer Treatment1
Yumiko Nishinaka, Mepur H. Ravindranath, and Reiko F. Irie2
Departments of Biotechnology Sciences fY. N., R. F. I.] and Glycolipid immunotherapy (M. H. RI, John Wayne Cancer Institute, Santa Monica, California 90404
on March 20, 2017. © 1996 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
Table 1 Reactivity of 155-81 human MAb in IA assay to five human melano
lines with different ganglioside profilesma
cellMelanoma
cell linesIA
score― Gangliosides―(nmol/g
dry weight of cell)
L55-81 L6l2 GM2 GD2GM3GD3Ml4
M24
M12
Ml5
M25+++
+ 4.0 2.4 7.3
+ — 8.2 0.7 0.3
— +++ <0.1 <0.1 18.1
— +++ 0.5 <0.1 20.6
— — <0. 1 0. 1 1 1 .74.2
0.4
2.8
2.0
10.0
HUMAN MAb TO GANGLIOSIDE GM2
Germany), or Advanced Immuno Chemical, Inc. (Long Beach, CA). A quan
titative determination of the individual glycosphingolipids was performed by
TLC densitometry, as described in a previous report (21).
Complement-dependent Cytotoxicity Assay. Cytotoxicitywas measured
in a 4-h 51Cr release assay. Target cells (1 X 106 cells) were labeled with 160
@tCi51Cr for 1 h at 37°C.After washing, the labeled cells (1.2 X l0@cells)
were mixed with 120 p1 of antibodies diluted with veronal buffer-containing
complement at 1:5 ratio. After a 4-h incubation at 37°C,the 5tCr release
activity in the supernatants was counted. The percentage of specific@ Cr
release was determined according to the following formula.
Percentage of specific release
— (release with antibody and complement— spontaneous release) < 100
— (maximum release — spontaneous release)
All test systems were done in triplicate for each experiment.
Biotinylatlon of the Antibody. Biotinylation of L55-8l human MAb was
performed with biotin hydrazide (Pierce, Rockford, IL) according to the
manufacturer's protocol. Briefly, antibody (0.1—0.5mg/ml) purified by ion
exchange chromatography and gel filtration was dialyzed against 0.1 Macetate
buffer (pH 5.5) containing 0.1 MNaCI. A 0.1 volume of 0.1 MNaIO4 was
added to the antibody solution to oxidize the antibody for 20 mm on ice in the
dark. The oxidized antibody was separated from excess reagent by a Sephadex
G-25 (PD-b) column. A 0.2 volume of 10 mr@ibiotin hydrazide was added to
the oxidized antibody solution. After incubation for 1 h at room temperature,
the biotinylated antibody was separated from excess biotin hydrazide by a
PD-b column.
Immunohistochemistry.Humanmalignantand normal specimenswere
collected shortly after surgery, immediately snap-frozen in liquid nitrogen, and
stored at —70°C.Four-p@mcryostat sections of Tissue-Tek-embedded tissues
were prepared and frozen at —20°Cuntil staining. Cytospin slides for mela
noma cell lines were prepared by spinning cells for S mm at 1000 rpm. Slides
were air-dried overnight at room temperature and stored at 4°Cuntil staining.
Cells and tissues on slides were fixed with cold acetone before immuno
staining. Immunostaining was carried out in an avidin-biotin complex immu
noperoxidase system. Sections were sequentially blocked with hydrogen per
oxide (0.3%) for 5 mm, avidin D blocking solution for 15mm, biotin blocking
solution (Vector SP200l ; Vector Laboratories, Inc., Burlingame, CA) for 15
mm,and4%BSAfor20mm.Biotinylatedantibodywasaddedtothe sections
and incubatedfor 1 h in a humiditychamberat room temperature.Sections
were stained using the Vectastain ABC Elite Reaction Kit solution (Vector
Laboratories, Inc.) and counterstained with Harris-hematoxylin. Staining in
tensity of the tissues was graded as + + + (very strong), + + (strong), + (mod
crate), ±(weak), and —(negative).
RESULTS
Binding Specificity of L55-81 Human MAb to GM2. The binding
specificity of L55-8l human MAb to the cell surface of melanoma cell
lines was assessed by IA assay using five different melanoma cell
lines expressing different GM2 antigen density. L55-81 demonstrated
positive reactivity to melanoma cell lines with a relatively high GM2
3.0
E 2.5
@2.0
@ 1.5
11.0
,@ 03
0.0 AsGM1 GM! GD1a GT1b GD1b GM3 GM2 GD3 GD2
Glycolipids Antigens
Fig. 1. Various glycolipids were tested for the binding activity of L55-81 human MAb
in ELISA. The human MAb (800 @sg/ml)was diluted with PBS-4% human serum
albumin: U, 1:50; @,1:100: D, 1:200. Glycolipids (3 nmol/well) were coated on each
well, and all tests were done in duplicate.
content, such as M14 and M24. Other cell lines (M12, MIS, and M25)
that expressed low levels of GM2 showed no binding of this human
MAb at a dose of 10 @tg/mi(Table 1). As a control, the binding of
anti-GM3 human MAb L-612 (18) to these cell lines was assessed
using the same assay. Cell lines with a high GM3 content (Ml2 and
MiS) had strong antibody binding, whereas those with relatively less
GM3 (such as M14, M24, and M25) had no or moderate binding.
These results demonstrated that these human MAbs had a differential
reactivity among cells with different ganglioside expression and sug
gested that L55-81 human MAb had specificity for GM2.
To confirm the specificity of L55-8l human MAb for GM2, ELISA
and TLC immunostaining were performed using a variety of ganglio
sides and neutral glycolipids. In ELISA, a significant absorbance
value was obtained only with GM2, and no positive reactivity was
obtained with asialo GMI, GM!, GD1,,,GT1b, GDIb, GMI, GD3, and GD2
(Fig. 1). For TLC immunostaining, the total gangliosides extracted
from M14 and M24, as well as a purified GM2 from M14, were used
as antigen sources. Fig. 2 is a representative staining result with the
L55-8l MAb. GM2 was the only ganglioside to react with the human
MAb, indicating that the human MAb has no cross-reactivity with
other gangliosides.
Antitumor Activity of L55-81 Human MAb in the Presence of
Complement. The abilityof the L55-8l humanMAbto mediatelysis
of the human melanoma M14 cell line, which has a high expression of
GM2on its membrane, was evaluated using@@ Cr release assay. When
M14 cells were exposed to antibody in the presence of human or
rabbit complement, significant@ ‘Cr release activity was observed at a
range of 0.2—120 j.tg/ml antibody (Fig. 3). In the absence of comple
ment, the human MAb did not show any cytotoxic activity, even at
120 @tg/ml. An irrelevant human 1gM MAb derived from the same
patient as L55-8l MAb had no 5tCr releasing activity against M14
cells at 120 tag/mi (data not shown).
Immunohistochemical Analysis of Human MAb L55-81. Pun
fled L55-81 human MAb was biotinylated as described in “Materials
and Methods.―This method involved the labeling of the carbohydrate
moiety of immunoglobulins (22, 23). The reactivity of the biotinylated
L55-81 was examined on 9 different histological-type biopsied tumor
tissues and normal tissues derived from 24 different organ sites (2—4
tissues per organ site). The optimal concentration of the biotinylated
L55-8l was predetermined with cytospin slides of the positive control
cell line M14. The reactivity ofMl4 was + + + at 20 @ag/ml(Fig. 4A),
+ + at 10 @gIml, + at 1 @ag/ml, ± at 0.1 p.g/ml, and — with no
antibody. Subsequent experiments on biopsied tissues were performed
a Ten pg/mi of human MAb were used in the IA assay. IA titer was graded using the
following scale: >75% of cells adhered, + + + ; 50% of cells adhered, + + ; 25%of cells
adhered, + ; and —,no adherence was observed.
b Gangliosides of these cell lines were purified and analyzed as described in “Materials
and Methods.―
5667
on March 20, 2017. © 1996 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
HUMAN MAb TO GANGLIOSIDE GM2
A
I
GM3
120 24 4.8 0.96 0.192 0.038
L55-81 human MAb (j.tg/ml)
Fig. 2. A, purified gangliosides from melanoma
cell lines Ml4 and M24 run on a TLC plate and
visualized with resorcinol-HCI reagent. STD,
standard gangliosides; M14 total, total gangliosides
from Ml4 cell line; M24 total, total gangliosides
from M24 cell line. B, TLC immunostaining of the
above-mentioned gangliosides with L55-81 human
MAb. Purified °M2from Ml4 and total ganglio
sides from Ml4 and M24 were run on a TLC plate.
Twenty @sg/m1of L55-81 human MAb were used.
GM2@--
@@bf'*@GD3GDIa
GD2@:—:T_GDIbSTD.M14
totalM24 totalM14 GM2M14 totalM24 total
with 20 ;.tg/mi of the antibody. The staining results on biopsied tumor
tissues are summarized in Table 2, and representative staining is
shown in Fig. 4, B and C. Positive reactivity of L55-81 was demon
strated on various types of tumor tissues: 70% of melanoma (7 of 10),
80% of colon carcinoma (4 of 5), 80% of ovary carcinoma (4 of 5),
20% of breast carcinoma (1 of 5), 20% of kidney carcinoma (1 of 5),
and 20% of prostate carcinoma (1 of 5). Of the five specimens each
tested, positive reactivity was not seen on lung carcinoma, pancreas
carcinoma, and lymphoma. Background staining was not observed,
and noncancerous tissues were clearly negative. Of the 24 normal
tissues tested (1—4cases/organ), none showed any detectable reaction
to the antibody. These normal tissues included colon (two), lung
(two), ovary (two), cerebellum (three), breast (two), heart (four),
thyroid (four), lymph node (two), eye (three), kidney (two), liver
(four), muscle (three), skin (two), spleen (two), salivary gland (two),
spinal cord (two), pons (two), cerebrum (one), esophagus (four), testis
(two), larynx (four), stomach (two), duodenum (four), and bladder
(two). A high statistical significance (P < 0.0001), based on Fisher's
exact test, was obtained with tumor tissues of melanoma, colon
carcinoma, and ovary carcinoma. However, due to the small number
of tissues tested, prostate, breast, and kidney carcinomas did not reach
a statistically significant level (P = 0.0746; Table 2). Two of three
cultured lung carcinoma cell lines were positive in our previous
studies with the original L55 human MAb (24), but none of the five
lung carcinomas tested showed positive staining. A definitive conclu
sion of the negativity or positivity of various histological-type malig
nancies must wait until more specimens are tested.
DISCUSSION
In 1982, we reported for the first time the establishment of a human
anti-GM2 MAb using B lymphocytes from a melanoma patient (17).
The availability of human MAb enabled us to define GM2 as a human
tumor-associated molecule that could induce immune responses in
man (11, 17). Since then, three murine anti-GM2 MAbs (25—27)and
one human MAb (28) have been reported, all of which have a lesser
degree of specificity.
The human MAb L55-8l reacted specifically to GM2 [Ga1NAc(3l-
4(NeuNAca2-3)Ga131-4Glcj3l-lceramide] but not with GM1 [Galf3l-
3GalNAc@l-4(NeuNAca2-3)Gal@l-4Glc@l-lceramide], °D2 [Gal
NAc@31-4(NeuNAca2-8)(NeuNAca2-3)Galf3l-4Glc@1-lceramide],
or GM3 (NeuNAca2-3Gal@l4Glc@1-1ceramide), indicating that the
epitope recognized by L55-81 contains both GaINAc and sialic acid
bound to lactose. This study confirms the monospecificity of the
MAb. In contrast, the human MAb MAb3-207 recently developed by
Yamaguchi et a!. (28) showed affinity for both GM2 and GD2, sug
gesting a lack of specificity and difference in the epitope specificity.
One of the major goals of this study was to establish an anti-GM2
antibody-producing cell line that can grow and produce a high level of
antibody in a serum-free medium. After several recloning procedures,
the L55-81 cell line was successfully adapted to AIM-V serum-free
medium. The amount of antibody produced in the spent tissue culture
medium is unusually high (10—30 .ag/ml; 5-day culture at an initial
4
Fig. 3. Complement-dependent cytotoxic activity of L55-8l human MAb to a GM2-
positive human melanoma Ml4 cell line. All tests were done in triplicate. The percentage
of cytolysis was shown by the percentage of 51Crrelease calculated according to the
formula described in “Materialsand Methods.―U, in the presence of human complement;
D, in the presence of rabbit complement. 5668
B
@ @..:.@ _
on March 20, 2017. © 1996 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
HUMAN MAb TO GANGLIOSIDE GM2
Fig. 4. Immunohistochemical staining of M14 cell line
(A), melanoma (B), and colon carcinoma (C) with bioti
nylated L55-81 in avidin-biotin complex peroxidase
assay. Twenty pg/mi of biotinylated L55-81 were used for
staining.
, @:‘@ @—-..i_..i@ @1..
@ .,,@ @@44,•@@ @.
•,@:.. . @. .“ #@__ p'@ a
- •—@@a • ‘ .@
@@ ..@ ,@ s_-.' @M@@_F @q'
5669
A
.@‘
‘!.
- ,; ..@.—o',@ @. .@ -@ .,@@
@ ,@4;@!4;.@,!@ 0 7―4 3.@?
-.:#@ ,_ .@ @/. & @,. _;
- ‘p 1 ,@@ @a-' • _ 2'
-@@ @,
I- 1'@@@ . -@e-@'
@. .
.)
‘:@.-@
on March 20, 2017. © 1996 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
HUMAN MAb TO GANGLIOSIDE 0M2
Table 2 Reactivity of biotinylated L55-81 antibody on biopsied tumor and normal
tissues in avidin-biotin complex peroxidase assay―
Tissues tested No. positive/no. tested (%)b
Tumor tissues
Melanoma 7/10 (70%) <0.0001
Colon ca. 4/5 (80%) <0.0001
Ovary ca. 4/5 (80%) <0.0001
Prostate ca. 1/5 (20%) 0.0746
Breast ca. 1/5 (20%) 0.0746
Kidney ca. 1/5 (20%) 0.0746
Pancreas ca. 0/5 (0%) 1.0000
Lung ca. 0/5 (0%) 1.0000
Lymphoma 0/5 (0%) 1.0000
Normal tissues
Cerebellum 0/3 (0%)
Cerebrum 0/1 (0%)
Pons 0/2 (0%)
Spinal cord 0/2 (0%)
Eye 0/3 (0%)
Skin 0/2 (0%)
Muscle 0/3 (0%)
Breast 0/2 (0%)
Salivary gland 0/2 (0%)
Larynx 0/4 (0%)
Esophagus 0/4 (0%)
Stomach 0/2 (0%)
Duodenum 0/4 (0%)
Colon 0/2 (0%)
Liver 0/4 (0%)
Lung 0/2 (0%)
Heart 0/4 (0%)
Thyroid 0/4 (0%)
Spleen 0/2 (0%)
Lymph node 0/2 (0%)
Bladder 0/2 (0%)
Kidney 0/2 (0%)
Ovary 0/2 (0%)
Testis 0/2 (0%)
‘IFresh snap-frozen surgical tissues specimens were used for each immunostaining.
Twenty @sg/mlof biotinylated L55-8l were used for staining.
b Degree of reactivity for each cancer type is as follows: of the seven positive
melanomas, one + + + , four + + , one + , and one ±; of the four positive colon ca., one
+++,one ++,one +,andone ±;ofthefourpositiveovaryca.,three ++ andone +;
one positive prostate ca., + ; one positive breast ca., + + + ; one positive kidney ca., + +.
C Significance between tumor tissues versus normal tissues. P based on Fisher's exact
test. P < 0.05 is considered a significant value.
concentration of l0@cells/ml) and is equivalent to that produced by
the same cell line grown in RPM! 1640 with 5% FCS. Of the many
subclones producing anti-GM2 antibodies established in our labora
tory, this is the only cell line that adapted to a serum-free medium and,
as far as we know from literature, the only serum-free cell line
available today that produces human MAb to GM2. The antibody has
been purified to almost 100% purity with relative ease and at a
reasonably low cost.
In the present study, we investigated the potential usefulness of
L55-81 human MAb for the treatment of cancer. The reactivity of
L55-8 I human MAb to various human cancer tissues and normal
tissues was tested by direct immunohistochemical staining using
biotinylated antibody. As suggested in previous reports citing the
original L55 human MAb (17), L55-8l human MAb reacts not only to
one particular histological type of cancer but to a variety of cancer
tissues. The antibody, however, shows dramatic negativity to normal
tissues. None of the tissues obtained from 24 normal organ sites were
positive. GM2 @5a biosynthetic intermediate of the abundant brain
ganglioside@ . When gangliosides are biochemically extracted
from brain tissues, GM2 is found as a minor ganglioside (<5% of total
gangliosides; Refs. 29 and 30). Our immunological assay, however,
did not detect GM2 in either compartment of brain tissues. The
differences in the ceramide portion of the ganglioside between ma
lignant cells and normal brain tissues might be responsible for the
binding of the anti-GM2 antibody (31). Nudelman et a!. (32) reported
that ganglioside GD3, obtained from malignant melanoma, contained
a different ceramide composition from that in the brain and suggested
that the ceramide portion might influence the antibody binding to the
carbohydrate structure of the ganglioside. Dohi et a!. (33) tested
the antibody-binding of a murine-human chimeric anti-GM2 MAb
(KM966) to human normal tissues. KM966 showed positive reactivity
to glycolipids extracted from normal brain tissues, whereas an immu
nohistological staining for KM966 was negative on normal brain
tissues. A similar phenomenon has been found in an anti-GM3 anti
body binding on tissues. Hakomori et a!. (34) hypothesized that
differential reactivity between normal cells and melanoma with anti
GM3 antibody might be due to the differences in conformation and
density of GMS on these respective membranes.
When comparing the frequency of positivity among the different
cancer tissues tested, we observed some unexpected results, namely,
nonneural-origin cancer tissues such as colon and ovarian carcinomas
that synthesized gangliosides at the lower level showed positive
reactivity to L55-81 as frequently and strongly as the neuroectoder
mal-origin cancer melanoma. Even among melanoma cell lines, the
cellular content of GM2 measured by biochemical assays did not
always reflect the binding of L55-81 to the cell surface. Experimental
results showed that M24, which has high GM2 content (8.2 nmol/g dry
weight), had lower cell surface reactivity with L55-81 than Ml4 (4.0
nmol/g dry weight; Table 1). There are three possible explanations:
(a) antigenic molecules detected by L55-8l on carcinoma cells are not
GM2but simplycross-reactwith GM2.In this regard,Patel et al. (35)
reported that a murine anti-GD2 MAb, MAb3F8, cross-reacted with
the glycoprotein neural cell adhesion molecule and that both GD2 and
the neural cell adhesion molecule have NeuAccs2-8NeuAc terminal
residue; (b) the carbohydrate moiety of GM2 is relatively short as
compared to that of many other gangliosides or glycoproteins. The
reactivity of L55-81 may have been interfered with partially by these
longer cell surface molecules in certain melanoma tissues; and (c)
gangliosides are normally located not only on cell membranes but also
in the Golgi apparatus, where they are biosynthesized (36). It is
possible that a larger amount of GM2 in the Golgi apparatus reflected
the total GM2 content rather than the amount of GM2 on the cell
surface membranes.
Since we reported GM2 as a target for cancer immunotherapy, it has
been tested in clinical Phase I/I! trials in the form of a pure molecule
or GM2-rich melanoma cell vaccine (37, 38). Patients who received
these vaccines in active specific immunotherapy produce 1gM anti
bodies to GM2, the levels of which correlated to good prognosis.
Recently, the Eastern Cooperative Oncological Group has initiated a
double-blind randomized trial specifically targeting GM2 in melanoma
patients with AJCC stage II or III melanoma with purified GM2
conjugated to adjuvants. The therapeutic potential of anti-GM2 anti
body has also been suggested in our preliminary experiments, which
used the original L55 human MAb. A patient with G@2-positive
cutaneous melanoma was treated with the human MAb via intrale
sional injection (39). A complete regression of the melanoma was
observed with 3.3 mg of the antibody. In the same experiment,
treatment with the antibody was ineffective in another patient with
G@2-negative melanoma, confirming immunological specificity of the
treatment efficacy by the antibody. The study has since been discon
tinued due to the high cost of clinical grade L55 human MAb. Due to
the high secretion rate of antibody by the L55-8l cell line and the
adaptation of the cells in a serum-free medium, which has reduced the
purification process, we are able to reduce the cost of production for
purified L55-8l significantly. To conclude, the new human MAb to
GM2 secreted from the highly stable cell line L55-8l has strong
antitumor activity in the presence of complement and may be one of
the most potent MAbs for the treatment of cancer.
5670
on March 20, 2017. © 1996 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
HUMAN MAb TO GANGLIOSIDE G@
ganglioside GM3 antigen on human cancers. Cancer Rca., 53: 5244—5250, 1993.
19. Tai, T., Cahan, L. D., Paulson, J. C., Saxton, R. E., and Irie R. F. Human monoclonal
antibody against ganglioside 002: use in development of enzyme-linked immunosor
bent assay for the monitoring of anti-G@2in cancer patients. J. NatI. Cancer Inst., 73:
627—633,1984.
20. Ravindranath, M. H., Ravindranath, R. M. H., Morton, D. L., and Graves, M. C.
Factors affecting the fine specificity and sensitivity of serum antiganglioside anti
bodies in ELISA. J. Immunol. Methods, 169: 257—272,1994.
21. Iwamori, M., Sunada, S., lshihara, E., Moki, M., Fujimoto, S., and Nagai, Y.
Differential expression of fucosyl GM, and a disialoganglioside with a Neu-Aca2-
6GaINAc linkage (GDI,) in various rat ascites hepatoma cells. FEBS Lett., 198:
66—70,1986.
22. O'Shannessy, D. J., and Quarles, R. H. Specific conjugation reactions of the oligo
saccharide moieties of immunoglobulins. J. AppI. Biochem.. 7: 347—355,1985.
23. O'Shannessy, D. J., and Quarles, R. H. Labeling of the oligosaccharide moieties of
immunoglobulins. J. Immunol. Methods, 99: 153—161,1987.
24. Inc. R. F., Tai, T., and Morton, D. L. Antibodies to tumor-associated gangliosides
(GM2 and GD2): potential for suppression of melanoma occurrence. in: M. Torisu and
T. Yoshida (eds.), Basic Mechanisms and Clinical Treatment of Tumor Metastases,
pp. 371—384.San Diego, CA: Academic Press, 1984.
25. Natoli, E. J., Livingston, P. 0., Pukel, C. S., Lloyd, K. 0., Wiegandt, H., Szalay, 3.,
Oettgen, H. F., and Old, L. 3. A murine monoclonal antibody detecting N-acetyl- and
N-glycolyl-G@2: characterization of cell surface reactivity. Cancer Res., 46: 41 16—
4120, 1986.
26. Bjerkvig, R., Engebraaten, 0., Laerum, 0. D., Fredman, P., Svennerholm, L.. Vrionis.
F. D., Wikstrand, C. J., and Bigner, D. D. Anti-GM2monoclonal antibodies induce
necrosis in GM2-rich cultures of a human glioma cell line. Cancer Res., 51: 4643—
4648,1991.
27. Miyake, M., Ito, M., Hitomi, S., Ikeda, S., Taxi, T., Kurata, M., Hino, A., Miyake, N.,
and Kannagi, R. Generation of two murine monoclonal antibodies that can discrim
mate N-glycolyl and N-acetyl neuraminic acid residues of °M2gangliosides. Cancer
Res., 48: 6154—6160,1988.
28. Yamaguchi, H., Furukawa, K., Fortunato, S. R., Livingston. P. 0., Lloyd. K. 0.,
Oettgen, H. F., and Old, L. 3. Human monoclonal antibody with dual GM2/GD2
specificity derived from an immunized melanoma patient. Proc. NaIl. Acad. Sci.
USA, 87: 3333—3337,1990.
29. Ando, S., Chang, N-C., and Yu, R. K. High-performance thin-layer chromatography
and densitometric determination of brain ganglioside compositions of several species.
Anal. Biochem., 89: 437—450,1978.
30. Nagai, Y., and Iwamori, M. Ganglioside distribution at different levels of organiza
tion and its biological implications. In: R. W. Ledeen, R. K. Yu, M. M. Rapport, and
K. Suzuki (eds.), Ganglioside Structure, Function, and Biomedical Potential, Vol.
174, pp. 135—146.New York: Plenum Press, 1984.
31. Itonori, S., Hidari, K., Sanai, Y., Taniguchi, M., and Nagai, Y. Involvement of the
acyl chain of ceramide in carbohydrate recognition by an anti-glycolipid monoclonal
antibody: the case of an anti-melanoma antibody, M259O, to G@3-ganglioside.
Glycoconj. 3., 6: 551—560,1989.
32. Nudelman, E., Hakomori, S., Kannagi, R., Levery, S., Yeh, M. Y., Hellstrom, K. E.,
and Hellstrom, I. Characterization of a human melanoma-associated ganglioside
antigen defined by a monoclonal antibody, 4.2. 3. Biol. Chem., 257: 12752—12756,
1982.
33. Dohi, T., Nakamura, K., Hanai, N., Taomoto, K., and Oshima. M. Reactivity of a
mouse/human chimeric anti-GM2antibody KM966 with brain tumors. Anticancer
Rca., 14: 2577—2582,1994.
34. Hakomori, S. General concept of tumor-associated carbohydrate antigens: their chem
ical, physical, and enzymatic basis. In: H. F. Oettgen (ed). Gangliosides and Cancer,
pp.57—67.NewYork:VCHPublishers,1989.
35. Patel, K., Rossell, R. 3., Pemberton, L. F., Cheung, N. K., Walsh, F. S., Moore, S. E.,
Sugimoto, T., and Kemshead, 3. T. Monoclonal antibody 3F8 recognizes the neural
cell adhesion molecule (NCAM) in addition to the ganglioside G02. Br. 3. Cancer, 60:
861—866,1989.
36. Teuamanti, G. An ouiline of ganglioside metabolism. in: R. W. Ledeen, R. K. Yu, M.
M. Rapport. and K. Suzuki (eds.), Ganglioside Structure, Function, and Biomedical
potential, Vol. 174, pp. 197—211. New York: Plenum Press, 1984.
37. Morton, D. L., Foshag, J., Hoon, D. S. B., Nizze, J. A., Wanek, L. A., Chang, C.,
Davtyan, D. G., Gupta, R. K., Elashoff, R.,and Irie, R. F. Prolongation of survival in
metastatic melanoma after active specific immunotherapy with a new polyvalent
melanoma vaccine. Ann. Surg., 216: 463—482,1992.
38. Livingston, P. 0. Approaches to augmenting the immunogenicity of melanoma
gangliosides: from whole melanoma cells to ganglioside-KLH conjugate vaccines.
Immunol. Rev., 145: 147—166,1995.
39. Inc. R. F., Matsuki, T., and Morton, D. L. Human monoclonal antibody to ganglioside
GM2 for melanoma treatment. Lancet, 8641: 786—788, 1989.
5671
ACKNOWLEDGMENTS
We thank the staff of Immunosciences,Inc. (New York, NY) for their
technical and pathological expertise in immunohistochemicalstudies. We also
thank Lan Sze for her technical assistance and Christina Riley for her editorial
assistance.
REFERENCES
1. Tai, T., Paulson, J. C., Cahan, L. D., and Inc. R. F. Ganglioside GM2as a human
tumor antigen (OFA-I-l). Proc. NatI. Aced. Sci. USA, 80: 5392—5396,1983.
2. Tsuchida, T., Saxton, R. E., Morton, D. L., and inc. R. F. Gangliosides of human
melanoma. J. Natl. Cancer Inst., 78: 45—54,1987.
3. Hamilton,W. B., Helling,F., Lloyd, K. 0., and Livingston,P. 0. Ganglioside
expression on human malignant melanoma assessed by quantitative immune thin
layer chromatography. Int. J. Cancer, 53: 566—573,1993.
4. Higashi, H., Hirabayashi, Y., Hirota, M., Matsumoto, M., and Kato, S. Detection of
ganglioside GM2in sera and tumor tissues of hepatoma patients. Jpn. J. Cancer Rca.,
78:1309—1313,1987.
5. Inc. R. F., and Ravindranath, M. H. Gangliosides as targets for monoclonal antibody
therapy of cancer. In: C. Borrebaech and J. Larrick (eds.), Therapeutic Monoclonal
Antibody, pp. 75—94.New York: Stockton Press, 1990.
6. Inc. R. F., and Morton, D. L. Regression of cutaneous metastatic melanoma by
intralesional injection with human monoclonal antibody to ganglioside GD2. Proc.
Natl. Acad. Sci. USA, 83: 8694—8698,1986.
7. Houghton, A. N., Mintzer, D., Cordon-Cardo, C., Welt, S., Fliegel, B., Vadhan, S.,
Carswell, E., Melamed, M. R., Oeugen, H. F., and Old, L. J. Mouse monoclonal IgG3
antibody detecting GD3 ganglioside: a Phase I trial in patients with malignant
melanoma. Proc. Nail. Acad. Sci. USA, 82: 1242—1246,1985.
8. Vadhan-Raj, S., Cordon-Cardo, C., Carswell, E., Mintzer, D., Dantis, L., Duteau, C.,
Templeton, M. A., Oettgen, H. F., Old, L. J., and Houghton, A. N. Phase I trial of a
mousemonoclonalantibodyagainstGD3gangliosidein patientswith melanoma:
inductionof inflammatoryresponsesat tumorsites.J. Clin.Oncol.,6: 1636—1648,
1988.
9. Bajorin, D. F., Chapman, P. B., Wong, G., Coit, D. G., Kunicka, J., Dimaggio, J.,
Cordon-Cardo, C., Urmacher, C., Dantes, L., Templeton, M. A., Liu, J., Oettgen,
H. F., and Houghton, A. N. Phase I evaluation of a combination of monoclonal
antibody R24 and interleukin 2 in patients with metastatic melanoma. Cancer Res.,
50: 7490—7495,1990.
10. Cheung, N. K., Burch, L., Kushner, B. H., and Munn, D. H. Monoclonal antibody 3F8
can effect durable remissions in neuroblastoma patients refractory to chemotherapy:
a Phase II trial. Prog. Clin. Biol. Res., 366: 395—400,1991.
11. Saleh, M. N., Khazaeli, M. B., Wheeler, R. H., Dropcho, E., Liu, T., Urist, M., Miller,
D. M., Lawson, S., Dixon, P., Russel, C. H.,and LoBuglio, A. F. Phase I trial of the
murine monoclonal anti-GD2 antibody l4G2a in metastatic melanoma. Cancer Res.,
52: 4342—4347, 1992.
12. Saleh, M. N., Khazaeli, M. B., Wheeler, R. H., Allen, L., Tilden, A. B., Grizzle, W.,
Reisfeld, R. A., Yu, A. L., Gillies, S. D., and LoBuglio, A. F. Phase I trial of the
chimeric anti-G02 monoclonal antibody chl4.l8 in patients with malignant mela
noma. Hum. Antib. Hybrid., 3: 19—24,1992.
13. Handgretinger, R., Bander, P., Dopfer, R., Klingebiel, T., Reuland, P., Treuner, J.,
Reisfeld, R. A., and Niethammer, D. A Phase I study of neuroblastoma with the
anti-ganglioside GD2 antibody l4G2a. Cancer Immunol. Immunother., 35: 199—204,
1992.
14. Murray, J. L., Cunningham, J. E., Brewer, H., Mujoo, K., Zukiwski, A. A., Podoloff,
D. A., Kasi, L. P., Bhadkamkar, V., Fritsche, H. A., Benjamin, R. S., Legha, S. S.,
Ater, J. L., Jaffe, N., Itoh, K., Ross, M. I., Bucana, C. D., Thompson, L., Cheung, L.,
and Rosenblum, M. G. Phase I trial of murmnemonoclonal antibody l4G2a admin
istered by prolonged intravenous infusion in patients with neuroectodermal tumors.
J. Clin. Oncol., 12: 184—193,1994.
15. Handgretinger, R., Anderson, K., Lang, P., Dopfer, R., Klingebiel, T., Schrappe, M.,
Reuland, P., Gillies, S. D., Reisfeld, R. A., and Niethammer, D. A Phase I study of
human/mouse chimeric anti-ganglioside GD2 antibody chl4. 18 in patients with neu
roblastoma. Eur. J. Cancer, 31: 261—267,1995.
16. Cahan, L. D., Inc. R. F., Singh, R., Cassidenti, A., and Paulson, J. C. Identification
ofahumanneuroectodennaltumorantigen(OFA-I-2)asgangliosideG02.Proc.Nail.
Acad. Sci. USA, 79: 7629—7633,1982.
17. inc. R. F., Sze, L. L., and Saxton, R. E. Human antibody to OFA-I, a tumor antigen
produced in vitro by Epstein-Barr virus-transformed human B-lymphoid cell lines.
Proc. NatI. Acad. Sci. USA, 79: 5666—5670,1982.
18. Hoon,D. S.B.,Wang,Y.,Sze,L.,Kanda,H.,Watanabe,T.,Morrison,S. L.,Morton,
D. L., and inc. R. F. Molecular cloning of a human monoclonal antibody reactive to
on March 20, 2017. © 1996 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
1996;56:5666-5671. Cancer Res
Yumiko Nishinaka, Mepur H. Ravindranath and Reiko F. Irie
with Potential for Cancer Treatment
M2
G
Development of a Human Monoclonal Antibody to Ganglioside
Updated version
http://cancerres.aacrjournals.org/content/56/24/5666
Access the most recent version of this article at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
.permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
on March 20, 2017. © 1996 American Association for Cancer Research.cancerres.aacrjournals.org Downloaded from
... 12,13 Gangliosides are abundant in nervous tissue where GM1, GD1a, GD1b, GT1b, and GQ1b account for most of them, while other gangliosides including GM2 are minor components in normal nervous systems. 12,13 Interestingly, GM2 is overexpressed in various cancer tissues including lung cancer, colon cancer, ovarian cancer, and malignant pleural mesothelioma, [14][15][16][17] suggesting that GM2 can be a potential target for cancer immunotherapy. Monoclonal antibody (mAb) against GM2 with enhanced activity of antibody-dependent cellular cytotoxicity (ADCC) was developed and examined in Phase I/II clinical trials, which resulted in insufficient efficacy while the safety was confirmed. ...
... GM2 is overexpressed in various cancer cells, while detected as a minor component in normal tissues including nervous and nonnervous cells. [12][13][14][15][16][17] The upregulated expression of GM2 in tumor cells has been shown to correlate with their malignant phenotypes, such as invasiveness of radiation-tolerant lung cancer cells and resistance to chemotherapeutic drugs. 23,24 It was also reported that GM2 on tumor cells can mediate immunosuppressive effects through T cell dysfunction and apoptosis. ...
Article
Full-text available
Background: While chimeric antigen receptor (CAR)-T cell therapy has demonstrated excellent efficacy in hematopoietic malignancies, its clinical application in solid cancers has yet to be achieved. One of the reasons for such hurdle is a lack of suitable CAR targets in solid cancers. Methods: GM2 is one of the gangliosides, a group of glycosphingolipids with sialic acid in the glycan, and overexpressed in various types of solid cancers. In this study, by using interleukin (IL)-7 and chemokine (C-C motif) ligand 19 (CCL19)-producing human CAR-T system which we previously developed, a possibility of GM2 as a solid tumor target for CAR-T cell therapy was explored in a mouse model with human small-cell lung cancer. Results: Treatment with anti-GM2 IL-7/CCL19-producing CAR-T cells induced complete tumor regression along with an abundant T cell infiltration into the solid tumor tissue and long-term memory responses, without any detectable adverse events. In addition, as measures to control cytokine-release syndrome and neurotoxicity which could occur in association with clinical use of CAR-T cells, we incorporated Herpes simplex virus-thymidine kinase (HSV-TK), a suicide system to trigger apoptosis by administration of ganciclovir (GCV). HSV-TK-expressing anti-GM2 IL-7/CCL19-producing human CAR-T cells were efficiently eliminated by GCV administration in vivo. Conclusions: Our study revealed the promising therapeutic efficacy of anti-GM2 IL-7/CCL19-producing human CAR-T cells with an enhanced safety for clinical application in the treatment of patients with GM2-positive solid cancers.
... Among all TACAs identified so far, GM2 is especially attractive for cancer vaccine development 39 because: (a) GM2 is relatively cancer-specific and expressed by several types of tumors, including melanoma, sarcoma, and renal cancer; 40 (b) GM2-reactive antibodies have been shown to mediate cytotoxicities against GM2-positive human cancer cell lines in vitro; 41-44 (c) GM2 is comparatively immunogenic, as evidenced by the presence of low titers of natural anti-GM2 IgM antibodies in some cancer patients [44][45][46] and by the findings that immunization of Published: xx xx xxxx OPEN www.nature.com/scientificreports/ 2 Scientific RepoRtS | 7: 11403 | melanoma patients with GM2 conjugates could elicit anti-GM2 antibodies; 43, 44 (d) the presence of GM2 antibodies in melanoma patients appears to be associated with improved survival and longer disease-free interval; 44 (e) no deleterious side effects associated with immune responses to GM2 have been observed yet 44 . As a result, GM2 synthesis and GM2-based cancer vaccines have been explored extensively by several groups 47-51 . ...
Article
Full-text available
An efficient method was developed for the synthesis of a GM2 derivative suitable for the conjugation with various biomolecules. This GM2 derivative was covalently linked to keyhole limpet hemocyanin (KLH) and monophosphoryl lipid A (MPLA) to form novel therapeutic cancer vaccines. Immunological evaluations of the resultant conjugates in mice revealed that they elicited robust GM2-specific overall and IgG antibody responses. Moreover, the GM2-MPLA conjugate was disclosed to elicit strong immune responses without the use of an adjuvant, proving its self-adjuvant property. The antisera of both conjugates showed strong binding and mediated similarly effective complement-dependent cytotoxicity to GM2-expressing cancer cell line MCF-7. Based on these results, it was concluded that both GM2-MPLA and GM2-KLH are promising candidates as therapeutic cancer vaccines, whereas fully synthetic GM2-MPLA, which has homogeneous and well-defined structure and self-adjuvant property, deserves more attention and studies.
... The level of anti-GM 2 IgM can be augmented by passive administration of a murine anti-GM 2 IgM such as KM696; however, the development of anti-mouse antibody responses 45 limits the efficacy of repeated administration. Nishinaka et al. 46 developed a human anti-GM 2 monoclonal antibody that can cause complement-dependent cytotoxicity, but its production is not cost-effective. Nakamura et al. 44,47 constructed mouse/human chimeric anti-GM 2 IgM KM966 (mouse residue 33%; human residue 67%) and humanized chimeric anti-GM 2 IgM KM8969 (mouse residue 10%; human residue 90%) with therapeutically higher complement-mediated cytotoxicity, antibodydependent cytotoxicity, and apoptosis. ...
Article
Full-text available
Pancreatic adenocarcinoma cells express gangliosides and sialyl Lewis (sLe) antigens. It is not known whether these carbohydrate antigens can be targeted by immunotherapy. The authors measured the expression of GM2 and sLe antigens on the surface of pancreatic carcinoma cells and the serum levels of total gangliosides, GM2, and antiganglioside antibodies in patients with pancreatic carcinoma. Cell surface GM2 and sLe antigens were measured by cell suspension enzyme linked immunoadsorbent assay (ELISA) in four pancreatic carcinoma cell lines. Sera from 20 pancreatic carcinoma patients and 20 age- and gender-matched healthy volunteers were analyzed for antiganglioside and anti-sLe immunoglobulin (Ig) M titers by ELISA. Serum levels of total gangliosides and GM2 also were measured. All cell lines expressed GM2 and sLe antigens. When compared with age- and gender-matched volunteers, patients had significantly higher serum levels of total gangliosides (25.6 ± 9.0 mg/dL vs. 15.6 ± 2.7 mg/dL; P < 0.001), GM2 (0.278 ± 0.415 mg/dL vs. 0.013 ± 0.018 mg/dL; P = 0.02), ELISA units of anti-GM2 IgM antibody (368 ± 95 vs. 155 ± 25; P = 0.04) and anti-GD1b IgM antibody (351 ± 91 vs. 138 ± 26; P = 0.03), but not anti-sLex IgM (1389 ± 345 vs. 1081 ± 224; P = 0.46) or anti-sLea IgM antibody (1097 ± 253 vs. 1200 ± 315; P = 0.80). Patients with unresectable tumors had higher serum levels of total gangliosides compared with patients with resectable tumors, and a serum level > 25 mg/dL was found to correlate significantly with poor overall survival (P < 0.02). Increased serum levels of total gangliosides and GM2 may reflect shedding or release of gangliosides from the surface of tumor cells. Production of IgM antibody against GM2 and GD1b indicates that these gangliosides are immunogenic antigens that may be potential targets for effective active immunotherapy. Cancer 2000;88:1828–36.
Article
Sialic acids (SAs) are nine-carbon monosaccharides existing at the terminal location of glycan structures on the cell surface and secreted glycoconjugates. The expression levels and linkages of SAs on cells and tissues, collectively known as sialoform, present the hallmark of the cells and tissues of different systems and conditions. Accordingly, detecting or profiling cell surface sialoforms is very critical for understanding the function of cell surface glycans and glycoconjugates and even the molecular mechanisms of their underlying biological processes. Further, it may provide therapeutic and diagnostic applications for different diseases. In the past decades, several kinds of SA-specific binding molecules have been developed for detecting and profiling specific sialoforms of cells and tissues; the experimental materials have expanded from frozen tissue to living cells; and the analytical technologies have advanced from histochemistry to fluorescent imaging, flow cytometry and microarrays. This review summarizes the recent bioaffinity approaches for directly detecting and profiling specific SAs or sialylglycans, and their modifications of different cells and tissues.
Chapter
Carbohydrate cell-surface antigens have proved to be unexpectedly potent targets for immune recognition and attack against cancers (reviewed in 1). Of the many tumorrestricted monoclonal antibodies derived by immunization of mice with human tumor cells, most have been directed against carbohydrate antigens expressed at the cell surface as glycolipids or mucins (2–4). Antibodies against cell-surface antigens such as these are ideally suited for eradication of free tumor cells and micrometastases. This is the role of antibodies against most infectious diseases and it has been accomplished against canc6 r cells as described below in a variety of preclinical models. In adjuvant immunization trials, the primary targets are individual tumor cells or early micrometastases, which may persist for long periods after apparent resection of all residual tumor (5–7). After surgery and completion of chemotherapy is the ideal time for immune intervention, and in particular for administration of cancer vaccines aimed at instructing the immune system to identify and kill these few remaining cancer cells. If antibodies of sufficient titer can be induced against tumor antigens to eliminate tumor cells from the blood and lymphatic systems, and to eradicate micrometastases (making establishment of new metastases no longer possible) this would dramatically change our approach to treating the cancer patient. Aggressive local therapies, including surgery, radiation therapy, and intralesional treatments, might result in long-term control of even metastatic cancers.
Article
In the last decade, several trials have been carried out to answer some questions. The first objective has been reproducing the results published initially by Kirkwood with high-dose interferon and testing them with longer follow-up. Furthermore, there have been tested another interferon schedules in order to find effective therapies with less toxicity and, finally, there have been initiated specific immunotherapy against melanoma targets, like certain gangliosides of cell membrane. Interferon, with a certain dose-dependent effect, has shown that offers improvement in relapse-free survival, but not in global survival, with a significant toxicity cost. Therefore, most of authors suggest that patients with high-risk melanomas should enter on clinical trials, if possible. If not, they recommend an adjuvant treatment with high-dose interferon, once advised its toxicity profile and the probability of effectiveness according to the results in the literature.
Article
BACKGROUND: Because of the challenge in defining prognostic markers predictive of recurrence or progression, carcinoembryonic antigen (CEA) remains the most frequently used marker in colorectal cancer, despite its low sensitivity. We hypothesized that TA90-IC status and serum ganglioside levels might be useful markers and might be of prognostic significance in colorectal cancer. METHODS: Serum samples from 68 patients undergoing surgical treatment for histologically proven colorectal cancer were analyzed for the presence of CEA, serum gangliosides, and TA90-IC. Forty-one patients had node-negative disease, whereas 27 patients had limited metastatic disease. The intent was curative resection, even for patients with metastatic disease. Cryopreserved serum specimens were analyzed in a blinded fashion for total serum ganglioside levels (by an assay that detects lipid-associated sialic acids), for CEA, and for TA90-IC (by a murine monoclonal antibody-based enzyme-linked immunosorbent assay). A positive value for TA90-IC levels was defined as an optical density (OD) of more than 0.410 at 405 nm. RESULTS: Serum ganglioside levels were elevated more frequently than CEA concentrations (84% vs 44%). The combination of serum ganglioside and CEA values was more sensitive (88%) than CEA value alone (44%) in identifying patients with early-stage colorectal cancer. TA90-IC levels were elevated more frequently than CEA concentrations (56% vs 32%). The combination of TA90-IC and CEA values was more sensitive (72%) than CEA value alone (32%) in identifying patients with advanced-stage colorectal cancer. At an enzyme-linked immunosorbent assay cutoff level of 0.410, 15 (56%) patients had positive TA90-IC values. Fourteen patients alive with residual disease had a median OD TA90-IC level of 0.879, and only three patients had levels below the OD cutoff value of 0.410. Thirteen patients with no evidence of disease had a median level of 0.277, and only four patients had OD levels > 0.410. TA90-IC was significantly higher in the alive with residual disease patients than those rendered no evidence of disease (P = 0.02). CONCLUSIONS: We speculate that a multiple-marker analysis that combines CEA values with serum ganglioside and TA90-IC values may be more sensitive than CEA value alone for detecting colorectal cancer. The potential prognostic significance of TA90-IC status in advanced disease warrants further investigation.
Article
This chapter describes the thin-layer chromatography (TLC) and high-performance liquid chromatography (HPLC) of glycosphingolipids (GSLs). Since their discovery, GSL structures in large numbers have been recorded, and the qualitative and quantitative GSL content have been determined for many organs and tissues in vertebrates. Because GSLs do not possess a characteristic chromophore, several precolumn derivatization reactions are developed. Preparative HPLC is superior to preparative TLC when large quantities of GSLs are required. Contrary to the simple concept of the fluid mosaic model of membrane structures, GSLs are not merely arbitrary head groups with dangling fatty acyl chains that provide passive structural support for membrane proteins. GSL-mediated mechanisms of action converge on two general schemes: (1) the interactions of GSLs with complementary receptors in opposing membranes (trans recognition) and (2) the modulation of activities of proteins in the same membrane (cis regulation). The chapter discusses some fundamental procedures for the successful isolation and structural characterization of GSLs.
Article
Ein Zielantigen für die Immuntherapie gegen Krebs ist das Gangliosid GM2, das auf den Zelloberflächen einiger humaner Krebsarten exprimiert wird. Durch eine effiziente Totalsynthese ist nun einheitliches und von biologischen Verunreinigungen freies GM2 auch in größerer Menge zugänglich. Damit ist der systematische Aufbau eines sicheren und wirksamen Impfstoffes gegen GM2 ⁺ ‐Krebsarten beträchtlich erleichtert. magnified image
Article
Full-text available
The effects of four anti-GM2 monoclonal antibodies (DMAb-1, DMAb-2, DMAb-3, and DMAb-5) were studied on spheroid cultures from a human glioma cell line (D-54 MG) that is known to express high levels of GM2. The spheroids developed central necrosis 48 h after antibody exposures at concentrations greater than 6 micrograms/ml. No necrosis was found with antibodies that had been absorbed with GM2 prior to exposure or with unrelated cytotoxic antibodies. Immunohistochemistry showed that the necrosis started shortly after the antibodies were evenly distributed throughout the spheroids. Light and transmission electron microscopy revealed that a small portion of the cells, mainly in the periphery of the spheroids, was unaffected by antibody exposure. New monolayer cultures established from antibody-treated cells expressed a 50% lower GM2 content as shown by flow cytometry and determination of ganglioside content throughout at least 12 passages. Thus, the GM2-rich D-54 MG cell line has subpopulations of cells with lower GM2 content. Spheroids obtained from this subpopulation developed only minor necrosis after antibody treatment. These results show that GM2 antibodies cause severe necrosis of GM2-containing glioma cells in vitro, but the effect depends on the concentration of antigen, and a threshold number of GM2 molecules is required.
Article
Human melanoma synthesizes a large quantity of gangliosides, glycosphingolipids containing sialic acid. The authors previously have demonstrated that the ganglioside profile differs among individual melanomas and is widely heterogenous. In the current study, a retrospective study was performed to compare the relationship between the quantity of five major gangliosides of human melanoma (GM3, GM2, GD3, GD2, and alkali-labile ganglioside) and nine clinical factors (sex, age, site, stage, tumor size, pigmentation, histopathologic type of primary tumor, chemosensitivity, and prognosis). Melanoma specimens studied were obtained from patients of our clinic and included 52 biopsy specimens and 28 cultured cell lines. Analysis of melanoma biopsy specimens have shown a differential ganglioside expression among different sites of tumor, pigmentation, and histopathologic types. Results of cultured melanoma cell lines differed from those of biopsy specimens, but ganglioside expression also differed among the site of tumor, tumor size, histopathologic types, and chemosensitivity. GM3 positively correlated with a good prognosis in both biopsy and cultured melanomas.
Article
The ganglioside composition of human malignant melanoma was studied with the use of 80 melanoma specimens, including 52 surgical specimens and 28 cultured cell lines. A ganglioside fraction was isolated and purified from each of these tissues, and the amount of each component ganglioside was assessed by thin-layer chromatography (TLC) and a TLC scanner. Five gangliosides (GM3, GD3, GM2, GD2, and alkali-labile ganglioside) were most commonly expressed by these melanomas. However, the total ganglioside amount (ranging from 33 to 302 μg/g wet wt of tissue) as well as the distribution of each ganglioside were widely heterogeneous in both biopsied and cultured melanomas. When the ganglioside expressions of cultured and biopsied melanomas were compared, GM2 and GD2 were minor components of biopsied melanomas but often became major components of cultured melanoma cells. Conversely, alkali-labile ganglioside was expressed more strongly on biopsied melanomas. This heterogeneity suggests that it will be necessary to analyze the ganglioside composition of biopsied melanomas before using monoclonal antibodies to melanoma-associated gangliosides for melanoma diagnosis or therapy.
Chapter
Certain classes of antibodies are capable of lysing tumor cells by either activating complement (complement-dependent cytotoxicity, or CDC) or by mediating immune effector cells (antibody-dependent cellular cytotoxicity, or ADCC). Non-cytotoxic antibodies are also capable of cell lysis after coupling with toxins, chemotherapeutic drugs, or radioisotopes. In addition, there are antibodies capable of blocking cell surface receptors, thereby inhibiting growth, metastasis, and invasion of the tumor cells. In view of these anti-tumor capabilities, a number of clinical trials involving monoclonal antibodies (Mab) targeted against human cancer have been performed. With the technical feasibility to produce large quantities of purified monoclonal antibodies with defined specificity, isotype, and function, monoclonal antibodies have generated much hope for the immunotherapy of human cancer.
Article
Improved resolution of complex brain ganglioside mixtures was achieved by high-performance thin-layer chromatography. The percentage distribution of individual gangliosides was then determined by direct densitometric seanning, employing a transmittance mode, of the resorcinol-positive spots on the plate. As little as 90 pmol (29 ng) of lipid-bound sialic acid could be detected with a good signal-to-noise ratio. A linear detector response was observed up to 3.0 μg of lipid-bound sialic acid. The brain white matter ganglioside patterns of eight animal species, including human, chimpanzee, monkey, chicken, bovine, sheep, and pig, were examined in detail. In addition, human brain gray matter, rat cerebral, rat brain gray matter, and rat cerebellar ganglioside patterns were also studied. Ganglioside GM4 (G7) was found to be one of the major components in primate and chicken brain white matter, but it represented only a minor ganglioside in other species. Other major gangliosides in all brain samples studied were GM1, GD1a, GD1b, and GT1b. GM1 was more abundant in white matter than in gray matter. GT1a, a recently discovered ganglioside species, was found in all species examined, but was most abundant in the rat cerebellum. The latter source also contained high proportions of GT1b and GQ1b.
Article
A new polyvalent melanoma cell vaccine (MCV) was administered to 136 stage IIIA and IV (American Joint Committee on Cancer) melanoma patients. Induction of cell-mediated and humoral immune responses to common melanoma-associated antigens present on autologous melanoma cells was observed in patients receiving the new MCV. This was accompanied by increased activation of tumor-infiltrating lymphocytes. Survival correlated significantly with delayed cutaneous hypersensitivity (p = 0.0066) and antibody responses to MCV (p = 0.0117). Of 40 patients with evaluable disease, nine (23%) had regressions (three complete). From our historical database of 126 stage IIIA and 1275 stage IV melanoma patients, there were no significant changes in the natural history of metastatic melanoma during the past 20 years. Univariate and multivariate analyses demonstrated prognostic significance for site of metastases (p = 0.0001) and immunotherapy with the new MCV (p = 0.0001). Overall our new MCV increased the median and 5-year survival of stage IIIA melanoma patients with regional soft tissue metastases twofold (p = 0.00024), and stage IV patients threefold (p = 0.0001) compared with previous immunotherapy and other treatments.
Article
The chimeric monoclonal anti-GD2 antibody ch14.18 is made up of the variable region of the murine anti-GD2 antibody 14.18 (or its IgG2a switch variant 14G2a) and the constant region of human IgG1k. Ch14.18 mediates antibody dependent cytotoxicity and complement dependent lysis in vitro. In a phase I trial, 13 patients with metastatic melanoma received ch14.18 as a single dose of 5-100 mg. Therapy was associated with an infusion-related abdominal/pelvic pain syndrome, which required intravenous morphine for control. The pharmacokinetics of ch14.18 best fit a two-compartment model with a T1/2 alpha of 24 +/- 1 hr and a T1/2 beta of 181 +/- 73 hr. Eight of 13 patients developed a weak-modest antibody response directed at the variable region of ch14.18. Clinical antitumor responses were not observed at the doses employed in this study. However, patients receiving greater than 45 mg of ch14.18 had antibody detectable on tumor cells analyzed by fluorescent activated cell sorter. Further modification of the therapeutic regime employing larger doses and frequent administration of ch14.18 are planned.
Article
Nine patients with neuroblastoma stage IV were treated with the murine monoclonal antibody 14.G2a, directed against disialoganglioside GD2. The antibody was injected daily for 5–10 days and the total applied dosage ranged between 100 mg/m2 and 400 mg/m2. The peak serum levels of mAb 14.G2a ranged from 28 µg/ml to 61 µg/ml. Pharmacokinetic data obtained in three patients indicated that the serum elimination of mAb 14.G2a fits a two-compartment model, with an α-half-time (t 1/2α ) between 0.66 h and 1.98 h and a β-half-time (t 1/2β ) between 30.13 h and 53.33 h. All patients presented with a human anti-(mouse IgG) antibody response either during or shortly after therapy. Eight patients showed a continuous decrease in complement component C4 during therapy, as well as an initial decrease in C3c and an initial increase in C3a, all suggesting an activation of the complement cascade. Side-effects consisted of allergic reactions like pruritus, exanthema, urticaria and of severe pain, predominantly located in the abdomen and lower extremities, which required the use of continuous intravenous morphine. Four patients additionally developed a transient hypertension and one patient experienced a transient nephrotic syndrome. Three patients were treated in an adjuvant setting and are not evaluable for tumor response. Of the remaining six patients, two had a complete remission, two showed a partial remission, and two patients did not respond to treatment.
Article
In a phase I trial, 12 patients with GD2 antigen-positive metastatic melanoma received the murine anti-GD2 monoclonal antibody 14G2a. The monoclonal antibody was administered in four doses over an 8-day period with total dose ranging from 10 to 120 mg. All patients receiving greater than 10 mg of 14G2a experienced transient abdominal/pelvic pain during the antibody infusion. Five patients had a delayed extremity pain syndrome following the third and fourth antibody infusion. Four of the five patients developed neurological toxicity, including two patients with significant although reversible motor neuropathy. Two of the patients developed hyponatremia secondary to a syndrome of inappropriate antidiuretic hormone. All 12 patients developed high levels of human anti-14G2a antibody. The plasma half-life of 14G2a was 42 +/- 6 (SD) h. One patient each had a partial response, mixed response, and stable disease, respectively. The very modest antitumor activity accompanied by dose-limiting neurological toxicity at total doses greater than 80 mg may restrict the clinical utility of murine 14G2a.