ArticlePDF Available

Innate NKT lymphocytes confer superior adaptive immunity via tumor-capturing dendritic cells

Rockefeller University Press
Journal of Experimental Medicine (JEM)
Authors:

Abstract and Figures

If irradiated tumor cells could be rendered immunogenic, they would provide a safe, broad, and patient-specific array of antigens for immunotherapies. Prior approaches have emphasized genetic transduction of live tumor cells to express cytokines, costimulators, and surrogate foreign antigens. We asked if immunity could be achieved by delivering irradiated, major histocompatibility complex-negative plasmacytoma cells to maturing mouse dendritic cells (DCs) within lymphoid organs. Tumor cells injected intravenously (i.v.) were captured by splenic DCs, whereas subcutaneous (s.c.) injection led only to weak uptake in lymph node or spleen. The natural killer T (NKT) cells mobilizing glycolipid alpha-galactosyl ceramide, used to mature splenic DCs, served as an effective adjuvant to induce protective immunity. This adjuvant function was mimicked by a combination of poly IC and agonistic alphaCD40 antibody. The adjuvant glycolipid had to be coadministered with tumor cells i.v. rather than s.c. Specific resistance was generated both to a plasmacytoma and lymphoma. The resistance afforded by a single vaccination lasted >2 mo and required both CD4+ and CD8+ T cells. Mature tumor capturing DCs stimulated the differentiation of P1A tumor antigen-specific, CD8+ T cells and uniquely transferred tumor resistance to naive mice. Therefore, the access of dying tumor cells to DCs that are maturing to activated NKT cells efficiently induces long-lived adaptive resistance.
Content may be subject to copyright.
The Journal of Experimental Medicine
JEM © The Rockefeller University Press $8.00
Vol. 202, No. 11, December 5, 2005 1507–1516 www.jem.org/cgi/doi/10.1084/jem.20050956
ARTICLE
1507
Innate NKT lymphocytes confer superior
adaptive immunity via tumor-capturing
dendritic cells
Kang Liu,
1
Juliana Idoyaga,
2
Anna Charalambous,
1
Shin-ichiro Fujii,
1
Anthony Bonito,
1
Jose Mordoh,
2
Rosa Wainstok,
2,3
Xue-Feng Bai,
4
Yang Liu,
4
and Ralph M. Steinman
1
1
Laboratory of Cellular Physiology and Immunology, The Rockefeller University, New York, NY 10021
2
Instituto Leloir, Instituto de Investigaciones Bioquimicas and
3
Departamento de Química Biológica, Facultad de Ciencias
Exactas y Naturales, Universidad de Buenos Aires, 1650 Buenos Aires, Argentina
4
Division of Cancer Immunology, Department of Pathology, Ohio State University Medical Center, Columbus, OH 43210
If irradiated tumor cells could be rendered immunogenic, they would provide a safe, broad,
and patient-specific array of antigens for immunotherapies. Prior approaches have emphasized
genetic transduction of live tumor cells to express cytokines, costimulators, and surrogate
foreign antigens. We asked if immunity could be achieved by delivering irradiated, major
histocompatibility complex–negative plasmacytoma cells to maturing mouse dendritic cells
(DCs) within lymphoid organs. Tumor cells injected intravenously (i.v.) were captured by
splenic DCs, whereas subcutaneous (s.c.) injection led only to weak uptake in lymph node or
spleen. The natural killer T (NKT) cells mobilizing glycolipid
-galactosyl ceramide, used to
mature splenic DCs, served as an effective adjuvant to induce protective immunity. This
adjuvant function was mimicked by a combination of poly IC and agonistic
CD40 antibody.
The adjuvant glycolipid had to be coadministered with tumor cells i.v. rather than s.c.
Specific resistance was generated both to a plasmacytoma and lymphoma. The resistance
afforded by a single vaccination lasted
2 mo and required both CD4
and CD8
T cells.
Mature tumor capturing DCs stimulated the differentiation of P1A tumor antigen-specific,
CD8
T cells and uniquely transferred tumor resistance to naive mice. Therefore, the access
of dying tumor cells to DCs that are maturing to activated NKT cells efficiently induces
long-lived adaptive resistance.
The use of autologous tumor cells as vaccines
dates back to the 1950s when it was found that
chemically induced tumors of inbred mice, if
injected as irradiated cells, could elicit protective
immunity in syngeneic hosts (1). The prospect of
rendering irradiated tumor cells immunogenic is
important because this would deliver a large
spectrum of epitopes to the immune system,
including critical regression antigens that may be
specific to an individual tumor (2). However, it
has been difficult to induce protective immunity
with inactive tumor cells (3–6). The injected cells
can even expand suppressor cells leading to
unresponsiveness (3). It remains a considerable
scientific challenge to learn to improve the im-
munogenicity of safe nonreplicating tumor cell
vaccines.
Tumor cells have been transduced to express
foreign proteins as surrogate antigens (7–10),
but this approach does not address the capacity
of the immune system to respond to a spectrum
of intrinsic tumor antigens. Tumor cells have
also been genetically modified to express individ-
ual costimulatory molecules from the B7 family
(B7.1, B7H, B7-DC) (11–13) or the TNF
superfamily (LIGHT) (14). These modifications
can improve T cell–mediated antitumor re-
sponses, although there is evidence that the
tumor cells must still be presented by host
antigen-presenting cells (15). In addition, tumor
cells have been transduced to secrete cytokines
such as interferons, interleukins, and hemato-
poietins that increase systemic immunity (16–
18). Tumors secreting IL-12 and GM-CSF are
most effective in eliciting T cell–mediated tumor
immunity (19–21). Both IL-12 and GM-CSF
The online version of this article contains supplemental material.
CORRESPONDENCE
Ralph M. Steinman:
steinma@mail.rockefeller.edu
Abbreviations used:
-Gal Cer,
-galactosyl ceramide; CFSE,
carboxyfluorescein diacetate
succinimidyl ester; NKT, natural
killer T.
PROTECTIVE IMMUNITY VIA MATURE, TUMOR-CAPTURING DCS | Liu et al.
1508
can mediate the recruitment of DCs (21–23), which are
specialized antigen-presenting cells for initiating immunity.
However, there are limitations to the clinical usefulness
of genetically modified tumors. These approaches typically
require the administration of live tumor cells, which raises a
safety issue (4, 5). To prioritize responses to patient-specific
antigens, tumor cell lines must be available from the patient
for purposes of genetic transduction, and this may be im-
practical. In addition, genetically modified tumors depend
on a few, often one, immune enhancing product.
An alternative to genetic transduction would be to
identify mechanisms required to induce immunity to irra-
diated tumor cells. Two principles would seem valuable in
this regard: (a) to learn to deliver dying whole tumor cells
directly to DCs that are abundant in lymphoid organs, and
(b) to render the DCs immunogenic through their differ-
entiation or maturation. A key feature of DCs is the effi-
ciency with which dying cells are processed for presenta-
tion on MHC class I and II products (24, 25). In vivo
dying cells injected intravenously are selectively taken
up by a subset of splenic DCs marked by expression of
CD8

and DEC-205. This leads to the presentation of a
surrogate antigen, ovalbumin, from the dying cells (26, 27).
Such “cross-presentation” of antigens by host cells, rather
than direct presentation by the tumor cells themselves,
primes protective T cells against several experimental tu-
mors (28–30), and this has now been observed with a cel-
lular human tumor vaccine as well (31). However, cross-
presentation of model antigens from dying cells in the
steady state leads to tolerance (26), whereas immunity de-
velops when the DCs simultaneously mature, for example,
in response to innate natural killer T (NKT) lymphocytes
(32, 33). NKT cells not only act as adjuvants by maturing
DCs but they also have additional valuable adjunct func-
tions in tumor resistance. NKT cells can produce IFN-
and lyse tumor targets, whereas the interaction of NKT
cells with DCs leads to the production of IL-12 and the ac-
tivation of other innate NK cells (34–37). Thus, NKT cells
are a potentially attractive means to link innate and adaptive
immunity against tumors.
Prior approaches to inducing immunity with safe irradi-
ated tumor cells have not directly addressed the need for tu-
mor cells to gain access to maturing DCs, to allow cross pre-
sentation and to induce strong T cell–mediated immunity.
Here, we have studied the nonimmunogenic plasmacytoma
cell line, J558, because it expresses a classical tumor antigen
called P1A (38) for which TCR transgenic T cells have been
made (39), and because J558 has an MHC class I–negative
variant that is genetically incapable of direct presentation
(40). We will describe that a single dose of irradiated J558
plasmacytoma cells, as well as A20 lymphoma cells, when
targeted to DCs, leads to long-lasting adaptive immunity as
long as the DCs are matured with innate NKT cells or a
combination of the TLR ligand, poly IC, and agonistic anti-
CD40 antibody.
RESULTS
Dying tumor cells are selectively captured by DCs
when given by the i.v. route
We first verified that DCs in mouse spleen could take up the
MHC class I–negative J558
variant of a mouse plasmacy-
toma. This variant has lost expression of cell surface MHC
class I and multiple antigen presentation genes, including
TAP-1, TAP-2, LMP-2, and LMP-7, due to malfunction of
Figure 1. Intravenous delivery of dying tumor cells to CD8
CD11c DCs in vivo. (A) Kinetics of uptake of 20 106 dying, irradiated,
CFSE-labeled J558 tumor cells injected i.v. or s.c., by CD11c spleen
(SPLN), draining (LN), and distal (dLN) lymph node DCs and analyzed with
flow cytometry as in B. (B) Flow cytometric assays to show selective
uptake of irradiated CFSE-labeled J558 tumor cells by CD8, CD11c
splenic DCs 2 h later (arrows). Comparison of CD11c and CD11c spleen
cells (top), and CD8 CD11c toCD8, CD11c DC subsets (bottom).
(C) Uptake by CD11c DC subsets of graded doses of CFSE-labeled tumor
cells i.v. 5, 20, or 106 irradiated CFSE-labeled J558 tumor cells were
injected i.v. Uptake 2 h after injection is shown in gated CD11c splenic
DCs (arrows).
JEM VOL. 202, December 5, 2005
1509
ARTICLE
the proto-oncogene
pml
(40, 41). Because the tumor cells
lack MHC class I, as we confirmed (Fig. S1 A, available at
http://www.jem.org/cgi/content/full/jem.20050956/DC1),
and also fail to express MHC class II (not depicted), recip-
ient antigen-presenting cells would have to capture and
process J558
cells to elicit CD8
and CD4
T cell re-
sponses. To verify that the J558
cells underwent cell death
after 75 Gy
-irradiation, we placed the cells in culture and
showed that with time, they could be stained with annexin
V and propidium iodide (see Materials and methods and Fig.
S1 B). We followed the uptake of carboxyfluorescein diace-
tate succinimidyl ester (CFSE)–labeled, irradiated, “dying”
J558
cells in vivo by lymph node and splenic DCs after in-
jection by the i.v. and s.c. routes, using flow cytometry. Fig.
1 A shows that the tumor cells, injected immediately after ir-
radiation, were phagocytosed by CD11c
splenic DCs. In
contrast, few CFSE-labeled cells were detected in splenic or
lymph node DCs when the tumor cells were injected by the
s.c. route (Fig. 1 A). CFSE-labeled tumor material was pri-
marily detected in the CD11c
DC-enriched populations
(Fig. 1 B, top) and, as expected from prior work (27), only
the CD8
CD11c
DC subset endocytosed the injected
CFSE-labeled tumor (Fig. 1 B, bottom). In experiments that
are not depicted, we established that few CFSE-labeled cells
were taken up by other CD11c
fractions of spleen, marked
for either CD11b, B220, or CD4. Fig. 1 C shows that the
uptake of J558
cells was limited by the dose of injected tu-
mor. Furthermore, irradiated A20 (MHC I
MHC II
),
J558 (MHC I
MHC II
), and J558
(MHC I
MHC II
)
were phagocytosed by DCs to similar extents (unpublished
data). Therefore, dying tumor cells are selectively captured
by DCs in vivo as long as the i.v. route is used.
-Gal Cer injection leads to rapid maturation of phagocytic
DCs in vivo
-Galactosyl ceramide (
-Gal Cer) is a nonmammalian gly-
colipid that is presented by CD1d molecules to an invariant
T cell receptor expressed by innate NKT lymphocytes (42).
A single i.v. dose of
-Gal Cer activates NKT cells to secrete
IL-4 and IFN-
, but this activation also leads to the matura-
tion of DCs in vivo, defined as the ability to initiate com-
bined CD4
and CD8
T cell immunity to coadministered
antigens (32, 33, 43). We established that the coadministra-
tion of
-Gal Cer with both J558 and A20 tumor cells did
not increase the level of tumor cell uptake by DCs; actually,
uptake could be slightly lower, probably because maturing
DCs are known to decrease their level of endocytic activity
(44). Also, in the presence of
-Gal Cer, tumor cell uptake
remained restricted to the CD8
subset of DCs. To evalu-
ate the maturation status of the DCs that had phagocytosed
dying tumor cells, we injected mice with CFSE-labeled, ir-
radiated MHC-negative, J558
cells in the presence or ab-
sence of
-Gal Cer. 5 h later, the DCs were analyzed by
flow cytometry for the expression of a number of cell surface
molecules that change during DC maturation. Injection of
tumor cells alone had little effect on the phenotype of the
total CD11c
splenic population relative to the PBS control
(Fig. 2, compare first and second rows). However, injection
of
-Gal Cer (not depicted) or coinjection of tumor cells
with
-Gal Cer (Fig. 2, third row) resulted in the maturation
of the total CD11c
DC population, as indicated by the up-
regulation of MHC II, CD80, CD86, B7-H1, and B7-DC
5 h later (Fig. 2, third row). When we looked selectively at
those DCs that had captured dying tumor cells (by examin-
ing cells positive for CD11c and CFSE), we found that the
phagocytic cells, which represent
3% of the splenic DCs
(Fig. 1 A), had higher levels of CD1d and other markers (Fig.
2, fourth row). The phagocytic cells strongly up-regulated
the expression of all the measured antigen-presenting and co-
stimulatory molecules in response to
-Gal Cer (Fig. 2, com-
pare fourth and fifth rows). Therefore, the administration of
-Gal Cer allows DCs that capture tumor cells to exhibit
numerous phenotypic changes typical of DC maturation.
Injection of dying tumor cells together with
-Gal Cer
induces tumor immunity
To determine if immunity was induced by delivery of dying
tumor cells to DCs, we used a tumor protection assay. Dif-
ferent groups of naive BALB/c mice were injected with
PBS,
-Gal Cer, 20
10
6
irradiated J558
tumor cells
alone, or J558
together with
-Gal Cer. 7 d after the vacci-
nation, we challenged the mice with 5
10
6
J558 live tu-
Figure 2. -Gal Cer injection rapidly matures CD1d-rich, splenic
DCs capturing dying cells. BALB/c mice were injected with PBS or 20
106 irradiated CFSE-labeled J558 tumor cells i.v. in the presence or
absence of 2 g -Gal Cer. 4.5 h later, bulk spleen cells were prepared
from the mice and stained with CD11c-allophycocyanin and PE-conjugated
mAbs CD40, 80, 86, B7-H1, B7-DC, Ld, I-Ad, and CD1d. The data are shown
for the CD11c total DC population (top three rows) and CD11c CFSE
phagocytic population (bottom two rows).
PROTECTIVE IMMUNITY VIA MATURE, TUMOR-CAPTURING DCS | Liu et al.
1510
mor cells s.c. The tumor grew progressively within 7–10 d
in mice that had received either PBS or
-Gal Cer alone
(Fig. 3 A). Injection with irradiated J558 tumor cells alone
protected only 15% mice from J558 challenge (3 out of 20
mice in four experiments), whereas 88% of the mice (22 out
of 25 in five experiments) were protected and remained tu-
mor-free after vaccination with the combination of irradi-
ated J558
with
-Gal Cer (Fig. 3 A). We were not able to
vaccinate mice with live J558
cells, which grew rapidly and
killed the mice. Vaccination with irradiated J558
and
-Gal
Cer was effective only when tumor cells were injected by
the i.v. and not the s.c. route (Fig. 3 B), as predicted by the
data in Fig. 1 A in which tumor cells had to be injected i.v.
to be picked up well by DCs. Corresponding to the uptake
data in Fig. 1 C, the level of protection varied with the dose
of injected tumor cells with 10
6
cells being ineffective (Fig. 3
C). To evaluate the specificity of the antitumor immune re-
sponse, we challenged the mice with either live Meth A sar-
coma cells s.c. or live A20 lymphoma cells after vaccination
with irradiated J558 with
-Gal Cer. Vaccination with dying
J558 L
d
and
-Gal Cer protected mice against a challenge
of J558, but not Meth A or A20 (Fig. 3 D). Likewise, immu-
nization with dying A20 with -Gal Cer led to protection
against only A20, but not J558 (Fig. 3 E). To compare -Gal
Cer with other DC maturation stimuli, we studied agonistic
anti-CD40 monoclonal antibody as well as the TLR3 ligand,
poly IC. The glycolipid was the only adjuvant that could in-
dependently induce protective immunity to irradiated tumor
injected i.v., but the combination of anti-CD40 and poly IC
was also active (Fig. 3 F). To assess memory, we verified that
Figure 3. Acquired resistance to J558 after vaccination with MHC I
tumor and -Gal Cer. (A) Mice were vaccinated with PBS, 2 g -Gal Cer,
20 106 irradiated MHC I–negative J558 cells with or without 2 g -Gal
Cer i.v. 1 wk later, mice were challenged with a lethal tumorogenic dose of
5 106 live MHC I J558 tumor cells s.c. (B) Mice were vaccinated with PBS,
20 106 irradiated J558 cells alone, or 20 106 irradiated J558 cells with
2 g Gal Cer i.v. or s.c. and, 1 wk later, challenged with 5 106 live MHC
class I J558 cells s.c. (C) Mice were vaccinated with 1, 5, or 20 106
irradiated J558 cells plus 2 g -Gal Cer i.v. and, 1 wk later, challenged with
5 106 J558 s.c. (D) Mice were vaccinated with 20 106 irradiated J558
cells plus 2 g -Gal Cer i.v. and, 1 wk later, challenged with 5 106 J558 or
to establish tumor specificity, with 5 106 Meth A sarcoma or 5 106 A20
lymphoma cells s.c. (E) Mice were vaccinated with 5 106 irradiated A20
cells plus 2 g -Gal Cer i.v. and 1 wk later, challenged with 5 106 J558 or
5 106 A20 cells s.c. (F) Mice were vaccinated with 5 106 irradiated J558
cells and either 2 g -Gal Cer i.v., 50 g CD40 i.p, 50 g polyI:C i.p., or
both CD40 and poly IC and, 3 wk later, each group was challenged with
5 106 J558 s.c. (G) 8 wk after vaccination, mice were challenged with a
lethal dose of 5 106 live J558 tumor cells. (H) To detect a therapeutic effect,
mice were injected with 1 or 5 106 live J558 tumor cells s.c., and 3 d later
treated with PBS, or 20 106 irradiated J558 cells with 2 g -Gal Cer i.v.
In all experiments, mice were monitored every other day for tumor growth
and scored positive when the tumors were palpable. Each group included at
least five mice, and one representative experiment of three is shown.
Figure 4. CD4 or CD8 T cell depletion abrogates tumor immunity
at the time of challenge. (A) Mice were vaccinated with 20 106 irradiated
J558 tumor cells plus -Gal Cer. 7 d after immunization, mice were
challenged with 5 106 MHC class I positive or negative J558 cells s.c.
(B) Wild-type BALB/c mice or J18/ mice were vaccinated with 20 106
irradiated J558 tumor cells plus -Gal Cer and, 3 wk after vaccination,
the mice were challenged with 5 106 J558 cells s.c. (C) 8 wk after
vaccination with 20 106 irradiated J558 tumor cells and -Gal Cer, mice
received 1 mg anti-CD4, anti-CD8, or control rat IgG. 1 d later, all mice
were challenged with 5 106 J558 tumor cells and monitored every other
day for tumor growth. Mice were scored positive when the tumors were
palpable. Each group included five mice; one representative experiment
of two is shown.
JEM VOL. 202, December 5, 2005 1511
ARTICLE
vaccinated mice were protected against J558 challenge 2 mo
(Fig. 3 G) and 4 mo (not depicted) after vaccination. We also
induced tumor resistance if we performed an immunother-
apy-type experiment and vaccinated 3 d after the injection of
the tumor cells (Fig. 3 H). However, therapeutic immunity
was manifest only when the tumor dose was 106 and not 5
106 cells, whereas protective immunity was evident in mice
challenged with both doses. Thus, long-lived tumor immu-
nity can be elicited by a single i.v. vaccination with irradiated
J558 and -Gal Cer, which also has a therapeutic effect.
Innate and adaptive T cells are required to generate
resistance after vaccination with dying tumor and -Gal Cer
To identify resistance mechanisms for the observed protec-
tive tumor immunity, we vaccinated mice with dying MHC
class I J558 plus -Gal Cer and challenged them with live
MHC class I–negative or positive J558 tumor cells. The vac-
cinated mice were protected against the MHC class I–posi-
tive J558 cells, but not the MHC class I negative J558 tu-
mor cells (Fig. 4 A), suggesting that CD8 T cells were
required for resistance. As expected, NKT cells were also re-
quired for effective vaccination because J281/ mice (also
termed J18/), which cannot respond to -Gal Cer be-
cause they lack NKT cells (45), failed to develop immunity
to dying cells plus glycolipid (Fig. 4 B). To further evaluate
the type of T cells required for protective immunity 8 wk af-
ter a single vaccination, the immune mice were injected with
depleting antibodies specific for CD4, CD8, or control IgG
and challenged with J558 cells 1 d later. We verified by
FACS that anti-CD4 and anti-CD8 antibodies depleted the
respective cell populations within 2 d, and that the depletion
lasted 2 wk, when they began to repopulate slowly as re-
ported (46). As shown in Fig. 4 C, mice injected with con-
trol IgG remained resistant to J558 challenge. However, de-
Figure 5. Proliferation and differentiation of tumor antigen-specific
T cells in response to dying tumor cell delivery in vivo. (A) 3 106
CFSE-labeled, naive CD8 P1CTL T cells were injected i.v. into BALB/c
mice. 1 d later, 20 106 J558 cells were injected i.v. with or without
2 g -Gal Cer coinjection. 3 d later, mice were killed and spleen cells
were stained with CD8-allophycocyanin and CD25 or CD62L-PE. (B) Spleen
cells obtained as in A were cultured at 107/ml with 1 g/ml P1A 35–43
peptide (LPYLGWLVF) for 4 h at 37C in the presence of 5 g/ml BFA. The
depicted cells were from gated CD8CFSE P1A transgenic T cells, and
intracellular IFN- and IL-2 were identified with PE-conjugated mAb
after fixation and permeabilization. Black lines are staining with isotype
control antibody; gray lines are staining with anti–IFN- or IL-2. Percentages
are the fraction of transgenic T cells expressing intracellular cytokine
above background.
PROTECTIVE IMMUNITY VIA MATURE, TUMOR-CAPTURING DCS | Liu et al.1512
pletion of either CD4 or CD8 T cells from vaccinated
mice significantly abrogated tumor immunity elicited by J558
with -Gal Cer (Fig. 4 C). Therefore, both innate NKT cells
and adaptive CD4 and CD8 T cells contribute to the tumor
resistance induced by DCs capturing dying cells in vivo.
-Gal Cer coinjection with dying cells better activates
antigen-specific CD8 T cells
To document the consequences of -Gal Cer for the quality
of an antigen-specific T cell response to irradiated tumor, we
took advantage of the P1CTL mouse, a CD8 TCR trans-
genic line specific for P1A tumor antigen presented on Ld
MHC class I molecules (39). 20 106 irradiated MHC class
I–negative, J558 cells with or without -Gal Cer were in-
jected into the mice that had received CFSE-labeled P1CTL
cells 1 d earlier. T cell proliferation and phenotype were an-
alyzed 3 d later with flow cytometry. In the absence of
-Gal Cer, DCs could cross-present P1A from dying tumor
cells to CD8 P1CTL T cells, driving the T cells into multi-
ple cycles of proliferation (Fig. 5, top row). This is consistent
with the capacity of CD8 DCs to present antigens on both
MHC class I and II products from dying cells in the steady
state (26, 27). However, the proliferating P1CTL T cells re-
tained markers typical of naive cells, i.e., low CD25 and
high CD62L (Fig. 5 A, white arrows). In contrast, in the
mice that had received J558 plus -Gal Cer, the T cells
proliferated more extensively and many began to show an
activation phenotype within 3 d, as indicated by the up-reg-
ulation of CD25 and down-regulation of CD62L (Fig. 5 A,
compare black and white arrows). Furthermore, T cells that
had been stimulated in the presence of -Gal Cer adjuvant
in vivo were able to produce significantly more IFN- and
IL-2 upon brief restimulation with P1A peptide in vitro
(Fig. 5 B, compare right with middle and left panels). There-
fore, DCs process antigens from tumor cells and induce the
proliferation of antigen-specific T cells in vivo, but a matu-
ration stimulus is required for the differentiation of effector
T cells (Fig. 5) as well as protective immunity (Figs. 3 and 4).
Evidence that mature DCs present tumor antigen
and transfer tumor immunity
To verify that mature DCs were responsible for the presenta-
tion of antigens from the captured dying tumor cells and also
elicited tumor immunity, we first isolated DCs from mice in-
jected with dying J558 tumor cells without or with -Gal
Cer. The CD11c DC-enriched and CD11c DC-depleted
Figure 6. Maturing DCs mediate P1A antigen presentation and elicit
tumor immunity in vivo. Mice were given -Gal Cer or PBS together with
irradiated J558- cells. 4 h later, CD11c and CD11c cells were isolated and
used to stimulate CD8 TCR transgenic T cells from P1CTL mice in vitro (A)
or in vivo (B). In A, in vitro T cell proliferation was measured by a [3H]thymidine
pulse at 40–50 h. In B, in vivo proliferation of CFSE-labeled P1CTL T cells
was measured 3 d later. (C) Splenic CD11c and CD11c cells were isolated
from mice injected with irradiated J558 cells and -Gal Cer 4 h earlier.
106 CD11c or 5 106 CD11c cells were then transferred i.v. into naive
BALB/c mice. 1 wk later, the mice were challenged with live 5 106 J558
cells. Mice were monitored every 3 d for tumor growth. Each group includes
12–17 mice pooled from three experiments.
JEM VOL. 202, December 5, 2005 1513
ARTICLE
cells from spleen were added as stimulators in culture of na-
ive CD8 P1CTL TCR transgenic T cells without further
antigen. When -Gal Cer had been coadministered, the iso-
lated DCs were much more effective at stimulating prolifera-
tion of naive CD8 T cells in culture, whereas CD11c cells
were inactive (Fig. 6 A, right, closed squares). Next, we iso-
lated CD11c DCs from spleens 4 h after immunization and
transferred the DCs to naive animals to test their capacity to
stimulate proliferation of CFSE-labeled CD8 P1CTL T
cells in vivo (Fig. 6 B). 3 d later, we detected P1CTL prolif-
eration only in response to DCs from mice given irradiated
tumor cells with -Gal Cer (Fig. 6 B, black arrow). CD11c
non-DCs from the same mice were not able to stimulate
P1CTL T cells, and DCs from mice that had received J558
without -Gal Cer failed to stimulate P1CTL above the
background (Fig. 6 B). Finally, to test if the antigen-present-
ing mature DCs were critical for inducing protective tumor
immunity, we transferred DCs or non-DCs from the vacci-
nated mice into naive mice and then challenged them with
live J558 tumor cells. As shown in Fig. 6 C, when naive
mice had been given 1.5 106 CD11c DCs from donor
mice injected with dying J558 cells together with -Gal
Cer, 58% of mice (10 out of 17 mice tested; P 0.01) were
fully protected. CD11c non-DCs and CD11c DCs from
mice injected with PBS or dying J558 cells alone, failed to
transfer protection to naive mice (0/12, Fig. 6 C). These data
provide direct evidence that mature antigen-capturing DCs
are responsible for presentation of tumor antigen and the
adjuvant action of -Gal Cer in vivo.
DISCUSSION
Genetic transduction of tumor cells is being used to increase
the adaptive immune response to tumor antigens, including
strategies in which cytokines and chemokines are introduced
to recruit DCs to the tumor. An alternative would be to
learn to deliver dying whole tumor cells to maturing DCs
within lymphoid tissues because these cells have dozens of
specializations pertinent to the initiation of immunity, in-
cluding ready access to the recirculating pool of T cells.
Here, we provide some mechanisms to address this goal.
The earliest attempts to increase the immunogenicity of
whole tumor cells and stimulate T cell–mediated tumor immu-
nity used bacterial components such as BCG or C. parvum as
adjuvants (47–49). Although these approaches have achieved
only limited success (50), they imply a need for coordination
between innate immunity and adaptive immunity against tu-
mors. Interestingly, innate NKT cells are required for the anti-
tumor effects of both IL-12 and GM-CSF (45, 51), the two cy-
tokines that are most often successful in improving immunity to
genetically modified tumor cells. Our findings indicate that
NKT cells can serve as superior adjuvants for protective antitu-
mor immunity. In prior studies, we studied the capacity of
NKT cells to mature DCs that were presenting the foreign anti-
gen, ovalbumin (32, 43), but in the current paper, we have ex-
amined the generation of long-lived T cell memory and
protection to a poorly immunogenic syngeneic tumor.
We find that a single dose of irradiated nonmodified tu-
mor cells, when directed to maturing DCs in vivo, here by
the i.v. route, leads to long-lived protective and combined
CD4 and CD8 T cell immunity. We compared the effi-
cacy of targeted i.v. delivery of J558 tumor to maturing DCs
with the use of DCs that were derived from bone mar-
row precursors and loaded with J558 tumor ex vivo as de-
scribed previously (6). However, two doses of 200,000 tu-
mor loaded DCs given 1 wk apart by the s.c. route, failed to
protect the mice to challenge with J558 s.c. The -Gal Cer
maturation stimulus we used to mature splenic DCs in vivo
induced superior protective immunity relative to other DC
stimuli, such as ligation of CD40, TLR4, and TLR3. This
glycolipid has been manufactured in a nontoxic form for hu-
man use (52). It is presented on CD1d molecules to activate
NKT lymphocytes (42, 45), which then mature the DCs as
mentioned (32, 33). When NKT cells are activated with a
single dose of -Gal Cer, there is a production of immune
enhancing cytokines, specifically TNF and IL-12 by the
DCs, and IFN- by NKT and NK cells, and there is an up-
regulation of CD40L on the NKT cells. To elicit protective
immunity to a syngeneic tumor, we find that the combina-
tion of a proinflammatory TLR ligand, poly IC, and agonis-
tic anti-CD40 antibody is able to mimic the effects of -Gal
Cer. Interestingly, each stimulus (poly IC, anti-CD40,
-Gal Cer) induces similar phenotypic changes of matura-
tion; i.e., increased expression of CD40, CD86, MHC class
II, B7-H1, B7-H2, and decreased interferon- receptor or
CD119, but for protective immunity, either -Gal Cer or
the combination of poly IC and CD40 is required.
When DCs were isolated from mice injected with irradi-
ated tumor cells, we could establish that the DCs from gly-
colipid-treated mice were better stimulators of CD8 T cells
specific for the P1A tumor antigen in vitro. Importantly,
transfer of these maturing DCs to naive mice protected
60% of them, whereas direct vaccination of dying tumor
cells with -Gal Cer protected 80–100% of the mice. The
DC transfer experiments might not have been optimal be-
cause they entailed the transfer of only 10-30,000 DCs
bearing tumor cells (only a small percent of the CD8 DCs
take up tumor cells in Fig. 1), which also might not lodge in
the recipient lymphoid tissues with high efficiency.
The harnessing of DCs in this paper offers some advan-
tages over genetic modification of tumor cells to improve
immunogenicity. First, maturing DCs express a plethora of
cytokines, chemokines, and accessory molecules (32, 53, 54)
as illustrated in Fig. 2. Second, because of the capacity of
DCs to cross-present cell-associated antigens in vivo (26,
55), the tumor cells become an effective source of antigen,
potentially a spectrum of antigens for a broad immune at-
tack. Third, by delivering tumor cells to the DCs, one en-
hances antigen presentation beyond the presenting capacities
of the tumor cells themselves, because DCs express such effi-
cient processing pathways for MHC class I (26), class II (55),
and CD1 (56), even if the tumor cells have dampened their
own antigen-presenting activities, as is often the case (57).
PROTECTIVE IMMUNITY VIA MATURE, TUMOR-CAPTURING DCS | Liu et al.1514
This is illustrated by our findings with the presentation of
P1A. This is a classical tumor antigen, one of the first specific
antigens to be described by Van den Eynde et al. (38). The
experiments in Fig. 5 reveal the processing and presentation
of the nonmutated P1A antigen from the tumor cells.
Nonetheless there are limitations to our experiments.
We would like to mention three. First, the delivery of tumor
cells to DCs, while enhanced by the use of the i.v. route of
injection, requires a relatively large dose of irradiated tumor
in mice, at least 5 106 cells. Efficacy might be further in-
creased, for example, by opsonizing the tumor or otherwise
increasing the frequency of antigen-capturing DCs. Like-
wise, human research will be needed to determine the effec-
tive dose that will result in immunization of patients (e.g.,
with hematologic malignancies like myeloma). Second, we
have tested only one CD1d-binding glycolipid, -Gal Cer,
and there are newer synthetic and natural glycolipids that
may prove to be more effective (58, 59). As mentioned,
-Gal Cer has already been found to be nontoxic in humans,
even at relatively high doses. Third, our studies do not ad-
dress the potentially important regulation imparted by sup-
pressor T cells which, beginning with the work of R. North
and colleagues, has been shown to be able to regulate the
function of protective or effector T lymphocytes (60, 61).
The approach in this paper takes full advantage of the pos-
itive feedback between NKT cells and DCs that occurs when
a single dose of -Gal Cer is given together with irradiated tu-
mor cells. We have observed specific immunity to two differ-
ent hematopoietic tumors, the J558 plasmacytoma and the
A20 lymphoma. We would like to suggest that vaccination
with dying tumor cells, under conditions where the tumor
cells are captured by DCs maturing in response to innate
NKT cells, be evaluated in humans, and that this be initiated
with hematologic malignancies such as the tumors tested here.
MATERIALS AND METHODS
Mice. 6-8-wk-old BALB/c females were obtained from Taconic. BALB/c
transgenic mice expressing a TCR specific for the tumor antigen P1A35–
43:Ld complex have been described previously (39). J18/ mice on
BALB/c background were gifts from M. Tsuji (New York University
School of Medicine, New York, NY). Mice were maintained under spe-
cific pathogen-free conditions. All experiments were conducted according
to institutional guidelines.
Cell lines. The plasmacytoma J558 and the MHC class I mutant cell line
J558Ld have been described previously (40). The Meth A fibrosarcoma
was provided by Z. Li (University of Connecticut Health Center, Farming-
ton, CT). The A20 lymphoma was provided by H.I. Levitsky (Sidney Kim-
mel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD).
The cell lines were cultured in RPMI 1640 medium supplemented with
10% FCS, 100 g/ml penicillin/streptomycin, and 2 mM glutamine. All
lines tested negative for Mycoplasma by Hoechst staining and PCR reaction
(American Type Culture Collection).
Reagents. Rat mAbs for MHC class II (TIB120, M5/114.15.2), granulo-
cytes (RB6-8C5, Gr-1), B220 (TIB146, RA3-3A1), F4/80 (HB198), CD4
(GK 1.5), and CD8 (TIB211, 3.155) were obtained from the American
Type Culture Collection. Anti-CD16/32, PE-conjugated anti-CD8,
CD11b, B220, CD4, V8.3, IFN-, IL-2, H-2Ld/H-2Db, CD62L, CD69,
B7-H1, B7-DC, and allophycocyanin-CD11c were obtained from BD Bio-
sciences or eBioscience. Sheep anti–rat IgG conjugated to magnetic beads
were obtained from Dynal. Anti-CD11c and CD8 microbeads were ob-
tained from Miltenyi Biotec. The other reagents were RPMI 1640
(GIBCO BRL), FCS (GIBCO BRL), CFSE (GE Healthcare), ACK buffer
(BioSource30% BSA solution (Sigma-Aldrich). -Gal Cer (2S, 3S, 4R-
1-O(-galactopyranosyl)-2(N-hexacosanoylamino)-1,3,4-octadecanetriol) was
provided by Kirin Brewery and diluted in PBS.
Induction of cell death. Tumor cells were harvested, washed twice with
RPMI 1640, resuspended to 10 106/ml in RPMI 1640, and irradiated
with 75 Gy. Detection of apoptotic tumor cells used the annexin V-FITC
Apoptosis Detection Kit (BD Biosciences), after which flow cytometry
(FACS Vantage SE, Becton Dickinson) was performed. Within 24 h, 24%
of the tumor cells were apoptotic, i.e., annexin V positive but PI negative
(Fig. S1 B). By 48 h, 57% of the irradiated cells underwent secondary ne-
crosis; and 72 h later, 84% of them were necrotic (PI). Therefore, we refer
to the irradiated cells that we injected as “dying cells.”
Tumor-specific TCR transgenic T cells. CD8 P1A-specific, T cells
were prepared from meshed cell suspensions of TCR transgenic lymph
nodes and spleen by depleting B220, CD4, F4/80, and MHC class II–
expressing cells using sheep anti–rat IgG Dynabeads. For CFSE labeling (GE
Healthcare), the cells at 107/ml in PBS were incubated with 5 M CFSE for
10 min at 37C. The reaction was stopped by washing three times with PBS.
In vivo delivery of dying tumor cells, DC maturation stimuli, and
tumor protection assay. 2 107 irradiated J558-Ld cells were injected
i.v. or s.c. into BALB/c mice with or without -Gal Cer as a DC matura-
tion stimulus. In some experiments, we compared -Gal Cer to agonisitic
anti-CD40 mAb (1C10, 25 g i.p.) or the Toll-like receptor ligands, poly
IC (50 g i.p., Invivogen) or lipopolysaccharide (20 g i.p., Sigma-
Aldrich). The mice had been given CFSE-labeled P1CTL CD8 T cells i.v.
1 d earlier, or were naive animals. In some experiments, mice were killed 3 d
later, and T cell division and activation in spleen were analyzed by flow cy-
tometry. Additionally, 7 d or 2 mo later, 5 106 live J558 tumor cells were
inoculated subcutaneously. 5 106 Meth A fibrosarcoma was used as a
control tumor for challenge. Tumor cell growth was measured with calipers
every other day. Mice were scored positive for tumor as soon as tumors be-
came palpable and grew progressively. Mice were killed when tumor size
exceeded 400 mm2.
Flow cytometry. T cell division, phagocytosis of CFSE-labeled tumor
cells, and acquisition of cell surface–activation markers were determined by
flow cytometry. In brief, spleens were harvested and low density spleno-
cytes were stained with CD11c-allophycocyanin and CD8-PE, CD11b-
PE, B220-PE, or CD4-PE for the up-take examination. For in vivo prolif-
eration of P1CTL T cells, spleens were harvested and the splenocyte
suspension was staining with CD8-Cy, V8.3-PE, CD25-PE, and CD62L-
PE. For intracellular cytokine staining, splenocytes were stimulated in vitro
with 1 g/ml P1A peptide in 5 g/ml brefeldin A (Sigma-Aldrich) at 37C
for 4 h. The cells were first stained with CD8-Cy-Chrome, fixed, perme-
abilized with cytofix-cytoperm buffer (BD Biosciences), and stained with
PE-conjugated mAbs to IL-2 or IFN-.
In vivo depletion of CD4 and CD8 T lymphocytes. Mice vacci-
nated with J558 and -Gal Cer were injected with ascites containing 1 mg of
rat monoclonal anti-CD4 (clone GK 1.5) or anti-CD8 (clone 53–6.72). The
mice received three daily injections, the first one i.v. 1 d before the challenge,
and two i.p. injections on the day of the challenge and 1 d after the challenge.
Control mice received 1 mg of rat IgG (Jackson ImmunoResearch Laborato-
ries). The depletion was monitored by staining with anti-CD4 and anti-CD8
antibodies followed by flow cytometry (BD Biosciences).
Online supplemental material. Fig. S1 A shows lack of MHC class I
expression on J558 cells relative to parental J558 plasmacytoma. Fig. S1 B
JEM VOL. 202, December 5, 2005 1515
ARTICLE
depicts death of J558 cells, untreated or 24/48/72 h after -irradiation, as
assessed by staining with annexin V and propidium iodide. Online sup-
plemental material is available at http://www.jem.org/cgi/content/full/
jem.20050956/DC1.
K. Liu was supported by a predoctoral fellowship from the Cancer Research Institute
and R.M. Steinman was supported by National Institutes of Health grant nos. AI
13013 and AI 51573.
The authors have no conflicting financial interests.
Submitted: 11 May 2005
Accepted: 26 October 2005
REFERENCES
1. Prehn, R.T., and J.M. Main. 1957. Immunity to methylcholanthrene-
induced sarcomas. J. Natl. Cancer Inst. 18:769–778.
2. Mumberg, D., M. Wick, and H. Schreiber. 1996. Unique tumor anti-
gens redefined as mutant tumor-specific antigens. Semin. Immunol.
8:289–293.
3. Rakhmilevich, A.L., R.J. North, and E.S. Dye. 1993. Presence of
CD4 T suppressor cells in mice rendered unresponsive to tumor an-
tigens by intravenous injection of irradiated tumor cells. Int. J. Cancer.
55:338–343.
4. Townsend, S.E., F.W. Su, J.M. Atherton, and J.P. Allison. 1994. Spec-
ificity and longevity of antitumor immune responses induced by B7-
transfected tumors. Cancer Res. 54:6477–6483.
5. Katsanis, E., M.A. Bausero, A. Panoskaltsis-Mortari, B.B. Dancisak, Z.
Xu, P.J. Orchard, C.G. Davis, and B.R. Blazar. 1996. Irradiation of
singly and doubly transduced murine neuroblastoma cells expressing
B7-1 and producing interferon- reduces their capacity to induce sys-
temic immunity. Cancer Gene Ther. 3:75–82.
6. Goldszmid, R.S., J. Idoyaga, A.I. Bravo, R. Steinman, J. Mordoh, and
R. Wainstok. 2003. Dendritic cells charged with apoptotic tumor cells
induce long-lived protective CD4 and CD8 T cell immunity against
B16 melanoma. J. Immunol. 171:5940–5947.
7. Wang, M., V. Bronte, P.W. Chen, L. Gritz, D. Panicali, S.A. Rosen-
berg, and N.P. Restifo. 1995. Active immunotherapy of cancer with a
nonreplicating recombinant fowlpox virus encoding a model tumor-
associated antigen. J. Immunol. 154:4685–4692.
8. Moore, M.W., F.R. Carbone, and M.J. Bevan. 1988. Introduction of
soluble protein into the class I pathway of antigen processing and pre-
sentation. Cell. 54:777–785.
9. Morgan, D.J., H.T.C. Kreuwel, S. Fleck, H.I. Levitsky, D.M. Pardoll,
and L.A. Sherman. 1998. Activation of low avidity CTL specific for a
self epitope results in tumor rejection but not autoimmunity. J. Immu-
nol. 160:643–651.
10. Staveley-O’Carroll, K., E. Sotomayor, J. Montgomery, I. Borrello, L.
Hwang, S. Fein, D. Pardoll, and H. Levitsky. 1998. Induction of anti-
gen-specific T cell anergy: an early event in the course of tumor pro-
gression. Proc. Natl. Acad. Sci. USA. 95:1178–1183.
11. Dohring, C., L. Angman, G. Spagnoli, and A. Lanzavecchia. 1994.
T-helper- and accessory-cell-independent cytotoxic responses to human
tumor cells transfected with a B7 retroviral vector. Int. J. Cancer. 57:1–6.
12. Liu, X., J.X. Gao, J. Wen, L. Yin, O. Li, T. Zuo, T.F. Gajewski, Y.X.
Fu, P. Zheng, and Y. Liu. 2003. B7DC/PDL2 promotes tumor immu-
nity by a PD-1–independent mechanism. J. Exp. Med. 197:1721–1730.
13. Liu, X., X.F. Bai, J. Wen, J.X. Gao, J. Liu, P. Lu, Y. Wang, P. Zheng,
and Y. Liu. 2001. B7H costimulates clonal expansion of, and cognate
destruction of tumor cells by, CD8 T lymphocytes in vivo. J. Exp.
Med. 194:1339–1348.
14. Tamada, K., K. Shimozaki, A.I. Chapoval, G. Zhu, G. Sica, D. Flies,
T. Boone, H. Hsu, Y.X. Fu, S. Nagata, et al. 2000. Modulation of
T-cell-mediated immunity in tumor and graft-versus-host disease mod-
els through the LIGHT co-stimulatory pathway. Nat. Med. 6:283–289.
15. Bai, X.F., J.X. Gao, J. Liu, J. Wen, P. Zheng, and Y. Liu. 2001. On
the site and mode of antigen presentation for the initiation of clonal
expansion of CD8 T cells specific for a natural tumor antigen. Cancer
Res. 61:6860–6867.
16. Golumbek, P.T., A.J. Lazenby, H.I. Levitsky, L.M. Jaffee, H. Kara-
suyama, M. Baker, and D.M. Pardoll. 1991. Treatment of established
renal cancer by tumor cells engineered to secrete interleukin-4. Science.
254:713–716.
17. Gansbacher, B., K. Zier, B. Daniels, K. Cronin, R. Bannerji, and E.
Gilboa. 1990. Interleukin 2 gene transfer into tumor cells abrogates tu-
morigenicity and induces protective immunity. J. Exp. Med. 172:
1217–1224.
18. Gansbacher, B., R. Bannerji, B. Daniels, K. Zier, K. Cronin, and E.
Gilboa. 1990. Retroviral vector-mediated -interferon gene transfer
into tumor cells generates potent and long lasting antitumor immunity.
Cancer Res. 50:7820–7825.
19. Cavallo, F., P. Signorelli, M. Giovarelli, P. Musiani, A. Modesti, M.J.
Brunda, M.P. Colombo, and G. Forni. 1997. Antitumor efficacy of ad-
enocarcinoma cells engineered to produce interleukin 12 (IL-12) or
other cytokines compared with exogenous IL-12. J. Natl. Cancer Inst.
89:1049–1058.
20. Dunussi-Joannopoulos, K., K. Runyon, J. Erickson, R.G. Schaub,
R.G. Hawley, and J.P. Leonard. 1999. Vaccines with interleukin-12-
transduced acute myeloid leukemia cells elicit very potent therapeutic
and long-lasting protective immunity. Blood. 94:4263–4273.
21. Dranoff, G., E. Jaffee, A. Lazenby, P. Golumbek, H. Levitsky, K.
Brose, V. Jackson, H. Hamada, D. Pardoll, and R.C. Mulligan. 1993.
Vaccination with irradiated tumor cells engineered to secrete murine
granulocyte-macrophate colony-stimulating factor stimulates potent,
specific, and long-lasting anti-tumor immunity. Proc. Natl. Acad. Sci.
USA. 90:3539–3543.
22. Soiffer, R., T. Lynch, M. Mihm, K. Jung, C. Rhuda, J.C. Schmollinger,
F.S. Hodi, L. Liebster, P. Lam, S. Mentzer, et al. 1998. Vaccination
with irradiated autologous melanoma cells engineered to secrete hu-
man granulocyte-macrophage colony-stimulating factor generates po-
tent antitumor immunity in patients with metastatic melanoma. Proc.
Natl. Acad. Sci. USA. 95:13141–13146.
23. Rodolfo, M., C. Zilocchi, B. Cappetti, G. Parmiani, C. Melani, and
M.P. Colombo. 1999. Cytotoxic T lymphocyte response against non-
immunoselected tumor antigens predicts the outcome of gene therapy
with IL-12-transduced tumor cell vaccine. Gene Ther. 6:865–872.
24. Albert, M.L., B. Sauter, and N. Bhardwaj. 1998. Dendritic cells ac-
quire antigen from apoptotic cells and induce class I-restricted CTLs.
Nature. 392:86–89.
25. Inaba, K., S. Turley, F. Yamaide, T. Iyoda, K. Mahnke, M. Inaba, M.
Pack, M. Subklewe, B. Sauter, D. Sheff, et al. 1998. Efficient presenta-
tion of phagocytosed cellular fragments on the MHC class II products
of dendritic cells. J. Exp. Med. 188:2163–2173.
26. Liu, K., T. Iyoda, M. Saternus, K. Kimura, K. Inaba, and R.M. Stein-
man. 2002. Immune tolerance after delivery of dying cells to dendritic
cells in situ. J. Exp. Med. 196:1091–1097.
27. Iyoda, T., S. Shimoyama, K. Liu, Y. Omatsu, Y. Maeda, K. Takahara,
Y. Akiyama, R.M. Steinman, and K. Inaba. 2002. The CD8 den-
dritic cell subset selectively endocytoses dying cells in culture and in
vivo. J. Exp. Med. 195:1289–1302.
28. van Mierlo, G.J., Z.F. Boonman, H.M. Dumortier, A.T. den Boer,
M.F. Fransen, J. Nouta, E.I. van der Voort, R. Offringa, R.E. Toes,
and C.J. Melief. 2004. Activation of dendritic cells that cross-present
tumor-derived antigen licenses CD8 CTL to cause tumor eradication.
J. Immunol. 173:6753–6759.
29. Yu, P., M.T. Spiotto, Y. Lee, H. Schreiber, and Y.X. Fu. 2003. Com-
plementary role of CD4 T cells and secondary lymphoid tissues for
cross-presentation of tumor antigen to CD8 T cells. J. Exp. Med. 197:
985–995.
30. Huang, A.Y.C., P. Golumbek, M. Ahmadzadeh, E. Jaffee, D. Pardoll,
and H. Levitsky. 1994. Role of bone marrow-derived cells in present-
ing MHC class I-restricted tumor antigens. Science. 264:961–965.
31. Thomas, A.M., L.M. Santarsiero, E.R. Lutz, T.D. Armstrong, Y.C.
Chen, L.Q. Huang, D.A. Laheru, M. Goggins, R.H. Hruban, and
E.M. Jaffee. 2004. Mesothelin-specific CD8 T cell responses provide
evidence of in vivo cross-priming by antigen-presenting cells in vacci-
nated pancreatic cancer patients. J. Exp. Med. 200:297–306.
32. Fujii, S., K. Shimizu, C. Smith, L. Bonifaz, and R.M. Steinman. 2003.
Activation of natural killer T cells by -galactosylceramide rapidly in-
PROTECTIVE IMMUNITY VIA MATURE, TUMOR-CAPTURING DCS | Liu et al.1516
duces the full maturation of dendritic cells in vivo and thereby acts as
an adjuvant for combined CD4 and CD8 T cell immunity to a co-
administered protein. J. Exp. Med. 198:267–279.
33. Hermans, I.F., J.D. Silk, U. Gileadi, M. Salio, B. Mathew, G. Ritter, R.
Schmidt, A.L. Harris, L. Old, and V. Cerundolo. 2003. NKT cells en-
hance CD4 and CD8 T cell responses to soluble antigen in vivo
through direct interaction with dendritic cells. J. Immunol. 171:5140–5147.
34. Kitamura, H., K. Iwakabe, T. Yahata, S. Nishimura, A. Ohta, Y.
Ohmi, M. Sato, K. Takeda, K. Okumura, L. Van Kaer, et al. 1999.
The natural killer T (NKT) cell ligand -galactosylceramide demon-
strates its immunopotentiating effect by inducing interleukin (IL)-12
production by dendritic cells and IL-12 receptor expression on NKT
cells. J. Exp. Med. 189:1121–1128.
35. Van Den Broeke, L.T., E. Daschbach, E.K. Thomas, G. Andringa, and
J.A. Berzofsky. 2003. Dendritic cell-induced activation of adaptive and
innate antitumor immunity. J. Immunol. 171:5842–5852.
36. Fujii, S., K. Shimizu, M. Kronenberg, and R.M. Steinman. 2002. Pro-
longed interferon- producing NKT response induced with -galacto-
sylceramide-loaded dendritic cells. Nat. Immunol. 3:867–874.
37. Mocikat, R., H. Braumuller, A. Gumy, O. Egeter, H. Ziegler, U. Reusch,
A. Bubeck, J. Louis, R. Mailhammer, G. Riethmuller, et al. 2003.
Natural killer cells activated by MHC class I(low) targets prime den-
dritic cells to induce protective CD8 T cell responses. Immunity. 19:
561–569.
38. Van den Eynde, B., B. Lethe, A. Van Pel, E. De Plaen, and T. Boon.
1991. The gene coding for a major tumor rejection antigen of tumor
P815 is identical to the normal gene of syngeneic DBA/2 mice. J. Exp.
Med. 173:1373–1384.
39. Sarma, S., Y. Guo, Y. Guilloux, C. Lee, X.F. Bai, and Y. Liu. 1999.
Cytotoxic T lymphocytes to an unmutated tumor rejection antigen
P1A: normal development but restrained effector function in vivo. J.
Exp. Med. 189:811–820.
40. Guilloux, Y., X.F. Bai, X. Liu, P. Zheng, and Y. Liu. 2001. Optimal
induction of effector but not memory antitumor cytotoxic T lympho-
cytes involves direct antigen presentation by the tumor cells. Cancer
Res. 61:1107–1112.
41. Zheng, P., Y. Guo, Q. Niu, D.E. Levy, J.A. Dyck, S. Lu, L.A. Shei-
man, and Y. Liu. 1998. Proto-oncogene PML controls genes devoted
to MHC class I antigen presentation. Nature. 396:373–376.
42. Kawano, T., J. Cui, Y. Koezuka, I. Toura, Y. Kaneko, K. Motoki, H.
Ueno, R. Nakagawa, H. Sato, E. Kondo, et al. 1997. CD1d-restricted
and TCR-mediated activation of V14 NKT cells by glycosylcer-
amides. Science. 278:1626–1629.
43. Fujii, S., K. Liu, C. Smith, A.J. Bonito, and R.M. Steinman. 2004.
The linkage of innate to adaptive immunity via maturing dendritic cells
in vivo requires CD40 ligation in addition to antigen presentation and
CD80/86 costimulation. J. Exp. Med. 199:1607–1618.
44. Garrett, W.S., L.M. Chen, R. Kroschewski, M. Ebersold, S. Turley, S.
Trombetta, J.E. Galan, and I. Mellman. 2000. Developmental control
of endocytosis in dendritic cells by Cdc42. Cell. 102:325–334.
45. Cui, J., T. Shin, T. Kawano, H. Sato, E. Kondo, I. Toura, Y. Kaneko,
H. Koseki, M. Kanno, and M. Taniguchi. 1997. Requirement for
V14 NKT cells in IL-12-mediated rejection of tumors. Science. 278:
1623–1626.
46. Udono, H., D.L. Levey, and P.K. Srivastava. 1994. Cellular require-
ments for tumor-specific immunity elicited by heat shock protein: Tu-
mor rejection antigen gp96 primes CD8 T cells in vivo. Proc. Natl.
Acad. Sci. USA. 91:3077–3081.
47. Ray, P.K., T.B. Poduval, and K. Sundaram. 1977. Antitumor immu-
nity, V.BCG-induced growth inhibition of murine tumors. Effect of
hydrocortisone, antiserum against theta antigen, and -irradiated BCG.
J. Natl. Cancer Inst. 58:763–767.
48. Dye, E.S., R.J. North, and C.D. Mills. 1981. Mechanisms of anti-
tumor action of Corynebacterium parvum. I. Potentiated tumor-specific
immunity and its therapeutic limitations. J. Exp. Med. 154:609–620.
49. Woodruff, M.F., and J.L. Boak. 1966. Inhibitory effect of injection of
Corynebacterium parvum on the growth of tumour transplants in
isogenic hosts. Br. J. Cancer. 20:345–355.
50. Sinniah, D., J.C. White, A. Omar, and C.P. Chua. 1978. Acute leuke-
mia in Malaysian children. Cancer. 42:1970–1975.
51. Gillessen, S., Y.N. Naumov, E.E. Nieuwenhuis, M.A. Exley, F.S. Lee,
N. Mach, A.D. Luster, R.S. Blumberg, M. Taniguchi, S.P. Balk, et al.
2003. CD1d-restricted T cells regulate dendritic cell function and anti-
tumor immunity in a granulocyte-macrophage colony-stimulating fac-
tor-dependent fashion. Proc. Natl. Acad. Sci. USA. 100:8874–8879.
52. Giaccone, G., C.J. Punt, Y. Ando, R. Ruijter, N. Nishi, M. Peters,
B.M. von Blomberg, R.J. Scheper, H.J. van der Vliet, A.J. van den
Eertwegh, et al. 2002. A phase I study of the natural killer T-cell ligand
-galactosylceramide (KRN7000) in patients with solid tumors. Clin.
Canc. Res. 8:3702–3709.
53. Granucci, F., C. Vizzardelli, E. Virzi, M. Rescigno, and P. Ricciardi-
Castagnoli. 2001. Transcriptional reprogramming of dendritic cells by
differentiation stimuli. Eur. J. Immunol. 31:2539–2546.
54. Huang, Q., D. Liu, P. Majewski, L.C. Schulte, J.M. Korn, R.A.
Young, E.S. Lander, and N. Hacohen. 2001. The plasticity of dendritic
cell responses to pathogens and their components. Science. 294:870–875.
55. Inaba, K., M. Pack, M. Inaba, H. Sakuta, F. Isdell, and R.M. Stein-
man. 1997. High levels of a major histocompatibility complex II - self
peptide complex on dendritic cells from lymph node. J. Exp. Med. 186:
665–672.
56. Wu, D.Y., N.H. Segal, S. Sidobre, M. Kronenberg, and P.B. Chap-
man. 2003. Cross-presentation of disialoganglioside GD3 to natural
killer T cells. J. Exp. Med. 198:173–181.
57. Ferrone, S., and F.M. Marincola. 1995. Loss of HLA class I antigens by
melanoma cells: molecular mechanisms, functional significance and
clinical relevance. Immunol. Today. 16:487–494.
58. Schmieg, J., G. Yang, R.W. Franck, and M. Tsuji. 2003. Superior pro-
tection against malaria and melanoma metastases by a C-glycoside ana-
logue of the natural killer T cell ligand -galactosylceramide. J. Exp.
Med. 198:1631–1641.
59. Wu, D., G.W. Xing, M.A. Poles, A. Horowitz, Y. Kinjo, B. Sullivan,
V. Bodmer-Narkevitch, O. Plettenburg, M. Kronenberg, M. Tsuji, et
al. 2005. Bacterial glycolipids and analogs as antigens for CD1d-
restricted NKT cells. Proc. Natl. Acad. Sci. USA. 102:1351–1356.
60. Berendt, M.J., and R.J. North. 1980. T-cell–mediated suppression of
anti-tumor immunity. An explanation for progressive growth of an im-
munogenic tumor. J. Exp. Med. 151:69–80.
61. North, R.J. 1985. Down-regulation of the antitumor immune re-
sponse. Adv. Cancer Res. 45:1–43.
... It is important that the B lymphocyte receives no help from the classic Tfh cells. In the second case, the NKTfh cell promotes the classic help to the B cell provided by the pMHC II restricted Tfh lymphocyte [93,107]. In this case, the NKTfh cell is, as a rule, activated only by DC via the CD1d restricted mechanism [93,94]. ...
... BIOCHEMISTRY (Moscow) Vol. 84 No. 9 2019 Therefore, depending on the activating signal, B cells can receive help from NKTfh cells either for the for mation of PCs secreting antibodies against a single unique glycolipid/lipid or a protein carrier or the adaptive help due to glycolipid activated DCs and adaptive CD4 + Tfh cells [107]. In this case, NKT cells enhance the effect of Tfh lymphocytes in the transformation of B cells into PCs with subsequent synthesis of antibodies against protein antigens only [93,94]. ...
Article
Full-text available
The review discusses the mechanisms of participation of natural killer T cells (NKT cells) in the induction of antiphospholipid antibodies (APA) that play a major pathogenetic role in the formation of antiphospholipid syndrome (APS), summarizes the data on APS pathogenesis, and presents modern concepts on the antibody formation involving follicular helper type II NK cells.
... Важно, что другой помощи от классических TFH клеток B лимфоцит не получает. Во втором случае NKTFH клетка является усилителем обычной (классической) реакции помощи B клетке со стороны pMHC II рестриктированного TFH лимфоцита [93,107]. При этом активация NKTFH клетки осуществляется, как правило, только DC по CD1d рестриктированному меха низму [93,94]. ...
... Таким образом, в зависимости от активаци онного сигнала В клетки могут получать по мощь от NKTFH клеток либо для формирова ния PC, секретирующих антитела против одно го уникального гликолипида/липида или белко вого носителя, либо адаптивную форму помощи от NKTFH клеток благодаря гликолипид акти вированным DC и адаптивным CD4 + TFH клет кам [107]. В этом случае NKT клетки будут уси ливать эффект TFH лимфоцитов для трансфор мации В клеток в PC с последующим синтезом антител только к белковым антигенам [93,94]. ...
Article
Full-text available
В данном обзоре рассматриваются механизмы потенциального участия натуральных киллерных Т клеток (NKT) в индукции антифосфолипидных антител, играющих основную патогенетическую роль в формировании антифосфолипидного синдрома. На основании литературных данных рассматриваются патогенез антифосфолипидного синдрома и современные аспекты формирования антител с участием фолликулярных хелперных NKT клеток II типа. Представлено несколько потенциальных механизмов участия NKT клеток в индукции антифосфолипидных антител.
... One of the major drawbacks of administering soluble free α-GalCer is that it causes type I NKT cell to adopt an anergic state causing unresponsiveness to sequential stimulation with α-GalCer (205). To circumvent this problem, mice were administrated DCs loaded with either α-GalCer alone or in combination with tumor antigens (180,182,187,190,206). α-GalCer-pulsed APCs induced a more prolonged cytokine response as well as powerful antitumor immune response than α-Galcer alone (180,207). ...
... α-GalCer-pulsed APCs induced a more prolonged cytokine response as well as powerful antitumor immune response than α-Galcer alone (180,207). Another recent immunotherapeutic approach has been to load autologous irradiated tumors, which act as source of tumor antigens with α-GalCer (121,182,187,188). A big improvement of this approach is CD1d-mediated cross-presentation of endogenous glycolipids and or α-GalCer from tumor cells to NKT cells, leading to DC maturation and consequently effective long-term T cell resistance to the tumor (128). ...
Article
Full-text available
Natural killer T (NKT) cells are specialized CD1d-restricted T cells that recognize lipid antigens. Following stimulation, NKT cells lead to downstream activation of both innate and adaptive immune cells in the tumor microenvironment. This has impelled the development of NKT cell-targeted immunotherapies for treating cancer. In this review, we provide a brief overview of the stimulatory and regulatory functions of NKT cells in tumor immunity as well as highlight preclinical and clinical studies based on NKT cells. Finally, we discuss future perspectives to better harness the potential of NKT cells for cancer therapy.
... Therefore, upon uptake of self-antigens in a non-inflammatory context (meaning without any danger signals), these antigens are presented on MHC molecules without increased co-stimulatory signals (Blander, 2007;Bonifaz et al., 2002;Coombes et al., 2007;Hawiger et al., 2001;Kretschmer et al., 2005;Yamazaki et al., 2008). Interestingly, cDC1 have been described to efficiently take up dead cells (Liu et al., 2002(Liu et al., , 2005Qiu et al., 2009;Zelenay et al., 2012). Furthermore, cDC1 regulate CD8 + T cell tolerance by inducing clonal deletion or anergy following cross-presentation (Blander and Nair-Gupta, 2013; Joffre et al., 2012;Luckashenak et al., 2008). ...
Chapter
Dendritic cells (DCs) are major regulators of adaptive immunity, as they are not only capable to induce efficient immune responses, but are also crucial to maintain peripheral tolerance and thereby inhibit autoimmune reactions. DCs bridge the innate and the adaptive immune system by presenting peptides of self and foreign antigens as peptide MHC complexes to T cells. These properties render DCs as interesting target cells for immunomodulatory therapies in cancer, but also autoimmune diseases. Several subsets of DCs with special properties and functions have been described. Recent achievements in understanding transcriptional programs on single cell level, together with the generation of new murine models targeting specific DC subsets, advanced our current understanding of DC development and function. Thus, DCs arise from precursor cells in the bone marrow with distinct progenitor cell populations splitting the monocyte populations and macrophage populations from the DC lineage, which upon lineage commitment can be separated into conventional cDC1, cDC2, and plasmacytoid DCs (pDCs). The DC populations harbor intrinsic programs enabling them to react for specific pathogens in dependency on the DC subset, and thereby orchestrate T cell immune responses. Similarities, but also varieties, between human and murine DC subpopulations are challenging, and will require further investigation of human specimens under consideration of the influence of the tissue micromilieu and DC subset localization in the future.
... NK cells can find numerous stressed cells that have or have not been opsonized by antibodies by producing cytokines and by playing a part in adaptive immune response leading to cell lysis of infected and damaged cells. CD1d Dependent NKT cells are a subset of innate lymphocyte that help in mediating anti-tumor immunity [45]. Various studies have shown that NKt cells can mediate immune suppression and are also responsible for immune suppression [46]. ...
Article
Full-text available
In recent years, cancer microenvironment comprises of several kinds of immune factors associating to the group of tumor environment. The role of mesenchymal stem cells (MSCs) and regulatory T cells (Tregs) play a vital factor in the immune environment. Mesenchymal stem cells are multipotent adult stem cells with immunomodulatory chattels, Recently MSCs are found to be promising candidates for cell based immunosuppression and immuno tolerance. The regulatory T cells or the suppressor T cells are a subpopulation of T cells modulating the immune environment and maintain tolerance to self-antigens. The mechanism intricate in MSCs to inhibit proliferation of proinflammatory T lymphocytes responsible to instigating autoimmune disease have to characterised but the mechanism behind this is the MSCs knack to generate Tregs cells and has been found to enrol foxp3 in maintaining immune homeostasis along with Natural killer cells and dendritic cells.
... An effective therapeutic response against B-cell lymphoma was obtained using a single dose of the novel antibody (Figure 1(b)), while no significant antitumor effect was observed with a single dose α-GalCer administration, as shown by previous studies. 19,32,37,38 In fact, the poor antitumor efficacy of α-GalCer alone in our tumor model mostly resembles the clinical scenario, where patients treated with this glycolipid did not experiment any response although they had a moderate iNKT-cell expansion and activation with increased IFN-γ production. [23][24][25][26] NKT14m antibody induced a potent iNKT-cell activation with a higher increase of IFN-γ production than α-GalCer (Figure 1(c)), suggesting that this antibody could be a better option to improve responses in cancer patients. ...
Article
Full-text available
Invariant natural killer T (iNKT) cells are a small population of T lymphocytes that expresses an invariant T cell receptor with a unique specificity for glycolipid antigens. Their activation using the glycolipid α-galactosylceramide (α-GalCer) triggers innate and adaptive immune responses. The use of α-GalCer in preclinical models as a single antitumor treatment showed moderate effect, but its efficacy in cancer patients was less effective. In addition, this glycolipid induces long-term iNKT-cell anergy precluding the possibility of retreatment. Recently, the first murine iNKT-cell agonistic antibody, NKT14m, has been developed. Here, we analyzed, for the first time, the antitumor efficacy of NKT14m in a B-cell lymphoma model. In a therapeutic setting, a single dose of NKT14m had a moderate antitumor efficacy that was associated with an increase of IFN-γ producing iNKT cells even after a second dose of the NKT14m antibody. Importantly, the combination of a single dose of NKT14m with cyclophosphamide had a potent antitumor efficacy and long-lasting immunity in vivo. Our findings provide the first evidence of the in vivo antitumor efficacy of NKT14m antibody, showing that, either alone or in combination with chemotherapy, induces an effective antitumor response. These results open new opportunities for iNKT-cell mediated immunotherapy to treat B-cell lymphoma.
... At the same time, the maturation and polarization of DCs and monocytes is promoted by iNKT cells (42,43). Several mechanisms could underlie this interplay, including CD1d engagement (44), cytokine production (45), CD40 ligation (46,47), purinergic signaling (48,49). iNKT cell-dependent signaling cues indeed direct the acquisition of either pro-inflammatory or anti-inflammatory effector phenotypes of myeloid cells (50)(51)(52)(53). ...
Article
Full-text available
CD1d-restricted Natural Killer T (NKT) cells are regarded as sentinels of tissue integrity by sensing local cell stress and damage. This occurs via recognition of CD1d-restricted lipid antigens, generated by stress-related metabolic changes, and stimulation by inflammatory cytokines, such as IL-12 and IL-18. Increasing evidence suggest that this occurs mainly upon NKT cell interaction with CD1d-expressing cells of the Mononuclear Phagocytic System, i.e., monocytes, macrophages and DCs, which patrol parenchymatous organs and mucosae to maintain tissue homeostasis and immune surveillance. In this review, we discuss critical examples of this crosstalk, presenting the known underlying mechanisms and their effects on both cell types and the environment, and suggest that the interaction with CD1d-expressing mononuclear phagocytes in tissues is the fundamental job of NKT cells.
... We previously showed that coadministration of OVA protein antigen plus αGalCer elicited both CD4 + and CD8 + T cell responses via DC maturation (30). Similar responses have been demonstrated in some cancer and infectious disease models (31)(32)(33). We elucidated the mechanism of iNKTlicensed DCs for T cell response; activated iNKT cells promote DC maturation via CD40/40L signaling and cytokines (IFNγ and TNFα). ...
Article
Full-text available
Vaccines against a variety of infectious diseases have been developed and tested. Although there have been some notable successes, most are less than optimal or have failed outright. There has been discussion about whether either B cells or dendritic cells (DCs) could be useful for the development of antimicrobial vaccines with the production of high titers of antibodies. Invariant (i)NKT cells have direct antimicrobial effects as well as adjuvant activity, and iNKT-stimulated antigen-presenting cells (APCs) can determine the form of the ensuing humoral and cellular immune responses. In fact, upon activation by ligand, iNKT cells can stimulate both B cells and DCs as via either cognate or non-cognate help. iNKT-licensed DCs generate antigen-specific follicular helper CD4+ T cells, which in turn stimulate B cells, thus leading to long-term antigen-specific antibody production. Follicular helper iNKT cell-licensed B cells generally produce rapid, but short-term antibody. However, under some conditions in the presence of Th cells, the antibody production can be prolonged. With regards to humoral immunity, the quality and quantity of Ab produced depends on the APC type and the form of the vaccine. In terms of cellular immunity and, in particular, the induction of cytotoxic CD8+ T cells, iNKT-licensed DCs show prominent activity. In this review, we discuss differences in iNKT-stimulated APC types and the quality of the ensuing immune response, and also discuss their application in vaccine models to develop successful preventive immunotherapy against infectious diseases.
Thesis
Full-text available
Pour optimiser les stratégies vaccinales anti-tumorales, l’activation des cellules du système immunitaire inné est cruciale pour générer l’expansion des lymphocytes T spécifiques des antigènes tumoraux. Les lymphocytes T Natural Killer invariant (iNKT) représentent une famille unique de lymphocytes T innés ayant des propriétés immunomodulatrices puissantes. Ces cellules reconnaissent via leur récepteur T des antigènes glycolipidiques présentés par la molécule CD1d exprimée par les cellules présentatrices d’antigènes. L'alpha-galactosylcéramide (α-GalCer), un puissant activateur des cellules iNKT, est en développement clinique dans le cancer. Les cellules dendritiques (DCs) sont équipées pour activer les cellules iNKT and promouvoir de puissantes réponses immunitaires adaptatives. Considérant la capacité unique des DC CD8α+ à présenter de façon croisée les antigènes aux lymphocytes T CD8+, notre objectif a visé à délivrer l’α-GalCer (considéré ici comme un adjuvant) et des antigènes tumoraux aux DC CD8α+ dans le but de générer de puissantes réponses T cytotoxiques anti-tumorales. Pour cela, les antigènes ont été incorporés dans des nanoparticules de PLGA décorées à leur surface avec des anticorps anti-Clec9a, un marquer exprimé spécifiquement par les DC CD8α+. Nos résultats montrent chez la souris que la co-délivrance simultanée de l’α-GalCer et d’auto-antigènes tumoraux (Trp2 et gp100) aux DC CD8α+ promeut une forte réponse anti-tumorale dans un contexte prophylactique et thérapeutique. Nous démontrons que cet effet vaccinal est dû aux cellules iNKT (mais pas aux lymphocytes T auxiliaires) et aux lymphocytes T CD8+. L’efficacité vaccinale est corrélée à un rapport supérieur entre les lymphocytes T CD8+ spécifiques des antigènes tumoraux et les lymphocytes T CD4+ régulateurs au sein des tumeurs. Chez l’homme, la co-administration de l’α-GalCer et de l’antigène tumoral (Mélan A) aux DC BDCA3+ (les équivalents humains des DC CD8α+) induit une forte expansion des lymphocytes T CD8+ spécifiques du Mélan-A in vitro. Nos résultats montrent pour la première fois que la tolérance aux auto-antigènes tumoraux peut être levée en exploitant la fonction «helper» des cellules iNKT et mettent en évidence de nouvelles approches thérapeutiques contre le développement tumoral.
Article
Full-text available
Invariant natural killer T (iNKT) cells are an integral component of the immune system and play an important role in antitumor immunity. Upon activation, iNKT cells can directly kill malignant cells as well as rapidly produce cytokines that stimulate other immune cells, making them a front line defense against tumorigenesis. Unfortunately, iNKT cell number and activity are reduced in multiple cancer types. This anergy is often associated with upregulation of co-inhibitory markers such as programmed death-1. Similar to conventional T cells, iNKT cells are influenced by the conditions of their activation. Conventional T cells receive signals through the following three types of receptors: (1) T cell receptor (TCR), (2) co-stimulation molecules, and (3) cytokine receptors. Unlike conventional T cells, which recognize peptide antigen presented by MHC class I or II, the TCRs of iNKT cells recognize lipid antigen in the context of the antigen presentation molecule CD1d (Signal 1). Co-stimulatory molecules can positively and negatively influence iNKT cell activation and function and skew the immune response (Signal 2). This study will review the background of iNKT cells and their co-stimulatory requirements for general function and in antitumor immunity. We will explore the impact of monoclonal antibody administration for both blocking inhibitory pathways and engaging stimulatory pathways on iNKT cell-mediated antitumor immunity. This review will highlight the incorporation of co-stimulatory molecules in antitumor dendritic cell vaccine strategies. The use of co-stimulatory intracellular signaling domains in chimeric antigen receptor-iNKT therapy will be assessed. Finally, we will explore the influence of innate-like receptors and modification of immunosuppressive cytokines (Signal 3) on cancer immunotherapy.
Article
Full-text available
Fragments of foreign antigens associated with class I molecules of the major histocompatibility complex (MHC) are presented at the cell surface to elicit an immune response. This presentation requires the coordinated expression of several genes contained in the MHC, including those encoding the MHC class I heavy chain, the proteins LMP-2 and LMP-7, which are involved in the proteasomal degradation of cytosolic antigens into peptide fragments that are destined for association with MHC class I molecules, and TAP-1 and TAP-2, which transport these fragments across the membrane of the endoplasmic reticulum at the start of their journey to the cell surface. In many virus-transformed cell lines, and spontaneous tumours, these genes are simultaneously repressed. However, the key factor(s) that are essential for their expression and repression have not been identified. Here we report that the proto-oncogene product PML induces expression of LMP-2, LMP-7, TAP-1 and TAP-2 in an MHC-class I-negative, recurrent tumour, leading to the re-expression of cell-surface MHC in tumours and to rejection of the tumours. PML also regulates MHC expression in untransformed fibroblasts. We conclude that malfunction of PML may enable a tumour to evade the immune defence of its host.
Article
Full-text available
MHC class I-restricted tumor antigen can be presented to CD81 T cells by two distinct mechanisms. Direct presentation involves degradation of cytosolic proteins by the proteosome into peptides, transport of the pep- tides across the endoplasmic reticulum membrane, and expression of the MHC-peptide complex on the tumor cell surface. Cross-presentation, on the other hand, involves uptake and intracellular processing of the tumor antigen by host antigen-presenting cells. Whereas it is clear that cross- presentation is necessary and sufficient for the induction of memory CTLs, it has not been tested whether such presentation is sufficient to induce effector CTLs. Here we analyzed the requirements of direct anti- gen presentation for the induction of effector and memory antitumor CTLs using a MHC class I2 mutant incapable of direct antigen presen- tation and its parent, the MHC class I1 J558 cell line. We report that in comparison with the MHC class I1 tumor cell, the MHC class I2 mutant
Article
Full-text available
B7H/B7RP (hereby called B7H) is a new member of the B7 family of costimulatory molecules and interacts with inducible costimulatory molecule (ICOS). Its function for CD8 T cells has not been reported. We report here that expression of B7H on the tumor cells reduced tumorigenicity and induced immunity to subsequent challenge with parental tumor cells. The immune protection correlates with an enhanced cytotoxic T lymphocyte (CTL) response against P1A, the major tumor antigen expressed in the J558 tumor. To understand the mechanism of immune protection, we adoptively transferred transgenic T cells specific for tumor antigen P1A into mice that bore P1A-expressing tumors. We found that while the transgenic T cells divided faster in mice bearing the B7H+ tumors, optimal B7H-induced clonal expansion of P1CTL required costimulation by B7–1 and B7–2 on the endogenous host antigen-presenting cells (APCs). Interestingly, when B7H+ and B7H− tumors were coinjected, P1CTL selectively eliminated the B7H+ tumor cells. Moreover, B7H expressed on the tumor cells made them highly susceptible to destruction by CTL in vivo, even if the CTL was administrated into mice with large tumor burdens. Tumors that recurred in the P1CTL-treated mice lost transfected B7H and/or H-2Ld, the class I molecule that presents the P1A peptide. Taken together, our results reveal that B7H costimulates clonal expansion of, and cognate destruction by CD8+ T lymphocytes in vivo.
Article
Full-text available
The natural killer T (NKT) cell ligand α-galactosylceramide (α-GalCer) exhibits profound antitumor activities in vivo that resemble interleukin (IL)-12–mediated antitumor activities. Because of these similarities between the activities of α-GalCer and IL-12, we investigated the involvement of IL-12 in the activation of NKT cells by α-GalCer. We first established, using purified subsets of various lymphocyte populations, that α-GalCer selectively activates NKT cells for production of interferon (IFN)-γ. Production of IFN-γ by NKT cells in response to α-GalCer required IL-12 produced by dendritic cells (DCs) and direct contact between NKT cells and DCs through CD40/CD40 ligand interactions. Moreover, α-GalCer strongly induced the expression of IL-12 receptor on NKT cells from wild-type but not CD1−/− or Vα14−/− mice. This effect of α-GalCer required the production of IFN-γ by NKT cells and production of IL-12 by DCs. Finally, we showed that treatment of mice with suboptimal doses of α-GalCer together with suboptimal doses of IL-12 resulted in strongly enhanced natural killing activity and IFN-γ production. Collectively, these findings indicate an important role for DC-produced IL-12 in the activation of NKT cells by α-GalCer and suggest that NKT cells may be able to condition DCs for subsequent immune responses. Our results also suggest a novel approach for immunotherapy of cancer.
Article
Full-text available
LIGHT was recently described as a member of the tumor necrosis factor (TNF) 'superfamily'. We have isolated a mouse homolog of human LIGHT and investigated its immunoregulatory functions in vitro and in vivo. LIGHT has potent, CD28-independent co-stimulatory activity leading to T-cell growth and secretion of gamma interferon and granulocyte−macrophage colony-stimulating factor. Gene transfer of LIGHT induced an antigen-specific cytolytic T-cell response and therapeutic immunity against established mouse P815 tumor. In contrast, blockade of LIGHT by administration of soluble receptor or antibody led to decreased cell-mediated immunity and ameliorated graft-versus-host disease. Our studies identify a previously unknown T-cell co-stimulatory pathway as a potential therapeutic target.
Article
Interleukin-12 (IL-12) is a heterodimeric cytokine mediating a dynamic interplay between T cells and antigen-presenting cells (APCs). Preclinical studies have demonstrated that recombinant murine IL-12 (rmIL-12) promotes specific antitumor immunity mediated by T cells in several types of tumors. However, the in vivo antitumor properties of IL-12 in acute myeloid leukemia (AML) have not been previously reported. We show here in a murine AML model that systemic administration of rmIL-12 significantly delays tumor growth but is incapable of rescuing mice from lethal leukemia. In contrast, AML cells genetically modified to express IL-12 (IL12-AML) using murine stem cell virus (MSCV) p40 + p35 elicit very potent antileukemic activity. Vaccines with lethally irradiated IL12-AML cells protect naive mice against challenge with wild-type AML cells and, more importantly, can cure mice bearing a considerable leukemic burden. Immunized mice show no signs of systemic IL-12 toxicity and their spleen histology is comparable with naive mice spleen. In vivo depletion of IL-12, interferon-γ (IFN-γ), or CD8+ T cells after injections with live IL12-AML cells abrogates completely the antileukemia immune responses. Studies on the in vitro effects of IFN-γ on AML cells demonstrate enhanced expression of major histocompatibility complex (MHC) and accessory molecules and induction of the costimulatory molecules B7.1 and B7.2, but no significant direct antiproliferative effect. 51Cr release assays show that rejection of live IL12-AML cells supports the development of long-lasting leukemia-specific cytotoxic T lymphocyte (CTL) activity. In conclusion, our results demonstrate that IL12-AML vaccination is a safe and potent immunotherapeutic approach that has a great potential to eliminate minimal residual disease in patients with AML.
Article
Malignant transformation of melanocytes may be associated with changes in the expression of major histocompatibility complex (MHC) HLA class I antigens. Interest in the characterization of abnormalities in the expression of MHC class I by melanoma cells has been rekindled by the current emphasis on the application of T-cell-based immunotherapy to melanoma. Here, Soldano Ferrone and Francesco Marincola review defects in class I expression as described in melanoma cells, as well as the molecular mechanisms, functional significance and clinical implications of such defects.
Article
We showed previously that mouse mastocytoma P815 expresses several distinct antigens that are recognized by cytolytic T lymphocytes (CTL) of syngeneic DBA/2 mice. Antigens P815A and P815B are usually lost jointly and are targets for immune rejection responses in vivo. We used a cosmid library and a CTL stimulation assay to obtain transfectants expressing tumor rejection antigen P815A. From these transfectants we retrieved gene P1A which transferred the expression of both P815A and B. This gene is unrelated to three previously isolated genes coding for tum-antigens. It encodes a putative protein of 224 amino acids which contains two highly acidic domains showing homology with similar regions of nuclear proteins. The P1A gene expressed by tumor P815 is completely identical to the gene present in normal DBA/2 cells. Expression of the gene was tested by Northern blots. Cells from liver, spleen, and a number of mast cell lines were negative, but mast cell line L138.8A produced a high level of P1A message and was lysed by CTL directed against antigens P815A and B. We conclude that major tumor rejection antigens of P815 are encoded by a gene showing little or no expression in most normal cells of adult mice.
Article
The anti-tumor mechanism in mice induced by a subcutaneous injection of syngeneic tumor cells admixed with Corynebacterium parvum was investigated. When mice were implanted in a hind footpad with x 2 1096) tumor cells admixed with 100 microgram C. parvum, the tumor that emerged grew progressively for about 9 d and then underwent progressive and complete regression. It was found that this C. parvum-induced regression was associated with the acquisition of a systemic, T cell-mediated mechanism of immunity to tumor-specific transplantation antigens, which enabled the host to cause the regression of an untreated test tumor growing simultaneously at a distant site. The generation of a C. parvum-potentiated anti-tumor response was dependent on the presence of tumor cells in close association with C. parvum, tumor immunogenicity, and the quantity of tumor antigen in the admixture. The anti-tumor immunity was specific for the tumor in the therapeutic admixture and could be adoptively transferred to normal recipients with Thy-1.2-positive lymphocytes, but not with serum. Complete regression of a distant test tumor by the C. parvum-tumor admixture was limited to tumors below a certain critical size.