ArticlePDF Available

Concurrent delivery of tumor antigens and activation signals to dendritic cells by irradiated CD40 ligand-transfected tumor cells resulted in efficient activation of specific CD8+ T cells

Authors:

Abstract and Figures

To improve the efficacy of tumor cell-based and dendritic cell (DC)-based cancer vaccines, this study explored the potential of a new cancer vaccine strategy, that is, the use of CD40 ligand-transfected tumor (CD40L-tumor) cells to simultaneously deliver both tumor-derived antigens (Ag) and maturation stimuli to DCs. Materials from frozen/thawed or irradiated human tumor cells, with or without surface CD40L, were internalized efficiently by immature DCs after coincubation. However, during the internalization process, only coculturing with irradiated CD40L-tumor cells resulted in concurrent, optimal DC maturation and production of proinflammatory chemokines and pro-Th1 cytokines, such as IL-6, IL-8, IL-12, IFN-gamma, and TNF-alpha. These activated DCs were the most potent cells to support the growth of CD8+, IFN-gamma-producing T cells, and to process tumor Ag for the generation of specific cytotoxic T cells in vitro. Animals vaccinated with irradiated CD40L-tumor cell-pulsed DCs were better protected against subsequent challenge of a weakly immunogenic tumor cell line than animals vaccinated with irradiated CD40L-tumor cells alone. Thus, our results strongly support the future clinical application of using DCs pulsed with irradiated CD40L-tumor cells as a cancer vaccine.
Content may be subject to copyright.
Concurrent delivery of tumor antigens and activation signals to
dendritic cells by irradiated CD40 ligand-transfected tumor cells
resulted in efficient activation of specific CD8
þ
T cells
Ko-Jiunn Liu, Li-Fan Lu, Hui-Ting Cheng, Yi-Mei Hung, Sheng-Ru Shiou, Jacqueline
Whang-Peng and Shin-Hun Juang
Cancer Research Cooperative Laboratory at National Taiwan University Hospital, Division of Cancer
Research, National Health Research Institutes, Taipei, Taiwan.
To improve the efficacy of tumor cell-based and dendritic cell (DC)-based cancer vaccines, this study explored the potential of a
new cancer vaccine strategy, that is, the use of CD40 ligand-transfected tumor (CD40L-tumor) cells to simultaneously deliver both
tumor-derived antigens (Ag) and maturation stimuli to DCs. Materials from frozen/thawed or irradiated human tumor cells, with or
without surface CD40L, were internalized efficiently by immature DCs after coincubation. However, during the internalization
process, only coculturing with irradiated CD40L-tumor cells resulted in concurrent, optimal DC maturation and production of
proinflammatory chemokines and pro-Th1 cytokines, such as IL-6, IL-8, IL-12, IFN-g, and TNF-a. These activated DCs were the
most potent cells to support the growth of CD8
þ
, IFN-g-producing T cells, and to process tumor Ag for the generation of specific
cytotoxic T cells in vitro. Animals vaccinated with irradiated CD40L-tumor cell-pulsed DCs were better protected against
subsequent challenge of a weakly immunogenic tumor cell line than animals vaccinated with irradiated CD40L-tumor cells alone.
Thus, our results strongly support the future clinical application of using DCs pulsed with irradiated CD40L-tumor cells as a cancer
vaccine.
Cancer Gene Therapy (2004) 11, 135–147. doi:10.1038/sj.cgt.7700663
Published online 28 November 2003
Keywords: dendritic cells; antigen processing; CD40 ligand
D
endritic cells (DCs) are ‘‘professional’’ antigen
presenting cells (APCs) involved in the initiation
and maintenance of both the innate and the adaptive
immune responses.
1
Protective cellular immunity was
induced by vaccination of DCs loaded with various forms
of tumor antigens (Ag) in several animal models.
2–4
Early
clinical trials using tumor Ag-pulsed DCs as vaccines for
the treatment of various advanced malignancies have been
reported.
5–7
Several DC-based clinical trials employed
preidentified tumor-associated Ag (TAA) or synthetic
peptides containing cytotoxic T lymphocyte (CTL)
epitopes of known TAA as the source of Ag. Alternative
approaches have been explored to expand DC-based
therapy to cancers for which appropriate TAA or relevant
CTL epitopes have not been determined. These ap-
proaches include pulsing DCs with tumor cell ly-
sates,
3,4,6,8,9
tumor-derived RNA,
10
necrotic or apoptotic
tumor cells,
11–15
and fusing DCs with tumor cells.
16,17
Gene-modified tumor cell vaccines, which also use
undefined tumor material as the source of tumor Ag,
represent another promising new approach for cancer
therapy. Immunization of animals with tumor cells
transfected with genes coding for costimulatory mole-
cules, cytokines, or chemokines can induce tumor-specific
CTL responses and inhibit the growth of parental tumor
cells.
18–21
One possible mechanism underlying the anti-
tumor effect of gene-modified tumor cell vaccine is that
tumor materials deposited at the injection site are
captured and subsequently presented to specific CTLs
by infiltrating or resident DCs.
22
Thus, tumor cells
transfected with genes that can promote such in vivo Ag
presentation processes are likely to serve as good tumor
vaccines. It has been shown that immunization with CD40
ligand (CD40L)-transfected tumor (CD40L-tumor) cells
can induce a protective immunity against tumor challenge
in animal models.
22–24
CD40L induces DC maturation
and triggers DCs to secrete various cytokines, including
IL-12, which are important in the induction of Th1 and
CTL responses.
25–27
Signaling through CD40 on the
surface of DCs plays an important role in DC migration
from the epidermal tissue to the lymph nodes,
28
and can
enable DCs to activate CTL in the absence of Th cells.
29
Received June 2, 2003.
Address correspondence and reprint requests to: Dr Ko-Jiunn Liu,
Cancer Research Cooperative Laboratory at National Taiwan
University Hospital, Division of Cancer Research, National Health
Research Institutes, 7, Chung-Shan South Road, 100 Taipei, Taiwan.
E-mail: kojiunn@nhri.org.tw
This work was supported by intramural grants from the National
Health Research Institutes, Taipei, Taiwan.
Cancer Gene Therapy (2004) 11, 135147
r
2004 Nature Publishing Group All rights reserved 0929-1903/04 $25.00
www.nature.com
/
cgt
Vaccination with CD40L-tumor cells could potentially
result in the uptake of tumor Ag by infiltrating DCs, and
the concurrent activation of DCs. Thus, the antitumor
immune response to CD40L-tumor cells could be more
potent than the one to unmodified tumor cells. However,
with this approach, it is difficult to optimally control and
monitor the efficiency of Ag loading and activation of
DCs in vivo. To overcome this drawback and utilize the
benefit of CD40L signaling, we incorporated the gene-
modified tumor cells with the DCs prior to vaccination.
In this study, we have used an in vitro system to
investigate the effect of coculturing human immature DCs
with different forms of human CD40L-tumor cells. By
using CD40L-tumor cells to deliver both tumor Ag and
stimulating signals to DCs during in vitro vaccine
preparation, we had better control of Ag loading and
activation of DCs. DCs cultured with irradiated CD40L-
tumor cells were able to process and present tumor Ag,
secrete substantial amounts of various pro-Th1 and
proinflammatory cytokines, and promote the growth
and generation of specific CD8
þ
T cells. When CD40L-
tumor cell-vaccinated animals were compared to animals
receiving DCs pulsed with irradiated CD40L-tumor cells,
the latter were found to be more resistant to subsequent
challenge with a weakly immunogenic B16-F1 tumor line.
Our results support DCs pulsed with irradiated CD40L-
transfected tumor cells as a candidate vaccine strategy in
human clinical studies and may provide the foundation
for the design of a better DC-based cancer vaccine.
Materials and methods
Human tumor transfectant expressing CD40L
A lung cancer cell line was previously established in our
laboratory from the pleural effusion of a lung cancer
patient. The lung cancer cells were transfected with either
an empty pcDNA3.1/Zeo() vector or a vector carrying
the human CD40L sequence and selected with 200 mg/ml
zeocin (Invitrogen, San Diego, CA). The expression of
CD40L by transfected tumor cells was determined by
staining with a PE-anti-CD154 mAb (BD PharMingen,
San Diego, CA). Cells expressing surface CD40L were
sorted using a FACS Vantage cell sorter (Becton
Dickinson, Mountain View, CA) and cloned. All cell
lines used in this study were negative for mycoplasma
contamination as determined by a nested PCR-based
mycoplasma detection kit (ATCC, Rockville, MD).
Preparation of human DCs
Peripheral blood mononuclear cells (PBMCs) from
healthy donors (obtained from the Chinese Blood
Foundation) were enriched by density-gradient centrifu-
gation with Ficoll-Hypaque (Amersham Pharmacia Bio-
tech, Uppsala, Sweden). Cells at the interface were
collected, washed and suspended in serum-free AIM-V
medium (Life Technologies, Rockville, MD). Cells were
plated into plastic tissue culture dishes (25 10
6
cells/
dish) and incubated at 371C for 2 hours. Adherent
mononuclear cells were cultured with RPMI-1640 med-
ium with 10% fetal bovine serum (FBS) in the presence of
1000 U/ml recombinant human IL-4 (Peprotech, Rock
Hill, NJ) and 500 U/ml granulocyte–macrophage colony-
stimulating factor (GM-CSF) (a gift from Kirin Brewery
Co., Japan). After 7 days, nonadherent and loosely
adherent cells were collected and used as the source of
immature DCs in the subsequent experiments.
Flow cytometric analysis
Cells to be analyzed for the expression of surface markers
were stained with different fluorescence-labeled mAb and
then analyzed using a flow cytometer (EPICS XL-MCL,
Beckman Coulter, Fullerton, CA). The mAbs used in this
study were as follows: flourescein isothiocyanate (FITC)-
anti-HLA-DR (Immunotech, Marseille Cedex, France),
phycoerythrin (PE)-anti-CD86 (Immunotech), FITC-
anti-CD80 (Immunotech), PE-anti-CD83 (BD PharMin-
gen), and PE-anti-CD11c (Immunotech). Isotype-
matched control mAb were obtained from BD PharMin-
gen and Caltag (Burlingame, CA).
Internalization of tumor materials by DCs
Tumor cells were labeled with a green fluorescent dye,
PKH67 (Sigma, St. Louis, MO) according to the
manufacturer’s instruction, and treated with one cycle
of freezing/thawing to induce necrosis or irradiation
(12,000 rad) to initiate apoptosis. One cycle of freezing/
thawing resulted in mostly intact cells that were stained
positive with trypan blue. Irradiation (12,000 rad) in-
itiated a slowly progressing apoptosis, as 10–20% cells
were stained positive with FITC-annexin V (Caltag) after
16 hours. DCs (1 10
6
cells) were cultured with equal
numbers of frozen/thawed or irradiated dye-labeled
tumor cells at 371C for 16 or 40 hours. The mixture of
cells was collected, and DCs were counterstained with a
PE-anti-CD11c mAb (Immunotech) or a PE-anti-
CD83 mAb (BD PharMingen). The internalization of
green fluorescent tumor materials by DCs was evaluated
with a fluorescent microscope (BX50, Olympus, Tokyo,
Japan) or a flow cytometer (EPICS XL-MCL, Beckman
Coulter).
Induction of maturation and activation of DCs
Immature DCs (2.5 10
5
cells/well) were cultured in a 24-
well plate with equal numbers of frozen/thawed or
irradiated tumor cells with or without CD40L expression.
Cells were collected after 16 or 40 hours and analyzed for
surface expressions of CD11c, CD83, CD80, CD86, and
HLA-DR by flow cytometry, or for the induction of an
allogeneic mixed leukocyte response (MLR). In some
experiments, the culture supernatants were harvested and
assayed for cytokine production.
Detection of cytokine production
The production of IL-12p70, IL-1b, IL-6, IL-8, IL-10,
and TNF-a in the culture supernatant was detected by a
Human Inflammation Cytometric Bead Array (CBA) Kit
Antigen loading and concurrent activation of DC
K-J Liu et al
136
Cancer Gene Therapy
(BD PharMingen). Briefly, beads of different FL-3
fluorescent intensities were preconjugated with mAb
against various cytokines by the manufacturer. These
beads were incubated with 50 ml of culture supernatants
(or cytokine standards) and PE-labeled mAb against
various cytokines at room temperature for 3 hours. Beads
were then subjected to flow cytometric analysis. The
amount of a given cytokine in the culture supernatant was
calculated by converting the intensity of PE fluorescence
to concentration after comparing to results obtained with
the cytokine standards. The production of IFN-g in the
culture supernatant was detected by a sandwich-ELISA
using an IFN-g-OptEIA set (BD PharMingen).
Allogeneic MLR
Serial diluted DCs, after coculturing with tumor cells,
were irradiated (3000 rad) and cultured with allogeneic,
nonadherent PBMCs (10
5
cells/well) in 96-well plates for 6
days. In all, 1 mCi of
3
H-TdR (Amersham Pharmacia
Biotech) was added to the culture for the last 16 hours.
The uptake of
3
H-TdR by the proliferating cells was then
determined by a liquid scintillation counter (Packard
BioScience, Downers Grove, IL).
T-cell surface marker, secreted and intracellular
cytokine analysis
DCs were cultured with different forms of tumor cells for
40 hours. After washing, tumor material-pulsed DCs
(2.5 10
5
cells/well) were cultured with allogeneic, non-
adherent PBMCs (2.5 10
6
cells/well) for 6 days.
Proliferating T cells were expanded with 20 U/ml IL-2
(Chiron, Amsterdam, The Netherlands) and cultured for
another 6 days. Cells were then collected and stained with
an FITC-anti-CD8 mAb and a PE-anti-CD3 mAb (Im-
munotech). The forward scatter (FSC) and the side scatter
(SSC) were used to gate on small lymphocytes for
subsequent analysis. For intracellular cytokine staining,
proliferating T cells were collected, washed and stimu-
lated with 20 ng/ml phorbol 12-myristate-13-acetate
(PMA) and 1000 ng/ml ionomycin (Sigma) in the presence
of GolgiStop (BD PharMingen) for 6 hours. Cells were
stained with an FITC-anti-CD8 mAb. After washing, cells
were fixed, permeabilized with Cytofix/Cytoperm (BD
PharMingen), and then stained with a PE-anti-IFN-g
mAb (BD PharMingen). The expression of intracellular
IFN-g by CD8
þ
T cells was analyzed with a flow
cytometer. Supernatants from day 6 and day 12 cultures
were collected and analyzed for cytokine secretion with a
human Th1/Th2 CBA kit (BD PharMingen).
Evaluation of T-cell responses
Immature DCs were generated from PBMCs of normal
donors with the HLA-A*0201 genotype and cultured with
equal numbers of tumor cells for 16 hours. After washing,
tumor cell material-pulsed DCs (1.5 10
5
cells/well) were
cultured with autologous CD8-enriched T cells (1.5 10
6
cells/well) in RPMI medium supplemented with 10%
human AB-type serum (BioWhittaker, Walkersville, MD)
in a 24-well plate. Autologous CD8-enriched T cells (80%
pure) were prepared by removing adherent cells and
CD4
þ
T cells with anti-CD4-coated magnetic beads and
a Magnetic Particle Concentrator (Dynal Biotech, Oslo,
Norway). IL-2 (20 U/ml) was added into the culture on
day 1, 3, and 6. On day 10, the T cells were restimulated
with irradiated autologous PBMCs pulsed with 50 mg/ml
of an HLA-A*0201-restricted HER2 peptide (HER2, 369-
377:KIFGSLAFL, Research Genetics, Huntsville, AL).
30
IL-2 (20 U/ml) was added into the culture on days 11 and
14. On day 17, T cells (1 10
5
cells/well) were stimulated
with irradiated (8000 rad) T2 cells (5 10
4
cells/well)
pulsed in advance for 3 hours with 50 mg/ml of the HER2
peptide or a control HLA-A*0201-restricted, HPV-16 E6
peptide (E6, 18-26:KLPQLCTEL, Research Genetics).
31
The secretion of IFN-g in the culture supernatant was
determined after 48 hours using a human Th1/Th2 CBA
kit (BD PharMingen). The number of viable cells at the
beginning (day 0) and the end of each stimulation
procedure (days 10 and 17) was determined by the trypan
blue exclusion assay. The cytotoxicity was determined by
a europium-ligand release assay (Wallac, Turku, Fin-
land). The counts of culture with unlabeled T2 cells were
subtracted from those of cultures with peptide-pulsed T2
cells before calculating the specific lysis.
Animal experiments
Murine DCs were generated by culturing bone marrow
cells with RPMI-1640 medium supplemented with 10%
FBS, 500 U/ml of murine GM-CSF, and IL-4 (Peprotech)
for 7 days as described.
26
CT-26 or B16-F1 tumor cells
(ATCC, Rockville, MD) were transfected with either a
control vector or a vector carrying the murine CD40L
gene. Transfected cells were selected and cloned similarly
to methods described above. Day-7 DCs were cocultured
with irradiated (20,000 rad) CD40L-tumor cells for 48
hours at a one-to-one ratio. Mice (n ¼ 5) were immunized
(s.c.) weekly with PBS, DC alone (1 10
6
cells), irradiated
CD40L-tumor alone (1 10
6
cells), or DCs (1 10
6
cells)
pulsed with irradiated CD40L-tumor cells. At 2 weeks
after the third immunization, C57BL/6 or BALB/c mice
were challenged (s.c.) with wild-type B16-F1 cells (1 10
4
cells) or CT-26 cells (2 10
4
cells), respectively. B16-F1
cells were treated with IFN-g 16 hours before injection to
induce the expression of MHC class I molecules. The
growth of tumor was monitored every 2–3 days. The size
of tumor was determined as follows: short diameter
2
long diameter 1/2. Mice were killed when the tumor
volume exceeds 2 cm
3
. This experiment was repeated once
using the same conditions.
Results
DCs can efficiently internalize tumor materials from
frozen/thawed or irradiated tumor cells transfected with
CD40L
Human cells (because they are most relevant to future
clinical applications) were investigated initially in an in
Antigen loading and concurrent activation of DC
K-J Liu et al
137
Cancer Gene Therapy
vitro system. The cDNA for human CD40L was
transfected into a human lung cancer cell line (Fig 1a)
to generate tumor cells that can both provide tumor
antigenic materials and DC stimulatory signals. Immature
DCs were generated by culturing adherent PBMCs with
GM-CSF and IL-4 for 7 days. They expressed a typical
surface staining phenotype of immature DCs (HLA-
DR
þ
, CD3
, CD11c
þ
, CD14
, CD20
, CD80
þ
,
CD83
, and CD86
þ
) and can be induced to express a
mature DC phenotype (CD86
high
and CD83
þ
) after
incubation with 1000 U/ml TNF-a for 40 hours (Fig 1b
and data not shown). To determine the capacity of
immature DCs to internalize tumor materials after
different treatments, tumor cells, with or without surface
CD40L (CD40L-tumor or vector-tumor cells, respec-
tively), were labeled with a green fluorescent dye (PKH67)
and treated with one cycle of freezing/thawing or
irradiation (12,000 rad). Irradiation at this dose initiated
only a slowly progressing apoptosis in the tumor cells, as
about 13 and 41% of cells were stained positive with
FITC-annexin V after 16 and 40 hours, respectively (Fig
1c). These treated tumor cells were then cultured with
equal numbers of immature DCs. After 16 hours, a
significant amount of green fluorescent material was
observed in CD11c
þ
DCs cultured with various forms of
tumor cells (Fig 2). These results indicate that immature
DCs were capable of internalizing antigenic materials
from tumor cells with or without CD40L expression on
the surface. Similar results were observed after cocultur-
ing for 40 hours (data not shown). Flow cytometric
analysis revealed that 65% (64/99) of all CD11c
þ
DCs
contained green fluorescent tumor materials (Fig 3a, left
panel) after coculturing with frozen/thawed CD40L-
tumor cells for 40 hours. Only a few intact tumor cells
(CD11c
PHK67
þ
) remained in the culture (the small
debris was not gated for analysis). In contrast, after
coculturing DCs with irradiated CD40L-tumor cells for
40 hours, 40% (28/70) of CD11c
þ
DCs contained green
fluorescent tumor materials, while a significant percentage
(about 29% of all gated cells) of tumor cells
(CD11c
PKH67
þ
) was still present in the culture (Fig
3a, right panel). These results indicate that immature DCs
appeared to engulf tumor materials more efficiently when
CD40L-tumor cells were treated with freezing/thawing. It
is likely that coculture with irradiated CD40L-tumor cells
led to maturation of immature DCs and thus down-
regulated their capacity to internalize tumor materials.
There were only a few necrotic cells (o5%) in the culture
Figure 1 (a) The expression of surface CD40L on transfected tumor cells was analyzed by flow cytometry. Upper panel: vector-tumor cells.
Lower panel: CD40L-tumor cells. Dotted line: cells stained with a PE-isotype-matched control mAb. Solid line: cells stained with a PE-anti-CD40L
mAb. (b) Immature DCs were generated by culturing adherent PBMCs with GM-CSF and IL-4 for 7 days. Cells were further cultured in the
presence (lower four panels) or absence (upper four panels) of TNF-a for 48 hours. The cell surface expression of HLA-DR, CD86, CD80, and
CD83 was determined by flow cytometry with specific mAb (solid line). Dotted line: isotype-matched control mAb. (c) CD40L-tumor cells were
irradiated (12,000 rad) and then cultured for 16 or 40 hours. Untreated tumor cells or irradiated cells after culture were collected and stained with
FITC-annexin V and PI, and analyzed by flow cytometry.
Antigen loading and concurrent activation of DC
K-J Liu et al
138
Cancer Gene Therapy
Figure 2 Vector-tumor (left four panels) or CD40L-tumor (right four
panels) cells were labeled with a green fluorescent dye, PKH67,
washed extensively and then treated with either one cycle of
freezing/thawing (frozen/thawed) or irradiation (irradiated). Dye-
labeled tumor cells were then cocultured with equal numbers of
immature DCs for 16 hours. The mixture of cells was collected and
DCs were counterstained with a PE-anti-CD11c mAb. The inter-
nalization of green fluorescent tumor materials by CD11c
þ
DCs was
examined using a fluorescent microscope. Image from the same cell
was displayed with filters for green (a, c, e, and g) or red (b, d, f, and
h) fluorescence. One representative result from three separate
experiments is shown.
Figure 3 (a) Immature DCs were cocultured with equal numbers of
PKH67-labeled, frozen/thawed (left panel) or irradiated (right panel)
CD40L-tumor cells for 40 hours. The mixture of cells was collected
and DCs were counterstained with a PE-anti-CD11c mAb. The
numbers in each quadrant represent the percentage of cells after
gating. One representative result from three separate experiments is
shown. (b) Immature DCs were cocultured with equal number of
frozen/thawed or irradiated vector-tumor (closed bars) or CD40L-
tumor (open bars) cells for 16 or 40 hours. The mixture of cells was
then collected and stained for the expression of HLA-DR and CD83.
The percentage of CD83
þ
cells in all cells positive for HLA-DR is
shown. (c, d) Immature DCs were cocultured with equal numbers of
irradiated, PKH67-labeled vector-tumor (closed bars) or CD40L-
tumor (open bars) cells for 4, 16, or 40 hours. The mixture of cells
was collected and DCs were counterstained with a PE-anti-CD11c
mAb or a PE-anti-CD83 mAb. The percentage of CD11c
þ
(c) or
CD83
þ
(d) cells from all cells associated with PHK67 green
fluorescence was determined. One representative result from two
separate experiments is shown.
Antigen loading and concurrent activation of DC
K-J Liu et al
139
Cancer Gene Therapy
up to 40 hours after irradiation (Fig 1c). Therefore, under
this condition, necrotic cells should not have a major role
in the uptake of tumor materials by DCs and the
induction of DC maturation.
Only irradiated CD40L-tumor cells can efficiently
induce the concurrent expression of CD83 on DCs
during the internalization of tumor materials
Ligation of surface CD40 on DCs can trigger their
maturation and activation. As shown in Figure 3b, less
than 25% of HLA-DR
þ
DCs expressed CD83 after
coculturing with frozen/thawed vector-tumor or CD40L-
tumor cells. Coculturing immature DCs with irradiated
CD40L-tumor cells, but not vector-tumor cells, resulted
in a dramatic induction of CD83 expression. Around 40%
of DR
þ
cells were CD83
þ
after 16 hours, while more
than 90% DR
þ
cells were CD83
þ
after 40 hours. These
results suggest that only coculturing immature DCs with
irradiated CD40L-tumor cells can lead to concurrent
internalization of tumor materials and maturation of
DCs. To simultaneously monitor these two events,
immature DCs were cocultured with irradiated, green
fluorescent dye-labeled tumor cells for 4, 16, or 40 hours.
The mixture of cells was collected and the distribution of
internalized green tumor material in DCs expressing
CD11c (all DCs) or CD83 (mature DCs) was evaluated.
Around 20% of green fluorescent material was associated
with CD11
þ
DC after 4 hours of coculture with
irradiated vector-tumor or CD40L-tumor cells, while
60–70% of green fluorescent tumor material was asso-
ciated with CD11c
þ
DC after 16 and 40 hours (Fig 3c).
The proportion of green fluorescent tumor material
associated with CD83
þ
DCs after culturing with irra-
diated CD40L-tumor cells for 4, 16, or 40 hours was
similar to that associated with CD11c
þ
DCs (Fig 3c and
d, open bars). However, less than 20% of green
fluorescent tumor material from irradiated vector-tumor
cells was associated with CD83
þ
DC even after 40 hours
(Fig 3d, closed bars). These results demonstrate that after
coculturing immature DCs with irradiated CD40L-tumor
cells, a significant amount of tumor material was
internalized by CD11c
þ
DCs, and that most of these
DCs also became CD83
þ
within 16 hours. Coculturing
immature DCs with irradiated vector-tumor cells also
resulted in efficient internalization of tumor materials into
CD11c
þ
DCs, but the majority of them remained CD83
even after 40 hours.
Only irradiated CD40L-tumor cells induce a significant
secretion of pro-Th1 and pro-inflammatory cytokines by
DCs
DCs are known to dictate and modulate immune
responses by releasing various cytokines and chemokines.
A significant level of IL-12p70, in addition to substantial
amounts of IFN-g and several proinflammatory cytokines
including IL-6 and TNF-a, was detected only in the
culture containing DCs and irradiated CD40L-tumor cells
after 40 hours (Fig 4). Significant levels of IL-8 were
present under all four culture conditions, and the highest
level was detected in the culture with irradiated CD40L-
tumor cells. A low level of IL-10 and IL-1b was detected
in the culture containing DCs stimulated with irradiated
CD40L-tumor cells. These results indicate that cocultur-
ing immature DCs with irradiated CD40L-tumor cells
drives the activation of DCs and the production of several
pro-Th1 and proinflammatory cytokines. This cytokine
and chemokine profile should foster the generation of an
effective antitumor CTL response.
DCs cultured with irradiated CD40L-tumor cells induce
a much stronger primary allogeneic MLR
We then evaluated the T-cell stimulatory function of these
DCs. DCs cultured with irradiated CD40L-tumor cells
clearly stimulated a much stronger allogeneic MLR than
those cultured with frozen/thawed CD40L-tumor cells or
vector-tumor cells (Fig 5a and c). This result further
demonstrated that irradiated CD40L-tumor cells were the
most effective stimulus of DC maturation in our
experimental setting. Nevertheless, DCs cultured for 16
or 40 hours with frozen/thawed or irradiated tumor cells,
with or without CD40L expression, stimulated allogeneic
MLR equally well in the presence of LPS (Fig 5b and d).
These results suggest that frozen/thawed or irradiated
vector-tumor cells and frozen/thawed CD40L-tumor cells
failed to provide necessary maturation stimuli for
immature DCs, but they did not interfere with the ability
of immature DCs to undergo maturation in the presence
of a valid DC maturation signal such as LPS.
DCs cultured with irradiated CD40L-tumor cells
preferentially promote the growth of CD8
þ
T cells
To further investigate the influence of tumor material-
pulsed DCs on the growth of T cells, DCs were cultured
with tumor cells after different treatments as described
above. After 40 hours, DCs were collected, washed and
cultured with allogeneic T cells. T cells were harvested and
examined on day 12, whereas the culture supernatant was
Figure 4 Immature DCs were cocultured with equal numbers of
frozen/thawed vector-tumor (vector/F), CD40L-tumor (CD40L/F),
irradiated vector-tumor (vector/R), or CD40L-tumor (CD40L/R) cells
for 40 hours. The culture supernatants from each culture were
collected and assayed for the production of IL-1b, IL-6, IL-8, IL-10,
IL-12p70, and TNF-a with a CBA kit. The production of IFN-g was
detected by an ELISA. The detection limits (pg/ml) are: IFN-g, 3.6;
TNF-a, 3.7; IL-1b, 7.2; IL-6, 2.5; IL-8, 3.6; IL-10, 3.3, and IL-12p70,
1.9. The symbol (*) indicates data exceeding the upper range of the
standard (5000 pg/ml). One representative result from three separate
experiments is shown.
Antigen loading and concurrent activation of DC
K-J Liu et al
140
Cancer Gene Therapy
collected on day 6 and day 12 for cytokine analysis. The
highest number of T cells was recovered from the culture
stimulated by DCs pulsed with irradiated CD40L-tumor
cells, and more than 40% (37/92) of all CD3
þ
T cells in
the culture were CD8
þ
(Fig 6a). The majority (478%,
29/37) of these CD8
þ
T cells expressed IFN-g after
stimulation with PMA and ionomycin, indicating that
they are in a rather activated state. The percentage of
CD8
þ
T cells of all CD3
þ
T cells in the other three
groups was lower than 13% (8/96, 5/94, and 12/96). These
results strongly suggest that DCs cultured with irradiated
CD40L-tumor cells can efficiently promote the growth of
CD8
þ
, IFN-g producing T cells. Similar results were also
observed in the experiments using CD45RO-depleted
autologous or allogeneic T cells (data not shown). Some
cells expressed lower levels of CD3 and CD8. These cells
were probably the CD56
þ
cells and thus were not gated
for analysis. Examination of the cytokine environment
during T-cell stimulation indicated that the culture
supernatant of T cells plus DCs pulsed with irradiated
CD40L-tumor cells contained the highest level of IFN-g
and TNF-a, and the lowest level of IL-5 (Fig 6b). This
cytokine profile should promote the development of Th1
and CTL responses. In contrast, the cytokine environ-
ment of the other three groups is more likely to favor a
Th2 response (Fig 6b).
DCs cultured with irradiated CD40L-tumor cells
generate more Ag-specific T cells in vitro
It has been reported that the proto-oncogene HER2 was
overexpressed in many cancers including the non-small-
cell lung cancer.
32
The parental and transfected human
lung cancer cell lines used in this study also expressed
similar levels of HER2 on the surface (data not shown).
To study the cross-presentation and T-cell stimulation
function of tumor material-pulsed DCs, we have exam-
ined the ability of DCs to process tumor materials and
Figure 5 Immature DCs were cultured with equal numbers of frozen/
thawed (open symbols) or irradiated (closed symbols), vector-tumor
(triangles), or CD40L-tumor (circles) cells for 16 hours (a, b) or 40
hours (c, d), with (b, d) or without (a, c)1mg/ml LPS. Cells were then
collected, washed and irradiated (3000 rad). Two-fold serial diluted
cells were cultured with allogeneic PBMCs (10
5
cells/well) in 96-well
plates for 6 days. The proliferation of allogeneic PBMCs was
determined in a thymidine incorporation assay.
Figure 6 DCs were cocultured with equal numbers of frozen/thawed
vector-tumor (vector/F), CD40L-tumor (CD40L/F), irradiated vector-
tumor (vector/R), or CD40L-tumor (CD40L/R) cells for 40 hours.
After washing, DCs were cultured with allogeneic nonadherent
PBMCs for 6 days. Proliferating T cells were expanded by adding IL-
2 and cultured for another 6 days. Cells were then collected, counted
and stained with an FITC-anti-CD8 mAb and a PE-anti-CD3 mAb.
The number of cells obtained from each culture is: vector/F, 2 10
6
vector/R, 2.2 10
6
; CD40L/F, 2.6 10
6
, and CD40L/R, 3.5 10
6
.
For intracellular cytokine staining, proliferating T cells were collected
and stimulated with PMA and ionomycin in the presence of GolgiStop
for 6 hours. Cells were stained with an FITC-anti-CD8 mAb. After
washing, cells were fixed, permeabilized, and then stained with a PE-
anti-IFN-g mAb. Expression of intracellular IFN-g by CD8
þ
T cell was
analyzed with a flow cytometer (a). Supernatants from day 6 and day
12 cultures were collected and analyzed for cytokine secretion (b).
Antigen loading and concurrent activation of DC
K-J Liu et al
141
Cancer Gene Therapy
generate HER2-specific T cells in vitro. DCs were
obtained from normal donors with the HLA-A*0201
genotype and cultured with tumor cells after different
treatments for 16 hours. These DCs were washed,
irradiated, and then cultured with autologous (HLA-
A*0201) CD8-enriched T cells in the presence of human
AB-type serum. After 10 days, T cells were restimulated
with irradiated autologous PBMCs pulsed with an HLA-
A*0201-restricted HER2/369-377 peptide to enrich for
HER2-specific T cells. Surviving T cells on day 17 were
collected and stimulated with T2 cells pulsed with the
HER2/369-377 peptide or a control HPV E6 peptide. We
have found that only T cells initially stimulated with DCs
cultured with either irradiated vector-tumor or CD40L-
tumor cells were able to secrete a significant amount of
IFN-g in response to T2 cells pulsed with the HER2/369-
377 peptide (Fig 7a). These results indicate that DCs
cultured with irradiated tumor cells can process inter-
nalized tumor materials and present the HER2/369-377
epitope to autologous T cells. T cells initially stimulated
with DCs pulsed with frozen/thawed tumor cells secreted
little IFN-g. Therefore, it is not likely that the second
stimulation with peptide-pulsed autologous PBMCs was
priming, instead of boosting, the HER2-specific response.
The cell numbers from all four groups decreased in the
first 10 days of stimulation probably due to the death of
nonspecific autologous T cells. However, after two in vitro
stimulations, a much higher number (44-fold) of T cells,
initially primed by DCs pulsed with irradiated CD40L-
tumor cells, were recovered (Fig 7b). In addition, after
four in vitro stimulations, significant cytotoxicity of these
CD8
þ
T cells against T2 cells pulsed with the HER2/369-
377 peptide could be detected (Fig 7c). In contrast, the
number of T cells from the other three groups declined
rapidly during the first two in vitro stimulations and these
T cells died out during the subsequent stimulations. These
results are consistent with our previous observations and
indicate that DCs pulsed with irradiated CD40L-tumor
cells are superior in supporting the growth of specific
CD8
þ
T cells during in vitro culture.
Animals vaccinated with DCs pulsed with irradiated
CD40L-tumor cells were more resistant to subsequent
challenge of a weakly immunogenic tumor
Our in vitro results with human cells suggested that DCs
pulsed with irradiated CD40L-tumor cells can serve as a
potent vaccine to generate tumor-specific T-cell responses.
An animal model was then established to further compare
the efficacy of tumor cell-based vaccine with cancer
vaccine consisting of DCs pulsed with irradiated CD40L-
tumor cells. Similar to results described above, upregula-
tion of CD86 and secretion of IFN-g and IL-12 were
Figure 7 Immature DCs were generated from an HLA-A*0201 donor
and cultured with equal numbers of frozen/thawed vector-tumor
(vector/F), CD40L-tumor (CD40L/F), irradiated vector-tumor (vector/
R), or CD40L-tumor (CD40L/R) cells for 16 hours. Tumor material-
pulsed DCs were collected and incubated with autologous CD8-
enriched T cells. On day 10, T cells were restimulated with
autologous PBMC pulsed with 50 mg/ml of an HLA-A*0201-restricted
HER2/369-377 peptide. On day 17, cells were collected and
counted. T cells were then incubated with irradiated T2 cells pulsed
with 50 mg/ml of the HER2/369-377 peptide (closed bars) or an HLA-
A*0201-restricted, HPV-16 E6 peptide (open bars). (a) The secretion
of IFN-g in the culture supernatant by T cells on day 19 was
determined. (b) T-cell numbers were determined at the beginning
(day 0) and the end of the two stimulation cycles (days 10 and 17).
(c) T2 cells were labeled with a fluorescence enhancing ligand and
then pulsed at 371C for 2 hours without peptides, with 50 mg/ml of the
HER2 peptide, or two control CEA and MET peptides. Different
numbers of effector T cells were incubated with labeled T2 cells (10
4
cells/well) for 4 hours at 371C. The counts of culture with unlabeled
T2 cells were subtracted from those of cultures with peptide-pulsed
T2 cells before calculating the specific release. One representative
result from two separate experiments is shown.
Antigen loading and concurrent activation of DC
K-J Liu et al
142
Cancer Gene Therapy
observed after culturing bone marrow-derived DCs with
irradiated B16-F1 or CT-26 tumor cells transfected with
murine CD40L (data not shown). Mice immunized with
PBS or DCs alone developed tumors quickly after
challenging with the wild-type tumor cells (Fig 8a,b,e,f).
For the more immunogenic CT-26 tumor, vaccination of
either irradiated CD40L-tumor or DCs pulsed with
irradiated CD40L-tumor both provided protection in
the majority of the mice (Fig 8g and h). However, for the
weakly immunogenic B16-F1 tumor, immunization with
DCs pulsed with irradiated CD40L-B16-F1 tumor cells
(Fig 8d) was better than vaccination with irradiated
CD40L-B16-F1 tumor cells alone (Fig 8c) in inhibiting or
delaying the growth of tumor. Vaccination with DCs
pulsed with irradiated vector-tumor cells failed to provide
such additional protection in both tumor models (data
not shown). These results suggested that for certain
weakly immunogenic tumors, the response to tumor
vaccine consisting of DCs pulsed with irradiated
CD40L-tumor cells might be more protective than that
to tumor vaccine consisting of just the irradiated CD40L-
tumor cells alone.
Discussion
DC-based clinical trials for advanced cancers with partial
effectiveness have been reported.
6,17,33,34
One aspect to
improve current DC-based vaccines could be the source
and format of tumor Ag. Both necrotic and apoptotic
tumor cells have been explored as the source of Ag.
However, the consequences of Ag presentation by DCs
after internalization of materials from necrotic or
apoptotic cells may be different. It has been proposed
that immature DCs that have taken up Ag from apoptotic
cells during normal tissue turnover may induce anergy in
T cells or generate regulatory T cells as a mechanism of
peripheral tolerance.
35
Nevertheless, some studies demon-
strated that DCs can acquire Ag from apoptotic cells and
induce Ag-specific T-cell responses.
12,14,15
Both apoptotic
cells and lysate or supernatant of necrotic cells have been
shown to induce DC maturation in some reports.
36,37
DCs
exposed to apoptotic cells mediated by virus infection or
in the presence of inflammatory stimuli may undergo
maturation.
38,39
Rovere et al have demonstrated that a
higher number of apoptotic cells may trigger DC
maturation and increase their ability to crossactivate T
cells.
13,40
It has been shown that the maturation of DCs is
better stimulated by necrotic tumor cells than by
apoptotic tumor cells or primary tissue cells, probably
due to the secretion of proinflammatory mediators or
heat-shock proteins during necrosis.
36,41
However, in
other reports, immature DCs were not matured after
coculturing with tumor cell lysate or supernatant of
necrotic cells alone.
42–44
To reconcile these contradictory
results, Salio et al
43
and Somersan et al
37
have suggested
Figure 8 C57BL/6 (a-d) or BALB/c (e-h) mice (n ¼ 5) were immunized weekly with PBS (a, e), DCs alone (b, f), irradiated CD40L-tumor alone
(c, g), or DCs pulsed with irradiated CD40L-tumor cells (d, h). At 2 weeks after the third immunization, mice were challenged with wild-type B16-
F1 (a-d) or CT-26 (e-h) tumor cells, respectively. The growth of tumor was monitored every 2–3 days. Mice were killed when the tumor volume
exceeded 2 cm
3
. Several mice (#) died when the tumors were still developing. The numbers in the parentheses indicate the ratio of tumor-free
animals. Results from two separate experiments are shown.
Antigen loading and concurrent activation of DC
K-J Liu et al
143
Cancer Gene Therapy
that DCs are triggered to maturation not simply by the
death of surrounding cells, but also by other coexisting
stimulatory signals. These factors include inflammatory
cytokines,
42
heat-shock proteins released by dead cells,
37
and microbial materials (e.g., mycoplasma) contained in
the cell lysate.
43
Feng et al
45
also demonstrated that only
stressed apoptotic cells (but not nonstressed apoptotic
cells) could efficiently stimulate DC maturation, possibly
through upregulation of surface heat-shock proteins. Our
results support the notion that exposure of immature DCs
to necrotic or apoptotic cells may not be sufficient to
induce DC activation. Additional stimulations are re-
quired to fully mature DCs. Since our tumor cell culture
was free from mycoplasma contamination, it is likely that
little (if any) heat-shock protein was released from the
tumor cells under the conditions of our experiment, and
hence DCs were not induced to maturation after
coculturing with necrotic or unmodified apoptotic tumor
cells. In our case, the concurrent signaling by functional
membrane-CD40L is required and sufficient for optimal
stimulation of DCs to secrete IL-12 and other proin-
flammatory cytokines, and to promote the growth of
CD8
þ
T cells. This is further supported by a recent study
indicating that both crosstolerance and crosspriming
require the presence of mature DCs and the determining
factor is whether DCs receive an additional activation
signal from CD4
þ
T cells, most likely through CD40–
CD40L interaction.
46
A combination of DC-based cancer vaccines with
costimulatory molecules, cytokines, and chemokines
could provide potential activation signals and allow for
a greater control of DC functions particularly in the
microenvironment of vaccine injection site. Soluble forms
of these immune-regulatory mediators are convenient to
handle, but may have limitations due to a short half-life if
delivered locally, or undesired toxicities if administered
systemically. One strategy of enhancing the efficacy of
DC-based vaccine is to use gene-modified tumor cells as a
provider of both tumor Ag and activation stimulus for
DCs in vitro. In our study, coculturing immature DCs
with frozen/thawed or irradiated vector-tumor cells
resulted in uptake of tumor materials by DCs, but failed
to induce DC maturation and activation. In contrast,
irradiated, but not frozen/thawed, CD40L-tumor cells can
efficiently induce the maturation and activation of DCs in
the absence of additional cytokines. DCs cultured with
irradiated CD40L-tumor cells secreted not only IL-12 but
also a substantial amount of IL-6, IL-8, IFN-g, and TNF-
a. Thus, vaccination with such tumor material-loaded,
activated DCs might create an inflammatory microenvir-
onment at the injection site, and effectively promote the
generation of Th1 and CTL responses. Indeed, more IFN-
g-producing, CD8
þ
T cells were obtained after stimulat-
ing allogeneic or autologous T cells with DCs precultured
with irradiated CD40L-tumors. Although soluble CD40L
may provide a similar stimulus, results reported pre-
viously
47
and from our own experiments (data not shown)
indicate that its stimulatory effect might be lower than
that of membrane-bound CD40L. We have also observed
that DCs cultured with either irradiated vector- or
CD40L-tumor cells can both process internalized tumor
materials and generate HER2/369-377-specific T cells in
vitro. However, T cells survived for a longer period in vitro
if they are initially primed by DCs cultured with
irradiated CD40L-tumor cells. Since DCs stimulated with
irradiated vector-tumor cells secrete very little IL-12, it is
possible that a large amount of IL-12 may be more
important in the maintenance (in contrast to the priming)
of peptide-specific CD8
þ
T cells. Nevertheless, other
CD40L-mediated signals may also increase the ability of
DCs to support the growth of CD8
þ
T cells. DCs
cocultured with irradiated vector-tumor cells did not
mature efficiently, but could still induce HER2/369-377-
specific T cells (Fig 7a). One possible explanation is that
these immature DCs also secreted a substantial amount of
IL-6 (Fig 4). Several investigators have reported that IL-6
plays an important role in the induction of CTL in
vitro.
48–51
A combination of IL-6 and IL-12 is likely to
further enhance the survival of CTLs,
51
as occurs in
cultures containing DCs pulsed with irradiated CD40L-
tumor cells. Our results are in agreement with these
previous findings.
We have observed that a low but significant amount of
IL-10 was present in the culture containing DCs and
irradiated CD40L-tumor cells. IL-10 has been shown to
suppress immature DC development.
52–54
However, the
presence of IL-10 in our cultures did not seem to interfere
with the induction of CD83 expression on DCs, and the
ability of DCs to mediate a strong allogeneic MLR. It has
been shown that mature DCs are more resistant to the
inhibitory effect of IL-10.
52,55
Therefore, it is likely that
DCs cocultured with irradiated CD40L-tumor cells have
undergone maturation and activation before the IL-10
can act. Signaling through CD40L and the presence of
TNF-a may counteract the inhibitory effect of IL-10 as
well.
56
Alternatively, IL-10 in this setting may be
stimulatory, since IL-10 was shown to promote the
maintenance of tumor-specific CD8
þ
T-cell effector
function.
57
This is in agreement with our finding that
CD8
þ
T cells initially stimulated with DCs cultured with
irradiated CD40L-tumor cells survived longer during the
in vitro stimulation procedure. The failure of frozen/
thawed CD40L-tumor cells to induce DC maturation may
be due to the denaturation of surface CD40L during the
freezing/thawing treatment. Although most tumor cells
remain intact after only one cycle of freezing/thawing, it is
also possible that frozen/thawed CD40L-tumor cells are
internalized or break down early in the culture before
sustained contact with DCs needed for efficient induction
of maturation is achieved.
This study was initiated mainly to evaluate the
potential use of DCs pulsed with CD40L-tumor cells as
a cancer vaccine in future clinical studies. Therefore,
human tumor cells and monocyte-derived DCs were used
in our in vitro system. We have demonstrated that pulsing
DCs ex vivo with irradiated CD40L-tumor cells can result
in simultaneous internalization of tumor materials and
activation of DCs without adding exogenous cytokines.
Pulsing with irradiated CD40L-tumor cells proved to be a
better strategy for increasing the capacity of DCs to
Antigen loading and concurrent activation of DC
K-J Liu et al
144
Cancer Gene Therapy
stimulate CD8
þ
T cells than pulsing with unmodified
tumor cells. Also, the in vitro irradiated CD40L-tumor
cell-stimulated production of various pro-Th1 cytokines
and proinflammatory chemokines should increase the
efficacy of vaccination in vivo. It is likely that the secretion
of IL-6, IL-8, IFN-g, and TNF-a at the vaccination site by
these activated DCs might attract and/or activate cells of
the innate immune system such as NK cells and
eosinophils and lead to an amplification of the immune
responses. Thus, pulsing DCs with irradiated CD40L-
tumor cells in vitro may allow Ag loading and activation
of DCs to be better controlled, and vaccination with DCs
pulsed with irradiated CD40L-tumor cells may have a
greater therapeutic effect than vaccination with irradiated
CD40L-tumor cells alone. Our preliminary experiments
demonstrated that animals immunized with DCs pulsed
with irradiated CD40L-tumor cells were indeed better
protected against a weakly immunogenic tumor cell line.
It is possible that tumor cell-based vaccines and DC-based
vaccines may generate different immune responses or
involve different effector cells. We are currently conduct-
ing animal experiments to clarify the underlying mechan-
isms of these two different immunization strategies. Since
most human cancer cells are likely to be of low
immunogenicity, our results suggest that DCs pulsed with
irradiated CD40L-tumor cells could be useful clinically
and may provide the basis of cancer vaccines that could
be tested in future clinical studies. Since sufficient tumor
cells may not always be available and freshly isolated
tumor cells are difficult to transfect, such an approach
(using autologous gene-modified tumor cells) may be
inappropriate for the treatment of some cancer patients.
However, in several recent clinical studies of cancer
vaccine, unmodified or gene-modified allogeneic tumor
cells were chosen as the source of tumor Ag.
18,58
The basis
for this choice was the assumption and observation that
tumor cells from different patients express some of the
same TAA (e.g., CEA, MUC-1, and HER2/neu), and that
DCs are superior in crosspresentation.
59,60
Therefore, it is
possible that well-characterized, allogeneic tumor cell
lines transfected with CD40 ligand could be used as the
source of tumor Ag to pulse and stimulate DCs in a future
clinical application of our proposed vaccination strategy.
Acknowledgments
We would like to thank a former collaborator, Dr Kun-
Tai Lu, at the National Taiwan University Hospital, for
providing malignant pleural effusions and a former senior
technician, Mrs Den-Mei Yang, for her technical assis-
tance in establishing the human lung cancer cell line.
References
1. Palucka K, Banchereau J. Dendritic cells: a link between
innate and adaptive immunity. J Clin Immunol. 1999;19:12–
25.
2. DeMatos P, Abdel-Wahab Z, Vervaert C, et al. Vaccination
with dendritic cells inhibits the growth of hepatic metastases
in B6 mice. Cell Immunol. 1998;185:65–74.
3. DeMatos P, Abdel-Wahab Z, Vervaert C, et al. Pulsing
of dendritic cells with cell lysates from either B16 mela-
noma or MCA-106 fibrosarcoma yields equally effective
vaccines against B16 tumors in mice. J Surg Oncol.
1998;68:79–91.
4. Fields RC, Shimizu K, Mule JJ. Murine dendritic cells
pulsed with whole tumor lysates mediate potent antitumor
immune responses in vitro and in vivo. Proc Natl Acad Sci
USA. 1998;95:9482–9487.
5. Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of patients
with B-cell lymphoma using autologous antigen-pulsed
dendritic cells. Nat Med. 1996;2:52–58.
6. Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of
melanoma patients with peptide- or tumor lysate-pulsed
dendritic cells (see comments). Nat Med. 1998;4:328–332.
7. Tjoa BA, Simmons SJ, Bowes VA, et al. Evaluation of phase
I/II clinical trials in prostate cancer with dendritic cells and
PSMA peptides. Prostate. 1998;36:39–44.
8. Schott M, Feldkamp J, Schattenberg D, et al. Induction of
cellular immunity in a parathyroid carcinoma treated with
tumor lysate-pulsed dendritic cells. Euro J Endocrinol.
2000;142:300–306.
9. Holtl L, Rieser C, Papesh C, et al. Cellular and humoral
immune responses in patients with metastatic renal cell
carcinoma after vaccination with antigen pulsed dendritic
cells. J Urol. 1999;161:777–782.
10. Nair SK, Boczkowski D, Morse M, et al. Induction
of primary carcinoembryonic antigen (CEA)-specific cyto-
toxic T lymphocytes in vitro using human dendritic
cells transfected with RNA. Nat Biotechnol. 1998;16:364–
369.
11. Inaba K, Turley S, Yamaide F, et al. Efficient presentation
of phagocytosed cellular fragments on the major histocom-
patibility complex class II products of dendritic cells. J Exp
Med. 1998;188:2163–2173.
12. Albert ML, Sauter B, Bhardwaj N. Dendritic cells acquire
antigen from apoptotic cells and induce class I-restricted
CTLs. Nature. 1998;392:86–89.
13. Rovere P, Vallinoto C, Bondanza A, et al. Bystander
apoptosis triggers dendritic cell maturation and antigen-
presenting function. J Immunol. 1998;161:4467–4471.
14. Russo V, Tanzarella S, Dalerba P, et al. Dendritic cells
acquire the MAGE-3 human tumor antigen from apoptotic
cells and induce a class I-restricted T cell response. Proc Natl
Acad Sci USA. 2000;97:2185–2190.
15. Jenne L, Arrighi JF, Jonuleit H, et al. Dendritic cells
containing apoptotic melanoma cells prime human CD8+ T
cells for efficient tumor cell lysis. Cancer Res. 2000;60:4446–
4452.
16. Gong J, Chen D, Kashiwaba M, et al. Induction of
antitumor activity by immunization with fusions of dendritic
and carcinoma cells. Nat Med. 1997;3:558–561.
17. Kugler A, Stuhler G, Walden P, et al. Regression of human
metastatic renal cell carcinoma after vaccination with tumor
cell–dendritic cell hybrids (see comments). Nat Med.
2000;6:332–336.
18. Jaffee EM, Abrams R, Cameron J, et al. A phase I clinical
trial of lethally irradiated allogeneic pancreatic tumor
cells transfected with the GM-CSF gene for the treatment
of pancreatic adenocarcinoma. Hum Gene Ther.
1998;9:1951–1971.
19. Greten TF, Jaffee EM. Cancer vaccines. J Clin Oncol.
1999;17:1047–1060.
Antigen loading and concurrent activation of DC
K-J Liu et al
145
Cancer Gene Therapy
20. Parmiani G, Rodolfo M, Melani C. Immunological gene
therapy with ex vivo gene-modified tumor cells: a critique
and a reappraisal. Hum Gene Ther. 2000;11:1269–1275.
21. Roth JA, Cristiano RJ. Gene therapy for cancer: what have
we done and where are we going? J Natl Cancer Inst.
1997;89:21–39.
22. Chiodoni C, Paglia P, Stoppacciaro A, et al. Dendritic
cells infiltrating tumors cotransduced with granulocyte/
macrophage colony-stimulating factor (GM-CSF) and
CD40 ligand genes take up and present endogenous
tumor-associated antigens, and prime naive mice for a
cytotoxic T lymphocyte response. J Exp Med. 1999;190:125–
133.
23. Fujita N, Kagamu H, Yoshizawa H, et al. CD40 ligand
promotes priming of fully potent antitumor CD4(+) T cells
in draining lymph nodes in the presence of apoptotic tumor
cells. J Immunol. 2001;167:5678–5688.
24. Grangeon C, Cormary C, Douin-Echinard V, et al. In vivo
induction of antitumor immunity and protection against
tumor growth by injection of CD154-expressing tumor cells.
Cancer Gene Ther. 2002;9:282–288.
25. de Saint-Vis B, Fugier-Vivier I, Massacrier C, et al. The
cytokine profile expressed by human dendritic cells is
dependent on cell subtype and mode of activation.
J Immunol. 1998;160:1666–1676.
26. Mackey MF, Gunn JR, Maliszewsky C, et al. Dendritic cells
require maturation via CD40 to generate protective
antitumor immunity. J Immunol. 1998;161:2094–2098.
27. Mackey MF, Barth Jr RJ, Noelle RJ. The role of CD40/
CD154 interactions in the priming, differentiation, and
effector function of helper and cytotoxic T cells. J Leukoc
Biol. 1998;63:418–428.
28. Moodycliffe AM, Shreedhar V, Ullrich SE, et al. CD40–
CD40 ligand interactions in vivo regulate migration of
antigen-bearing dendritic cells from the skin to draining
lymph nodes. J Exp Med. 2000;191:2011–2020.
29. Ridge JP, Di Rosa F, Matzinger P. A conditioned dendritic
cell can be a temporal bridge between a CD4+ T-helper and
a T-killer cell (see comments). Nature. 1998;393:474–478.
30. Fisk B, Blevins TL, Wharton JT, et al. Identification of an
immunodominant peptide of HER-2/neu protooncogene
recognized by ovarian tumor-specific cytotoxic T lympho-
cyte lines. J Exp Med. 1995;181:2109–2117.
31. Bartholomew JS, Stacey SN, Coles B, et al. Identification of
a naturally processed HLA A0201-restricted viral peptide
from cells expressing human papillomavirus type 16 E6
oncoprotein. Eur J Immunol. 1994;24:3175–3179.
32. Giatromanolaki A, Koukourakis MI, O’Byrne K, et al.
Non-small cell lung cancer: c-erbB-2 overexpression corre-
lates with low angiogenesis and poor prognosis. Anticancer
Res. 1996;16:3819–3825.
33. Lodge PA, Jones LA, Bader RA, et al. Dendritic cell-based
immunotherapy of prostate cancer: immune monitoring of a
phase II clinical trial. Cancer Res. 2000;60:829–833.
34. Fong L, Brockstedt D, Benike C, et al. Dendritic cells
injected via different routes induce immunity in cancer
patients. J Immunol. 2001;166:4254–4259.
35. Steinman RM, Turley S, Mellman I, et al. The induction of
tolerance by dendritic cells that have captured apoptotic
cells. J Exp Med. 2000;191:411–416.
36. Sauter B, Albert ML, Francisco L, et al. Consequences of
cell death: exposure to necrotic tumor cells, but not primary
tissue cells or apoptotic cells, induces the maturation of
immunostimulatory dendritic cells (see comments). J Exp
Med. 2000;191:423–434.
37. Somersan S, Larsson M, Fonteneau JF, et al. Primary
tumor tissue lysates are enriched in heat shock proteins and
induce the maturation of human dendritic cells. J Immunol.
2001;167:4844–4852.
38. Cella M, Salio M, Sakakibara Y, et al. Maturation,
activation, and protection of dendritic cells induced by
double-stranded RNA. J Exp Med. 1999;189:821–829.
39. Curtsinger JM, Schmidt CS, Mondino A, et al. Infla-
mmatory cytokines provide a third signal for activation
of naive CD4+ and CD8+ T cells. J Immunol.
1999;162:3256–3262.
40. Rovere P, Sabbadini MG, Vallinoto C, et al. Delayed
clearance of apoptotic lymphoma cells allows cross-pre-
sentation of intracellular antigens by mature dendritic cells.
J Leukoc Biol. 1999;66:345–349.
41. Gallucci S, Lolkema M, Matzinger P. Natural adjuvants:
endogenous activators of dendritic cells. Nat Med.
1999;5:1249–1255.
42. Vegh Z, Mazumder A. Generation of tumor cell lysate-
loaded dendritic cells preprogrammed for IL-12 production
and augmented T cell response. Cancer Immunol Immun-
other. 2003;52:67–79.
43. Salio M, Cerundolo V, Lanzavecchia A. Dendritic cell
maturation is induced by mycoplasma infection but not by
necrotic cells. Eur J Immunol. 2000;30:705–708.
44. Hoffmann TK, Meidenbauer N, Muller-Berghaus J, et al.
Proinflammatory cytokines and CD40 ligand enhance cross-
presentation and cross-priming capability of human den-
dritic cells internalizing apoptotic cancer cells. J Immun-
other. 2001;24:162–171.
45. Feng H, Zeng Y, Graner MW, et al. Stressed apoptotic
tumor cells stimulate dendritic cells and induce specific
cytotoxic T cells. Blood. 2002;100:4108–4115.
46. Albert ML, Jegathesan M, Darnell RB. Dendritic cell
maturation is required for the cross-tolerization of CD8+ T
cells. Nat Immunol. 2001;2:1010–1017.
47. Felzmann T, Buchberger M, Lehner M, et al. Functional
maturation of dendritic cells by exposure to CD40L
transgenic tumor cells, fibroblasts or keratinocytes. Cancer
Lett. 2001;168:145–154.
48. Quentmeier H, Klaucke J, Muhlradt PF, et al. Role of IL-6,
IL-2, and IL-4 in the in vitro induction of cytotoxic T cells. J
Immunol. 1992;149:3316–3320.
49. Bass HZ, Yamashita N, Clement LT. Heterogeneous
mechanisms of human cytotoxic T lymphocyte generation.
II. Differential effects of IL-6 on the helper cell-independent
generation of CTL from CD8+ precursor subpopulations. J
Immunol. 1993;151:2895–2903.
50. Ming JE, Steinman RM, Granelli-Piperno A, et al. IL-6
enhances the generation of cytolytic T lymphocytes in the
allogeneic mixed leucocyte reaction. Clin Exp Immunol.
1992;89:148–153.
51. Gajewski TF, Renauld JC, Van Pel A, et al. Costimulation
with B7-1, IL-6, and IL-12 is sufficient for primary
generation of murine antitumor cytolytic T lymphocytes in
vitro. J Immunol. 1995;154:5637–5648.
52. Allavena P, Piemonti L, Longoni D, et al. IL-10 prevents the
differentiation of monocytes to dendritic cells but promotes
their maturation to macrophages. Eur J Immunol.
1998;28:359–369.
53. Buelens C, Verhasselt V, De Groote D, et al. Interleukin-10
prevents the generation of dendritic cells from human
peripheral blood mononuclear cells cultured with interleu-
kin-4 and granulocyte/macrophage-colony-stimulating fac-
tor. Eur J Immunol. 1997;27:756–762.
Antigen loading and concurrent activation of DC
K-J Liu et al
146
Cancer Gene Therapy
54. Morel AS, Quaratino S, Douek DC, et al. Split activity of
interleukin-10 on antigen capture and antigen presentation
by human dendritic cells: definition of a maturative step.
Eur J Immunol. 1997;27:26–34.
55. Kalinski P, Schuitemaker JH, Hilkens CM, et al. Prosta-
glandin E2 induces the final maturation of IL-12-deficient
CD1a+CD83+ dendritic cells: the levels of IL-12 are
determined during the final dendritic cell maturation and are
resistant to further modulation. J Immunol. 1998;161:2804–
2809.
56. Brossart P, Zobywalski A, Grunebach F, et al. Tumor
necrosis factor alpha and CD40 ligand antagonize
the inhibitory effects of interleukin 10 on T-cell stimu-
latory capacity of dendritic cells. Cancer Res. 2000;60:4485–
4492.
57. Fujii S, Shimizu K, Shimizu T, et al. Interleukin-10
promotes the maintenance of antitumor CD8(+) T-cell
effector function in situ. Blood. 2001;98:2143–2151.
58. Arienti F, Belli F, Napolitano F, et al. Vaccination of
melanoma patients with interleukin 4 gene-transduced
allogeneic melanoma cells. Hum Gene Ther. 1999;10:2907–
2916.
59. Nouri-Shirazi M, Banchereau J, Bell D, et al. Dendritic cells
capture killed tumor cells and present their antigens to elicit
tumor-specific immune responses. J Immunol.
2000;165:3797–3803.
60. Berard F, Blanco P, Davoust J, et al. Cross-priming of naive
CD8 T cells against melanoma antigens using dendritic cells
loaded with killed allogeneic melanoma cells. J Exp Med.
2000;192:1535–1544.
Antigen loading and concurrent activation of DC
K-J Liu et al
147
Cancer Gene Therapy
... 15 We are interested in evaluating the combination of gene-modified tumor cells with DC-based cancer vaccine as an improvement of current therapy. 16 We proposed to use macrophage inflammation protein-3a (MIP-3a)-transfected tumor cells for in vivo preconditioning of vaccination sites of DC-based vaccine. MIP-3a, also known as LARC (liver and activation-regulated chemokine) and as exodus-1, is a CC chemokine (CCL20) capable of inducing migration of immune cells including immature DCs and epidermal Langerhans cells that express CCR6, the receptor for MIP-3a. ...
... Mouse DCs were generated by culturing bone marrow (BM) cells with murine granulocyte macrophage colony stimulating factor (GM-CSF) and interleukin (IL)-4 (Peprotech, London, England) for 7 days as described previously. 16,23 To increase the purity of BM-derived DCs (BMDCs), cells after GM-CSF/IL-4 culture were enriched by density gradient centrifugation with 80% Ficoll-Paque (Amersham Pharmacia Biotech, Uppsala, Sweden) and cells in the interface were collected as the source of BMDCs. The resulting cells were subjected to flow cytometry analysis and at least 85% of cells were gated based on forward scatter/side scatter setting to detect the expression of various cell markers. ...
... The generation of CD40L-transfected B16F1 tumor cells and their ability to stimulate maturation of DCs have been described previously. 16 For lung metastasis model, mice were immunized with PBS alone, or MIP-tumor vaccine followed by DC vaccine either at different sides of the flank or at the same site on one flank. They were boosted once 2 weeks later with the same protocol. ...
Article
Full-text available
Macrophage inflammation protein-3alpha (MIP-3alpha) is a chemokine expressed in inflamed tissue and capable of inducing migration of immature dendritic cells (DCs) or Langerhans cells. We postulated that conditioning vaccination sites with MIP-3alpha might enhance the efficacy of subsequently administered DC-based cancer vaccines. Our results demonstrate that subcutaneously injection of irradiated tumor cells expressing MIP-3alpha induces substantial cell infiltration to the injection site. Vaccination of irradiated tumor cells expressing MIP-3alpha followed by DCs pulsed with irradiated tumor cells can effectively suppress tumor growth in animals, which is significantly better than vaccination with irradiated MIP-3alpha-producing tumor cells or DCs pulsed with tumor cells alone. The protective effect was most evident when the MIP-3alpha-producing tumor cells and DC-based vaccines were injected at the same site. These results support the notion that this combination vaccination strategy might generate a more effective immune response to suppress the growth of tumor cells in animals.
... These efforts include improvement of DC stimulation [25,26], pre-conditioning of the vaccination site [27], and measures to maintain a long-lasting T cell response with cytokines or other biological agents [28]. We previously reported in animal models a strategy for pulsing DCs with CD40 ligand-transfected tumor cells and pre-conditioning with MIP-3α-transfected tumor cells [29,30]. Both approaches resulted in a better immune response against the tumor and effectively suppressed tumor growth and metastasis. ...
Article
Full-text available
Background To better evaluate and improve the efficacy of dendritic cell (DC)-based cancer immunotherapy, we conducted a clinical study of patients with advanced colorectal cancer using carcinoembryonic antigen (CEA)-pulsed DCs mixed with tetanus toxoid and subsequent interleukin-2 treatment. The tetanus toxoid in the vaccine preparation serves as an adjuvant and provides a non-tumor specific immune response to enhance vaccine efficacy. The aims of this study were to (1) evaluate the toxicity of this treatment, (2) observe the clinical responses of vaccinated patients, and (3) investigate the immune responses of patients against CEA before and after treatment. Methods Twelve patients were recruited and treated in this phase I clinical study. These patients all had metastatic colorectal cancer and failed standard chemotherapy. We first subcutaneously immunized patients with metastatic colorectal cancer with 1 × 106 CEA-pulsed DCs mixed with tetanus toxoid as an adjuvant. Patients received 3 successive injections with 1 × 106 CEA-pulsed DCs alone. Low-dose interleukin-2 was administered subcutaneously following the final DC vaccination to boost the growth of T cells. Patients were evaluated for adverse event and clinical status. Blood samples collected before, during, and after treatment were analyzed for T cell proliferation responses against CEA. ResultsNo severe treatment-related side effects or toxicity was observed in patients who received the regular 4 DC vaccine injections. Two patients had stable disease and 10 patients showed disease progression. A statistically significant increase in proliferation against CEA by T cells collected after vaccination was observed in 2 of 9 patients. Conclusions The results of this study indicate that it is feasible and safe to treat colorectal cancer patients using this protocol. An increase in the anti-CEA immune response and a clinical benefit was observed in a small fraction of patients. This treatment protocol should be further evaluated in additional colorectal cancer patients with modifications to enhance T cell responses. Trial registrationClinicalTrials.gov (identifier NCT00154713), September 8, 2005
... This goes along with reports, where irradiated and CD40L transfected tumor cells have been investigated to activate DCs resulting in tumor specific activation of CD8 þ cells. 45 Although we performed coinfections with oncolytic Ad and CD40L-encoding adenoviral vector, our results represent a promising example for the opportunity to modulate immune responses induced by adenoviral oncolysis via transgene expression. Future studies should investigate further genetic strategies for improving human DC functions by viral oncolysate. ...
Article
Oncolytic adenoviruses are emerging agents for treatment of cancer by tumor-restricted virus infection and cell lysis. Clinical trials have shown that oncolytic adenoviruses are well tolerated in patients but also that their antitumor activity needs improvement. A promising strategy toward this end is to trigger systemic and prolonged antitumor immunity by adenoviral oncolysis. Antitumor immune activation depends in large part on antigen presentation and T cell activation by dendritic cells (DCs). Thus, it is likely that the interaction of lysed tumor cells with DCs is a key determinant of such "oncolytic vaccination." Our study reveals that human DCs effectively phagocytose melanoma cells at late stages of oncolytic adenovirus infection, when the cells die showing preferentially features of necrotic cell death. Maturation, migration toward CCL19 and T cell stimulatory capacity of DCs, crucial steps for immune induction, were, however, not induced by phagocytosis of oncolysate, but could be triggered by a cytokine maturation cocktail. Therefore, oncolytic adenoviruses and adenoviral oncolysate did not block DC maturation, which is in contrast to reports for other oncolytic viruses. These results represent a rationale for inserting immunostimulatory genes into oncolytic adenovirus genomes to assure critical DC maturation. Indeed, we report here that adenoviral transduction of melanoma cells with CD40L during oncolysis triggers the maturation of human DCs with T cell stimulatory capacity similar to DCs matured by cytokines. We conclude that triggering and shaping DC-induced antitumor immunity by oncolytic adenoviruses "armed" with immunostimulatory genes holds promise for improving the therapeutic outcome of viral oncolysis in patients.
... These results indicate that enhanced secretion of IL-12, IFN-γ, and TNF-α is due to CD40L incorporation into the SHIV-VLP. Our data are consistent with a previous report that CD40L could deliver both tumor-derived antigens (Ag) and maturation stimuli simultaneously to DCs while also leading to the production of proinflammatory chemokines and pro-Th1 cytokines like IL-6, IL-8, IL-12, IFN-γ, and TNF-α [53]. Another report showed that HIV-1-infected DCs had up-regulated cell surface markers but failed to produce IL-12 in response to CD40L stimulation, suggesting that HIV-1 infection disables DC function [54]. ...
Article
Engagement of CD40 with CD40L induces dendritic cell (DC) maturation and activation, thereby promoting immune responses. The objective of this study was to investigate whether immunization with chimeric CD40L/SHIV virus-like particles (CD40L/SHIV-VLP) could enhance immune responses to SIV Gag and HIV Env proteins by directly activating DCs. We found that CD83, CD40, and CD86 were significantly up-regulated and significantly increased cytokines production were observed after hCD40L/SHIV-VLP treatment in human CD14(+) monocyte-derived DCs as compared to SHIV-VLP treatment. Mice immunized with mCD40L/SHIV-VLP showed more than a two-fold increase in HIV Env-specific IgG antibody production, an increase in SIV Gag and HIV Env-specific IFN-gamma and IL-4 producing cells, and an increase in HIV Env-specific cytotoxic activity compared to that in SHIV-VLP immunized mice. Furthermore, multifunctional CD4(+) Th1 cells, which simultaneously produce IFN-gamma, IL-2 and TNF-alpha triple cytokines, and CD8(+) T-cells, which produce IFN-gamma were elevated in the mCD40L/SHIV-VLP immunized group. These data demonstrate that chimeric CD40L/SHIV-VLP potently induce DC activation and enhance the magnitude of both humoral and cellular immune responses to the SIV Gag and HIV Env proteins in the mouse model. Therefore, incorporation of CD40L into VLP may represent a novel strategy to develop effective HIV vaccines.
... CD40L has been shown to improve immunogenicity in several therapeutic cancer (40,58) and prophylactic vaccine (20,46) studies. Skountzou et al. demonstrated that incorporating CD40L into an SIV virus-like particle enhances humoral and cellular immune responses (61). ...
Article
Full-text available
A vaccine for the prevention of human immunodeficiency virus (HIV) infection is desperately needed to control the AIDS pandemic. To address this problem, we developed vesicular stomatitis virus glycoprotein-pseudotyped replication-defective simian immunodeficiency viruses (dSIVs) as an AIDS vaccine strategy. The dSIVs retain characteristics of a live attenuated virus without the drawbacks of potential virulence caused by replicating virus. To improve vaccine immunogenicity, we incorporated CD40 ligand (CD40L) into the dSIV envelope. CD40L is one of the most potent stimuli for dendritic cell (DC) maturation and activation. Binding of CD40L to its receptor upregulates expression of major histocompatibility complex class I, class II, and costimulatory molecules on DCs and increases production of proinflammatory cytokines and chemokines, especially interleukin 12 (IL-12). This cytokine polarizes CD4(+) T cells to Th1-type immune responses. DC activation and mixed lymphocyte reaction (MLR) studies were performed to evaluate the immunogenicity of CD40L-dSIV in vitro. Expression levels of CD80, CD86, HLA-DR, and CD54 on DCs transduced with the dSIV incorporating CD40L (CD40L-dSIV) were significantly higher than on those transduced with dSIV. Moreover, CD40L-dSIV-transduced DCs expressed up to 10-fold more IL-12 than dSIV-transduced DCs. CD40L-dSIV-transduced DCs enhanced proliferation and gamma interferon secretion by naive T cells in an MLR. In addition, CD40L-dSIV-immunized mice exhibited stronger humoral and cell-mediated immune responses than dSIV-vaccinated animals. The results show that incorporating CD40L into the dSIV envelope significantly enhances immunogenicity. As a result, CD40L-dSIVs can be strong candidates for development of a safe and highly immunogenic AIDS vaccine.
Article
Full-text available
The tumor vaccine MGN1601 was designed and developed for treatment of metastatic renal cell carcinoma (mRCC). MGN1601 consists of a combination of fourfold gene-modified cells with the toll-like receptor 9 agonist dSLIM, a powerful connector of innate and adaptive immunity. Vaccine cells originate from a renal cell carcinoma cell line (grown from renal cell carcinoma tissue), express a variety of known tumor-associated antigens (TAA), and are gene modified to transiently express two co-stimulatory molecules, CD80 and CD154, and two cytokines, GM-CSF and IL-7, aimed to support immune response. Proof of concept of the designed vaccine was shown in mice: The murine homologue of the vaccine efficiently (100%) prevented tumor growth when used as prophylactic vaccine in a syngeneic setting. Use of the vaccine in a therapeutic setting showed complete response in 92% of mice as well as synergistic action and necessity of the components. In addition, specific cellular and humoral immune responses in mice were found when used in an allogeneic setting. Immune response to the vaccine was also shown in mRCC patients treated with MGN1601: Peptide array analysis revealed humoral CD4-based immune response to TAA expressed on vaccine cells, including survivin, cyclin D1, and stromelysin.
Article
Despite recent attempts to take advantage of dendritic cell (DC)-based vaccines for cancer immunotherapy, the results of clinical studies have been disappointing. This is mainly as a result of the diverse immune escape mechanisms used by the tumor together with the insufficient ability of DCs to mount an effective immune response against these mechanisms. In this regard, several approaches have been devised to improve the efficacy of DC-based vaccines. However, the application of each individual approach per se might not be sufficient to overwhelm the diverse immune escape mechanisms. In this review, we focus on current strategies for the ex vivo potentiation of DC-based vaccines, with an emphasis on combinational therapy methods as a promising alternative for tumor immunotherapy.
Article
Dendritic cells (DCs) pulsed by tumor antigens have been widely used as tumor vaccines to specifically trigger the cytotoxicity of CD8+ T cells. But the tumor microenviroment with enriched immunosuppressants hampered DC maturation and co-stimulation. CD40/CD40L signaling, one of the most important co-stimulatory molecules is capable of effectively skewing the immune response by promoting DCs maturation and co-stimulation. To establish a novel specific immunotherapeutic approach for the use of DC vaccine in the treatment of B lymphoma, hu-SCID mice bearing B lymphoma were vaccinated by different combination of tumor antigen pulsed DC or imDC vaccines and immune-enhancing agencies such as agonist CD40mAb and T cells. The results of immature DCs combined with agonistic CD40mAb were encouraging with achievement of tumor regression and induction of antigen-specific immune responses. These findings demonstrated the potential utility of imDC-based tumor vaccination combining with agonistic CD40mAb in the treatment of malignant lymphoma.
Article
Die Immuntherapie stellt eine mögliche Therapiealternative bei malignen Erkrankungen dar. In dieser Arbeit wurden im murinen Modell dendritische Zellen in vitro generiert, mit bestrahlten Tumorzellen koinkubiert und mit CpG-Oligonukleotiden aktiviert. Diese Tumorvakzine wurde subkutan gegen subkutan induzierte Tumoren eingesetzt. Sowohl prophylaktisch wie auch therapeutisch konnte eine potente antitumorale Immunantwort induziert werden, die systemisch wirksam, lang anhaltend und tumorspezifisch war. Durch einen Vergleich zwischen zwei Mausstämmen konnten Faktoren, die möglicherweise die antitumorale Immunantwort beeinflussen, identifiziert werden.
Article
5C11, a murine monoclonal antibody with a high specificity for human CD40 molecule, is a promising candidate for cancer targeting therapy. We have therefore attempted to construct a humanized antibody of 5C11 to minimize its immunogenicity for potential clinical use. A chimeric version of 5C11 (ch-5C11) was generated by transferring these mouse variable regions onto a human framework. This chimeric antibody retained reactivity to human CD40. In vitro, ch-5C11 could effectively inhibit B lymphoma Daudi cell proliferation, suggesting that it might have the potential to be developed for future clinical use.
Article
It has been more than 100 years since the first reported attempts to activate a patient's immune system to eradicate developing cancers. Although a few of the subsequent vaccine studies demonstrated clinically significant treatment effects, active immunotherapy has not yet become an established cancer treatment modality. Two recent advances have allowed the design of more specific cancer vaccine approaches: improved molecular biology techniques and a greater understanding of the mechanisms involved in the activation of T cells. These advances have resulted in improved systemic antitumor immune responses in animal models. Because most tumor antigens recognized by T cells are still not known, the tumor cell itself is the best source of immunizing antigens. For this reason, most vaccine approaches currently being tested in the clinics use whole cancer cells that have been genetically modified to express genes that are now known to be critical mediators of immune system activation. In the future, the molecular definition of tumor-specific antigens that are recognized by activated T cells will allow the development of targeted antigen-specific vaccines for the treatment of patients with cancer.
Article
IL-10 secretion by tumor cells was demonstrated to be one of the mechanisms by which tumor cells can escape from immunologie recognition. In immature DC, IL-10 inhibits IL-12 production and induces a state of antigen specific anergy in T cells. Here, we analyzed the effects of different activation stimuli including LPS, TNF-α and CD40 ligation on IL-10 mediated inhibition of DC development and T-cell stimulatory capacity. The addition of IL-10 to the cultures containing GM-CSF and IL-4 with or without LPS completely inhibited the generation of DC from peripheral blood monocytes. These cells remained CD 14+ and expressed high levels of IL-10 receptor (IL-10R) suggesting that IL10 mediates its effects by upregulating the IL- 10R. In contrast, the simultaneous incubation of monocytes with GM-CSF/IL-4, and IL-10 and TNF-α or soluble CD40 ligand (sCD40L) resulted in the generation of CD83+ DC, induction of nuclear localized RelB and inhibition of IL-10R up-regulation. DC grown in the presence of IL-10 and TNF-α or sCD40L elicited efficient CTL responses against viral and tumor associated peptide antigens, which, however, were reduced as compared to DC cultures generated without IL-10. IL10 decreased the production of IL-6 and the expression of IL-12 in the presence of TNFa or sCD40L but it had no effect on IL-15, IL-18 and TNF-α secretion. Addition of IL10 to the cultures induced the upregulation of the inflammatory chemokines MIP-la, MCP-2 and MCP-4, suggesting that the DC generated in the presence of IL-10 and TNFa or sCD40L have an intermediate activation status that is modulated by IL-10. Our results show that TNF-α or CD40 ligation can antagonize the IL-10 mediated inhibition on DC function suggesting that the presence of IL-10 not necessarily results in T-cell anergy.
Article
Synthetic peptide analogues of sequences in the HER-2 protooncogene (HER-2) were selected based on the presence of HLA-A2.1 anchor motifs to identify the epitopes on HER-2 recognized by ovarian tumor-reactive CTL. 19 synthetic peptides were evaluated for recognition by four HLA-A2 ovarian-specific cytotoxic T lymphocyte (CTL) lines obtained from leukocytes associated with ovarian tumors. The nonapeptide E75 (HER-2, 369-377:KIFGSLAFL) was efficient in sensitizing T2 cells for lysis by all four CTL lines. This peptide was specifically recognized by cloned CD8+ CTL isolated from one of the ovarian-specific CTL lines. E75-pulsed T2 cells inhibited lysis by the same CTL clone of both an HLA-A2+ HER-2high ovarian tumor and a HER-2high cloned ovarian tumor line transfected with HLA-A2, suggesting that this or a structurally similar epitope may be specifically recognized by these CTL on ovarian tumors. Several other HER-2 peptides were recognized preferentially by one or two CTL lines, suggesting that both common and private HER-2 epitopes may be immunogenic in patients with ovarian tumors. Since HER-2 is a self-antigen, these peptides may be useful for understanding mechanisms of tumor recognition by T cells, immunological tolerance to tumor, and structural characterization of tumor antigens.
Article
What makes a protein immunogenic, particularly for strong T cell–mediated immunity? To a first approximation, this determination seems to be made by dendritic cells (DCs). Immature DCs, as in skin ([1][1])([2][2])([3][3])([4][4]), lung ([5][5]), blood ([6][6])([7][7]), and spleen ([7][7])([8][8
Article
We evaluated the effects of interleukin (IL)-10 on the differentiation of dendritic cells (DC) obtained by culturing plastic-adherent peripheral blood mononuclear cells for 7 days in presence of granulocyte/macrophage-colony-stimulating factor (GM-CSF) + IL-4. The addition of IL-10 at the initiation of culture resulted in the generation of macrophage-like cells with expressing high levels of CD14 and decreased levels of CD1a and CD1c. Furthermore, cells generated in presence of IL-10 secreted lower levels of IL-12, but higher levels of IL-8 compared with DC generated in absence of IL-10, both spontaneously and after CD40 engagement. Finally, cells generated in presence of IL-10 were less efficient than DC in stimulating the production of IL-2, interferon-gamma, and IL-4 by allogeneic T cells. We conclude that IL-10 prevents the generation of DC induced by GM-CSF + IL-4 and favors the development of macrophages with a lower T cell stimulatory potential, but secreting higher levels of IL-8 than DC.
Article
Interleukin-10 (IL-10) is a multifunctional cytokine that can exert suppressive and stimulatory effects on T cells. It was investigated whether IL-10 could serve as an immunostimulant for specific CD8⁺ cytotoxic T cell (CTL) in vivo after vaccination and, if so, under what conditions. In tumor prevention models, administration of IL-10 before, or soon after, peptide-pulsed primary dendritic cell immunization resulted in immune suppression and enhanced tumor progression. Injection of IL-10, however, just after a booster vaccine significantly enhanced antitumor immunity and vaccine efficacy. Analysis of spleen cells derived from these latter animals 3 weeks after IL-10 treatment revealed that the number of CD8⁺CD44hi CD122⁺ T cells had increased and that antigen-specific proliferation in vitro was enhanced. Although cytotoxicity assays did not support differences between the various treatment groups, 2 more sensitive assays measuring antigen-specific interferon-γ production at the single-cell level demonstrated increases in the number of antigen-specific responder T cells in animals in the vaccine/IL-10 treatment group. Thus, IL-10 may maintain the number of antitumor CD8⁺ T cells. In adoptive transfer studies, the ability of IL-10 to maintain CTL function could be enhanced by the depletion of CD4⁺ T cells. This suggests that IL-10 mediates contrasting effects on both CD4⁺ and CD8⁺ T cells that result in either immune dampening or immune potentiation in situ, respectively. Appreciation of this dichotomy in IL-10 immunobiology may allow for the design of more effective cancer vaccines designed to activate and maintain specific CD8⁺ T-cell effector function in situ.
Article
Dendritic cells (DC) represent potent APCs that are capable of generating tumor-specific immunity. We performed a pilot clinical trial using Ag-pulsed DC as a tumor vaccine. Twenty-one patients with metastatic prostate cancer received two monthly injections of DC enriched and activated from their PBMC. DC were cocultured ex vivo with recombinant mouse prostatic acid phosphatase as the target neoantigen. Following enrichment, DC developed an activated phenotype with up-regulation of CD80, CD86, and CD83 expression. During culture, the DC maintained their levels of various adhesion molecules, including CD44, LFA-1, cutaneous lymphocyte-associated Ag, and CD49d, up-regulated CCR7, but lost CD62 ligand and CCR5. In the absence of CD62 ligand, such cells would not be expected to prime T cells efficiently if administered i.v. due to their inability to access lymphoid tissue via high endothelial venules. To assess this possibility, three patient cohorts were immunized with Ag-pulsed DC by i.v., intradermal (i.d.), or intralymphatic (i.l.) injection. All patients developed Ag-specific T cell immune responses following immunization, regardless of route. Induction of IFN-γ production, however, was seen only with i.d. and i.l. routes of administration, and no IL-4 responses were seen regardless of route, consistent with the induction of Th1-type immunity. Five of nine patients who were immunized by the i.v. route developed Ag-specific Abs compared with one of six for i.d. and two of six for i.l. routes. These results suggest that while activated DC can prime T cell immunity regardless of route, the quality of this response and induction of Ag-specific Abs may be affected by the route of administration.
Article
BACKGROUNDA phase I trial involving patients with advanced prostate cancer was conducted to assess the safe administration of dendritic cells (DC) and HLA-A0201-specific prostate-specific membrane antigen (PSMA) peptides (PSM-P1 or -P2). Thirty-three of the phase I participants were subsequently enrolled in a phase II trial, which involved six infusions of DC pulsed with PSM-P1 and -P2 peptides.METHODS Clinical monitoring was conducted up to 770 days from the start of the phase I study. Data collected included: complete blood count, bone and total alkaline phosphatase, prostate markers, physical examination, performance status, bone scan, ProstaScint® scan, and chest X-ray, as well as assays to monitor cellular immune responses.RESULTSNine partial responders were identified in the phase II study based on National Prostate Cancer Project (NPCP) criteria, plus 50% reduction of prostate-specific antigen. Four of the partial responders were also responders in the phase I study, with an average response duration of 225 days. Their combined average total response period was over 370 days. Five other responders were nonresponders in the phase I study. Their average partial response period was 196 days.CONCLUSIONS The responses observed in the phase I and II clinical trials were significant and of long duration. The partial-responder group included patients who continued to respond from phase I, as well as those who started to respond during the phase II trial. Prostate 36:39–44, 1998. © 1998 Wiley-Liss, Inc.
Article
A human melanoma line genetically modified to release interleukin 4 (IL-4) was utilized to immunize advanced melanoma patients in order to elicit or increase a specific anti-melanoma immune response, which may affect distant lesions. Twelve metastatic melanoma patients were injected subcutaneously at least three times with 5 x 10(7) IL-4 gene-transduced and irradiated allogeneic melanoma cells per dose. Both systemic and local toxicities were mild, consisting of transient fever and erythema, swelling, and induration at the vaccination site. Two mixed but not complete or partial clinical responses were recorded. To assess the immune response of vaccinated patients, both serological and cell-mediated activities were evaluated. Antibodies to alloantigens could be detected in 2 of 11 patients tested. Mixed tumor-lymphocyte cultures were performed, utilizing autologous and allogeneic HLA-A2-matched melanoma lines as simulators and targets. A significant increase in IFN-gamma release was detected in 7 of 11 cases when postvaccination lymphocytes were stimulated by the untransduced allomelanoma cells. However, induction of a specific recognition of autologous melanoma cells by PBLs was obtained after vaccination in only one of six cases studied. This response involved the melanoma peptide Melan-A/MART-1(27-35) that was recognized in an HLA-A2-restricted fashion. These results indicate that vaccination with allogeneic melanoma cells releasing IL-4 locally can expand a T cell response against antigen(s) of autologous, untransduced tumor, although in a minority of patients.