ArticlePDF Available

Reversal of CD8+ T Cell Ignorance and Induction of Anti-Tumor Immunity by Peptide-Pulsed APC

Authors:

Abstract and Figures

In the present report, we have studied the potential of naive and activated effector CD8(+) T cells to function as anti-tumor T cells to a solid tumor using OVA-specific T cells from TCR-transgenic OT-I mice. Adoptive transfer of naive OT-I T cells into tumor-bearing syngeneic mice did not inhibit tumor cell growth. The adoptively transferred OT-I T cells did not proliferate in lymphoid tissue of tumor-bearing mice and were not anergized by the tumor. In contrast, adoptive transfer of preactivated OT-I CTL inhibited tumor growth in a dose-dependent manner, indicating that E.G7 was susceptible to immune effector cells. Importantly, naive OT-I T cells proliferated and elicited an anti-tumor response if they were adoptively transferred into normal or CD4-deficient mice that were then vaccinated with GM-CSF-induced bone marrow-derived OVA-pulsed APC. Collectively, these data indicate that even though naive tumor-specific T cells are present at a relatively high fraction they remain ignorant of the tumor and demonstrate that a CD8-mediated anti-tumor response can be induced by Ag-pulsed APC without CD4 T cell help.
Content may be subject to copyright.
Reversal of CD8
T Cell Ignorance and Induction of
Anti-Tumor Immunity by Peptide-Pulsed APC
1
Nava Dalyot-Herman,* Oliver F. Bathe,
and Thomas R. Malek
2
*
In the present report, we have studied the potential of naive and activated effector CD8
T cells to function as anti-tumor T cells
to a solid tumor using OVA-specific T cells from TCR-transgenic OT-I mice. Adoptive transfer of naive OT-I T cells into
tumor-bearing syngeneic mice did not inhibit tumor cell growth. The adoptively transferred OT-I T cells did not proliferate in
lymphoid tissue of tumor-bearing mice and were not anergized by the tumor. In contrast, adoptive transfer of preactivated OT-I
CTL inhibited tumor growth in a dose-dependent manner, indicating that E.G7 was susceptible to immune effector cells. Impor-
tantly, naive OT-I T cells proliferated and elicited an anti-tumor response if they were adoptively transferred into normal or
CD4-deficient mice that were then vaccinated with GM-CSF-induced bone marrow-derived OVA-pulsed APC. Collectively, these
data indicate that even though naive tumor-specific T cells are present at a relatively high fraction they remain ignorant of the
tumor and demonstrate that a CD8-mediated anti-tumor response can be induced by Ag-pulsed APC without CD4 T cell
help. The Journal of Immunology, 2000, 165: 6731–6737.
I
t is now well-established that some tumors express Ags that
result in induction of anti-tumor immune responses (1, 2).
Although in some cases such a response results in eradication
of the tumor, more often an anti-tumor immune response is inef-
fective, and the tumor ultimately grows. There are a number of
specific mechanisms by which a tumor evades an ongoing immune
response. These vary widely and include: development of tu-
mor-Ag loss variants; down-regulation of surface MHC molecules
or other molecules required for Ag presentation by the tumor cells
(3, 4); induction of anergy of tumor-specific T cells due to lack of
costimulatory molecules on the tumor cells (5–7); and suppression
of the immune response by tumor secretion of inhibitory cytokines
(8, 9). Tumor Ags by nature are usually dysregulated self-proteins
or variants of self-Ags. Therefore, anti-tumor immunity may also
fail due to a weak response to the Ags expressed by the replicating
tumor or because potentially tumor-reactive T cells are tolerant to
the tumor Ags.
Although the fate and function of adoptively transferred tumor-
specific effector T cells have been extensively studied (10–12),
comparatively little is known concerning the initial activation of
tumor-specific T cells in vivo, primarily due to their low frequency
in the peripheral lymphocyte pool. With the introduction of TCR-
transgenic mice, this problem has been overcome. Analysis of an
Ag-specific T cell response to nominal Ags is facilitated by adop-
tive transfer of a relatively low, but detectable, number of TCR-
transgenic T cells to normal mice and then challenging such ani-
mals with the appropriate Ag (13). This approach allows direct
phenotypic and functional characterization of the responding Ag-
specific transgenic T cells during the course of the immune re-
sponse and avoids the complications inherent in direct Ag stimu-
lation of the TCR-transgenic mouse, in which all the T cells are Ag
responsive.
More recently, this method has been adapted to study the in-
duction of anti-tumor immunity in vivo. In this setting, a prede-
termined number of naive TCR-transgenic T cells are adoptively
transferred to mice bearing a tumor that was transfected with an
Ag recognized by the transgenic T cells (14–18). The present
study employed this TCR-transgenic strategy to compare the ca-
pacity of naive and activated effector-transgenic CD8
T cells to
generate an anti-tumor immune response to a solid tumor. We used
OT-I TCR-transgenic T cells that are specific for OVA
257–264
pep-
tide bound to H-2K
b
(19) as the source of anti-tumor-specific T
cells and the OVA-transfected EL4 cell line, E.G7, as the tumor
cells expressing a tumor-specific Ag, i.e., OVA (20). We demon-
strated that the naive OT-I T cells are functionally blind or igno-
rant of the OVA tumor Ag. This failure of OT-I T cells to respond
to this tumor was overcome by proper Ag presentation, as supplied
by peptide-pulsed professional APC, leading to an effective anti-
tumor immune response. These findings provide a relevant strategy
to overcome tumor Ag ignorance for cancer immunotherapy.
Materials and Methods
Animals
The OT-I TCR-transgenic mice (19) were maintained by breeding het-
erozygous OT-I TCR-transgenic mice to wild-type C57BL/6. The progeny
mice were screened by PCR for the expression of the OVA-TCR gene. Six-
to 10-wk-old female C57BL/6 or CD4-deficient mice on the C57BL/6
background were purchased from The Jackson Laboratory (Bar
Harbor, ME).
Cell lines
EL-4, a thymoma derived from the C57BL/6 mouse (H-2
b
), was obtained
from American Type Culture Collection (ATCC, Manassas, VA). OVA-
transfected EL-4, designated as E.G7 (20), was a gift from Dr. M. Bevan
(University of Washington, Seattle, WA). These cell lines were maintained
in RPMI 1640 containing 5% FCS, glutamine (30
g/ml), penicillin (100
U/ml), streptomycin (100
g/ml), and 2-ME (5 10
5
M) (complete
medium).
*Department of Microbiology and Immunology and
Department of Surgery, Divi-
sion of Surgical Oncology, University of Miami School of Medicine, Miami,
FL 33101
Received for publication March 24, 2000. Accepted for publication September
13, 2000.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
This work was supported by Grant DAMD17-98-1-8208 from the Department of
Defense.
2
Address correspondence and reprint requests to Dr. Thomas Malek, Department of
Microbiology and Immunology, University of Miami School of Medicine, 1600 NW
10th Avenue, Miami, FL 33136. E-mail address: tmalek@med.miami.edu
Copyright © 2000 by The American Association of Immunologists 0022-1767/00/$02.00
Abs and other reagents
OVA peptide (SIINFEKL) (21) was synthesized by Research Genetics
(Huntsville, AL). Directly conjugated mAbs, including CyChrome-conju-
gated anti-CD8
, PE-conjugated anti-mouse V
2 TCR, and FITC-conju-
gated anti-mouse V
5.1, 5.2 TCR, were purchased from PharMingen (San
Diego, CA). CFSE was purchased from Molecular Probes (Eugene, OR).
Cells were labeled with CFSE as previously described (22). Briefly, cells
(2 10
7
/ml) were incubated with 5
M CFSE (froma5mMstock in
DMSO) in serum-free medium for 10 min at 37°C and washed twice with
cold complete medium and twice with HBSS.
Tumor challenge and adoptive transfer of transgenic T cells
Normal C57BL/6 or CD4
/
mice were injected with 1 10
6
E.G7 cells
in 0.2 ml HBSS s.c. into the midline of the abdomen. The tumor cells were
freshly thawed and grown in culture for 5–10 days before each injection.
Spleen cell suspensions from heterozygous OT-I mice (6 wk of age) were
prepared as previously described (15). Splenocytes containing the indicated
number of transgenic OT-I T cells were injected i.v. in 0.5 ml HBSS 5–7
days after tumor challenge.
FACS analysis
Spleens and draining lymph nodes (LN)
3
(inguinal, brachial) were col-
lected and subjected to FACS analysis as previously described (15). Be-
tween 50,000 and 100,000 events per sample were collected on a FACScan
flow cytometer (Becton Dickinson, San Jose, CA) and analyzed using
CellQuest software (Becton Dickinson). OT-I-transgenic T cells were enu-
merated either by three-color staining for CD8, V
2-TCR, and V
5.1,
5.2-TCR or by the fraction of CSFE-labeled (FITC) OT-I spleen cells that
also stained for CD8. The latter approach is valid because essentially all
CD8
cells in OT-I mice express the transgenic TCR.
Purification of CD8
OT-I cells
OT-I T cells were purified by a combination of negative and positive se-
lection. First, B cells were depleted on anti-Ig-coated plates, followed by
further depletion by incubation of the nonadherent cells with anti-CD24
(JIID), anti-NK1.1, and anti-MHC class II and Cfor 45 min at 37°C. The
OT-I T cells were then subjected to positive selection using magnetic beads
containing anti-CD8 (Miltenyi Biotec, Auburn, CA). The resulting cells
were 95% pure as judged by FACS analysis.
T cell proliferation assay
T cells (2 10
5
/well) were cultured in flat-bottom 96-well plates in com-
plete medium containing the indicated stimuli for 3–4 days. EL4 or E.G7
cells were always irradiated (20,000 rad). Then 1
Ci [
3
H]thymidine was
added to the cultures for the last 5–6 h. The cells were harvested with an
automated cell harvester, and the radioactivity incorporated in DNA was
measured in a scintillation counter. Data of triplicate values that varied by
10% of the mean are displayed as cpm, i.e., cpm from experimental
culture minus cpm from cultures containing only medium.
Culture for APCs
A single-cell suspension of bone marrow cells from normal C57BL/6 mice
was cultured at 0.5 10
6
/ml in complete medium containing 2 ng/ml
murine GM-CSF (PeproTech, Rocky Hill, NJ). Four to 5 days later, ad-
herent cells were collected after incubation with PBS containing 5 mM
EDTA at 37°C for 15 min. The cells were washed with HBSS, incubated
with 1
M OVA peptide for1hat37°C, and washed three times with
HBSS. Mice were injected i.v. with the OVA-pulsed APC in 0.5 ml
of HBSS.
Generation and assay of CTL
Splenocytes (1 10
6
) from OT-I-transgenic mice were cultured in com-
plete medium containing 1 nM OVA peptide, 20 U/ml IL-2, and 40 U/ml
IL-4. After 3 days, the cells were collected, washed, and re-cultured at 1
10
5
cells/ml in complete medium containing 20 U/ml IL-2 and 40 U/ml
IL-4 for 2 days. The CTL activity of the cells was measured by a standard
51
Cr release assay as previously described (23) using
51
Cr-labeled EL4 or
E.G7 cells as the targets.
Results
Activation of OT-I in vitro
In this study, the OVA-specific MHC class I-restricted (H2
b
) OT-I
TCR-transgenic CD8
T cells were used as tumor-specific T cells
by adoptive transfer to mice bearing the E.G7 tumor (OVA-trans-
fected EL4) as a solid tumor. The presence of the OT-I cells in
vivo was assessed by the coexpression of V
5 and V
2ofthe
transgenic TCR on CD8
T cells. In our initial studies, we deter-
mined the potential to functionally measure low numbers of OT-I
T cells in peripheral lymphoid tissue by evaluating the prolifera-
tive response to OVA peptide or E.G7 in vitro. Dose-response
studies demonstrated that OT-I T cells developed strong prolifer-
ative responses to 10
7
–10
9
M OVA peptide, with detectable
responses in cultures containing as little as 10
12
M OVA peptide
(Fig. 1B). Furthermore, by mixing OT-I spleen cells with normal
C57BL/6 spleen cells in such a manner that the fraction of trans-
genic OT-I cells was predetermined, readily detectable prolifera-
tive responses were routinely generated by a relatively high dose
of OVA peptide (10
9
M) when the cultures contained as few as
1 10
3
OT-I T cells, which is only 0.5% of the total number of
spleen cells in culture (Fig. 1A). Thus, OT-I cells are exquisitely
sensitive to OVA peptide, and OT-I T cells were functionally de-
tected in lymphoid tissues when present at a frequency of 1 in 200.
The E.G7 cells used in this study secreted 560 pg of OVA/ml/
1 10
6
cells after a 24-h culture as determined by ELISA (data
not shown). The addition of as much as 1.0 mg/ml of native sol-
uble OVA protein induced only minimal proliferation of OT-I T
cells in vitro (data not shown). Despite this relatively low level of
3
Abbreviation used in this paper: LN, lymph node.
FIGURE 1. Response of OT-I cells in vitro. A, Response by limiting
number of OT-I cells. B, Dose response to OVA peptide. C, Response to
cell associated-OVA. D, Response by purified OT-I cells. C57BL/6 spleen
cells (2 10
5
/well) containing the indicated number of OT-I T cells (A)or
2 10
3
/well OT-I T cells (B and C) were stimulated for 3 days with the
indicated concentration of OVA peptide, anti-CD3 (5% supernatant), or the
indicated number of irradiated EL4, E.G7, or OVA-pulsed APC. D, A total
of 2.5 10
4
purified OT-I cells were stimulated for 3 days with the in-
dicated concentration of OVA peptide or the indicated number of irradiated
E.G7 in the presence or absence of 1 10
5
accessory cells. Data are
representative of two to five experiments.
6732 REVERSAL OF TUMOR-SPECIFIC T CELL IGNORANCE
secretion of OVA, the E.G7 cells were immunogenic, as the irra-
diated E.G7, but not the parental EL4 thymoma, induced prolifer-
ation by OT-I cells in a fashion comparable to peptide-pulsed APC
(Fig. 1C). Taken together, these data suggest that cell-associated
OVA is much more immunogenic than soluble OVA protein. Fur-
thermore, E.G7 directly stimulated substantial proliferation by
highly purified OT-I T cells in the absence of accessory cells (Fig.
1D). By contrast, the response of these purified T cells to soluble
OVA peptide was dependent upon accessory cells. These results
indicate that the E.G7 can directly present OVA to OT-I.
OT-I T cells lack anti-tumor activity in vivo
Given the potent immunogenicity of E.G7 for OT-I T cells in vitro,
we examined the ability of OT-I T cells to mount an anti-tumor
immune response in vivo. Five days after C57BL/6 mice received
10
6
E.G7 s.c., OT-I T cells were adoptively transferred to these
tumor-bearing mice, and tumor size was measured over time. Each
mouse received 2.5 10
6
OT-I T cells, yielding mice in which
5% of their CD8
T cells were OT-I. As reported by Jenkins and
coworkers (13), this number of transgenic T cells seeds the pe-
ripheral immune compartment so that the transgenic T cells are
present at a frequency that is still detectable by FACS analysis, but
these cells are not at such a high level as to cause imbalance in the
peripheral immune compartment. When compared with control
mice that did not receive OT-I cells, it is quite apparent that rate of
tumor growth was comparable between both groups of mice (Fig.
2). Transfer of a larger number (4 10
6
) of OT-I T cells still failed
to affect tumor cell growth (data not shown). Thus, OT-I cells did
not generate as obvious an anti-tumor response to E.G7 growing as
did a solid tumor.
To examine whether the adoptively transferred OT-I cells pro-
liferated to E.G7 in vivo, we determined the proportion of OT-I T
cells in the spleen and draining LN of tumor-bearing animals by
three-color FACS analysis by staining for CD8, V
2, and V
5. In
a normal mouse, 0.2% and 0.5% of spleen and LN cells, re-
spectively, express these three surface molecules. After adoptive
transfer of OT-I T cells, the fraction of cells bearing these markers
initially increased to 0.6% for the spleen and 1.3% for the
draining LN (Fig. 3A). Importantly, these numbers decreased over
time for both control and tumor-bearing animals to a level similar
to that seen in normal mice. The total spleen and LN recovery, and
the proportion of CD8
T cells from mice that were adoptively
transferred with OT-I cells, in the presence or absence of E.G7,
was always comparable (data not shown), further indicating that
there was no obvious lymphoid cell expansion by the tumor. These
findings suggest that OT-I T cells did not proliferate in response to
the E.G7 tumor.
To further evaluate whether OT-I cells responded to E.G7 in
vivo, CFSE-treated OT-I T cells were adoptively transferred to
E.G7-bearing mice. CFSE is an intracellular fluorescent label for
which the fluorescence intensity decreases proportionally upon cell
division (22). When CD8 cells in the draining LN and spleens of
control and tumor-bearing mice were subjected to FACS analysis,
the large majority of cells maintained the original fluorescence
FIGURE 2. Anti-tumor activity of naive OT-I cells. Normal C57BL/6
mice were injected with E.G7 (1 10
6
). Seven days later, mice received
OT-I T cells as indicated, and the tumor size was measured. Detectable
tumor is considered to be above 0.5 cm
2
. The mice were sacrificed when
the tumor reached a size of 2.0 cm
2
. Data shown contain eight mice/group
and are representative of three experiments.
FIGURE 3. OT-I cells do not respond to E.G7 in vivo. A, Proportion of
OT-I T cells in tumor-bearing mice. The percentage of V
2, V
5, CD8
three-color-positive cells was determined for the spleens and LN of normal
and tumor-bearing mice as described in Materials and Methods. Data
shown contain four mice/group and is representative of three experiments.
B, Cell division by OT-I cells. OT-I spleen cells were labeled with CFSE
before transferring to normal or tumor-bearing mice (2.5 10
6
). Some
mice received 0.5 10
6
OVA-pulsed APC 1 day later. Three days after
transfer of APC, LN cells were analyzed for cell division by two-color
FACS analysis. Shown are histograms of cells double-stained for CD8 and
CFSE. The percentage of cells that have divided, as seen by a reduction in
CFSE staining intensity, is listed in C. Quantitative analyses of five to six
mice in each group (mean SD), based on FACS histograms as shown in B.
6733The Journal of Immunology
intensity, a profile characteristic of cells that have not undergone
cell division. Representative FACS histograms are shown in Fig.
3B, and the results from the analysis of multiple mice are summa-
rized in Fig. 3C. Very few OT-I cells exhibited a decrease in CFSE
staining after adoptive transfer to normal or E.G7-bearing mice. In
contrast, in recipients that were adoptively transferred with CFSE-
treated OT-I cells and subsequently challenged with OVA-pulsed
APC, 50% of the CD8 T cells exhibited a reduced level of CFSE
staining (Fig. 3C), demonstrating a normal and strong response by
OT-I T cells upon encountering Ag in association with a profes-
sional APC. Thus, these data further indicate that naive OT-I T
cells are nonresponsive to E.G7 tumor in vivo.
Because the OT-I T cells were adoptively transferred to mice
with an established tumor, we considered that E.G7 might have
anergized or otherwise suppressed activation of the transgenic T
cells. To test this possibility, the proliferative responses in vitro by
splenic and LN T cells to OVA peptide were assessed 4–7 days
after adoptive transfer to normal or tumor-bearing mice. In both
groups of mice, not only were the OVA-specific proliferative re-
sponses comparable, but the percentage of OT-I cells recovered
from the spleen and LN were also very similar (Table I). This
finding indicates that the failed anti-tumor response by OT-I T
cells was not the result of Ag-specific nonresponsiveness induced
by E.G7 in vivo. Collectively, these data indicate that the failed
anti-tumor response by naive OT-I is largely due to immunological
ignorance of OVA in the context of E.G7.
OT-I effector T cells generate an anti-tumor response
Although E.G7 as a solid tumor routinely failed to activate naive
OT-I T cells in vivo, these cells remained as potential targets for
immune effector cells, if such cells were successfully induced. To
determine whether E.G7 was susceptible to OT-I effector cells, we
tested whether the adoptive transfer of preactivated OT-I T cells
generated an anti-tumor response. Before adoptive transfer, the
OT-I cells were cultured for 3 days with OVA peptide and exog-
enous IL-2 and IL-4, and the effector cells were further expanded
for 2 days by culture with only the cytokines. After this 5-day
culture period, the OT-I cells exhibited potent CTL activity to
E.G7, but not to EL4 (Fig. 4A). These effector CTL were adop-
tively transferred 5 days after s.c. injection of E.G7, and when
compared with control mice, the growth of the tumor was delayed,
especially at a relatively high dose (6 10
6
) of OT-I effector cells
(Fig. 4B). Thus, E.G7 was recognized by, and was accessible to, in
vitro-generated OT-I CD8
effectors.
Because naive OT-I T cells were readily activated in vivo by
OVA-pulsed APC (see Fig. 3), we next tested whether the in
vivo induction of OT-I effector cells also induced anti-tumor
immunity. Naive OT-I T cells were adoptively transferred to
mice 5 days after s.c. injection of E.G7, and 24 h later the mice
received OVA-pulsed bone marrow-derived APC. As shown
earlier, the tumor grew quickly in mice that received tumor in
the presence of naive OT-1 (Fig. 5A). By contrast, coadminis-
tration of OT-I and OVA-pulsed APC resulted in a substantial
delay in the progression of the tumor. In 40% of the mice, no
tumor was detected 45 days after injection of E.G7, and several
mice that were observed longer remained tumor-free on day 60.
Importantly, there was no inhibition of growth when tumor-
bearing mice received OVA-pulsed APC in the absence of OT-I
T cells. This result indicates that the inhibition of tumor growth
Table I. E.G7 tumor cells do not anergize OT-I cells
a
Expt.
Days after
Adoptive
Transfer Cells from:
% OT-I
b
cpm 10
3c
Spleen LN Spleen LN
1 7 Tumor-bearing mice 0.53 0.03 1.39 0.06 22.0 3.3 88.3 24.0
Normal mice 0.67 0.03 1.23 0.03 24.3 0.4 76.0 16.6
2 4 Tumor-bearing mice 0.56 0.08 1.50 0.30 15.0 4.2 92.0 12.3
Normal mice 0.55 0.12 1.48 0.21 13.5 3.1 101.3 7.4
3 4 Tumor-bearing mice 0.58 0.02 0.94 0.25 23.1 4.0 71.5 21.0
Normal mice 0.60 0.07 1.10 0.15 21.0 1.0 63.5 15.4
a
Normal C57BL/6 mice were injected with E.G7 (1 10
6
). Five day later mice received 4 10
6
OT-I cells (experiment 1) and 2.5 10
6
OT-I cells (experiments 2
and 3). Spleen and LN were harvested at the indicated days, after adoptive transfer, and were subjected to analyses. Data shown contain three to four mice per group in each
experiment.
b
Determined by FACS analysis of three color positive cells for CD8, V
2, and V
5.
c
Determined by proliferation of spleen and LN after culture for 3 days with 1 nM OVA peptide.
FIGURE 4. Activity of effector OT-I CTL cells. A, CTL activity in vitro
as measured by
51
Cr-release assay against E.G7 and EL4. B, Anti-tumor
activity of OT-I effector cells. Normal C57BL/6 mice were injected with
E.G7 (1 10
6
). Five days later, mice received the indicated number of T
cells and tumor size recorded as described in Fig. 2. Effector OT-I CTL
cells were generated as described in Materials and Methods. Data shown
contain five mice/group and are representative of two experiments.
6734 REVERSAL OF TUMOR-SPECIFIC T CELL IGNORANCE
is dependent upon the presence of the adoptively transferred
transgenic T cells. Thus, after appropriate activation either in
vitro or in vivo, E.G7 was susceptible to tumor (OVA)-specific
OT-I effector cells.
To determine whether this anti-tumor response required CD4
T cells, we transferred OT-I T cells to tumor-bearing CD4-defi-
cient mice (Fig. 5B). Similar to E.G7-bearing C57BL/6 normal
mice, the growth of E.G7 was delayed in the CD4
/
recipient
mice, if they received OVA-pulsed APC, and two of six mice
remained tumor-free after 50 days.
The E.G7 tumor eventually grew in all mice that received in
vitro-induced OT-I effector cells and in some mice that were stim-
ulated with OVA-pulsed APC in vivo. The E.G7 cells were ex-
cised from one such mouse in each of the treatment groups and
grown in culture for at least 7 days. These cells were then used as
targets for OT-I CTL generated by in vitro culture. The E.G7 cells
obtained from the mice treated with ex vivo-induced OT-I effector
cells were nearly as good targets for OVA-specific CTL as the
parental E.G7 (Fig. 6). Thus, the tumor outgrowth from this mouse
appears to be the result of a failure of the adoptively transferred
CTL to completely kill the tumor. However, E.G7 from the in vivo
APC-treated mice were not lysed by the OVA-specific CTL, sug-
gesting that tumor outgrowth in this case was caused by the se-
lection of a tumor variant that escaped the effector OT-I CTL. In
addition, ELISA analysis confirmed that these cells failed to se-
crete a detectable OVA level (data not shown). Collectively, these
data raise the possibility that in vivo-induced effector T cells in-
duced a more potent anti-tumor immune response than adoptive T
cell immunotherapy.
Discussion
In the present study, we showed that a tumor-immune response
could not be elicited in tumor-bearing mice after the adoptive
transfer of a relatively high number of naive transgenic tumor-
specific CD8
T cells. Importantly, as assessed at several different
time points after adoptive transfer, the tumor-specific transgenic T
cells were not activated to proliferate in both the spleen and the
draining LN in the presence of the growing solid tumor. The fail-
ure to activate the T cells was not because the tumor anergized
them, because OT-I T cells derived from tumor-bearing mice
readily proliferated upon Ag challenge in vitro. Therefore, our data
indicate that failed anti-tumor responses to E.G7 were due to im-
munological ignorance, i.e., a failure of the host immune system to
recognize OVA, in this case in the context of a tumor cell, during
the induction phase of the immune response.
It was surprising that the OT-I T cells remained ignorant of
E.G7 when adoptively transferred in vivo considering the robust
proliferative response of the T cells to irradiated E.G7 tumor cells
in vitro. The activation of OT-I T cells in vivo has been shown to
be dependent on Ag presentation by short-lived bone marrow-de-
rived APC within the draining LN (24). Furthermore, when OVA
is cell associated, e.g., in transgenic
-islet cells, activation of
OT-I T cells is dependent upon cross-priming, i.e., the
-islet-
associated OVA is ultimately processed by an exogenous class I
pathway and presented to OT-I T cells by professional APC (24).
Cross-priming is facilitated by either destruction of the Ag-con-
taining cells and/or a high level of expression of the cell-associated
Ag (25). In the case of
-islet cells, when cross-priming was not
facilitated, the OVA-containing
-islets were ignored by OT-I T
cells (26).
The failure of OT-I T cells to recognize E.G7 is not analogous
to that described for OVA-containing
-islets, because E.G7 were
able to directly present OVA to OT-I. It is likely that two factors
promoted ignorance of E.G7. First, ignorance in our model appears
to be at least in part the result of lack of contact between the OT-I
tumor-specific T cells and OVA associated with the E.G7 tumor
cells. Naive OT-I T cells appear not to readily migrate to the site
of the tumor, preventing their direct activation by E.G7. Consistent
with this hypothesis, so far we have not detected OT-I cells within
the tumor site as determined by using CFSE-labeled OT-I cells and
FACS analysis of the excised tumors (data not shown). Similarly,
E.G7 does not obviously traffic to the spleen or the LN following
s.c. or i.v. injections (data not shown). These observations suggest
that OT-I T cells do not encounter E.G7 either at the site of the
tumor or within the draining LN. Second, there is no indication
that the E.G7 was able to cross-prime OT-I T cells by host
APC. E.G7 produced relatively low levels of OVA, and minimal
destruction of the rapidly growing E.G7 tumor is expected, espe-
cially early after injection of the tumor, conditions that would not
favor cross-priming by the APC of the recipient mice. The obser-
vation that professional APC consistently activated OT-I T cells in
the spleen and draining LN of E.G7-bearing mice demonstrates
that ignorance to E.G7 is not due to a failure of the naive OT-I T
cells to ultimately migrate to secondary lymphoid tissue, or to
generalized immune suppression by E.G7, although we cannot ex-
clude other means by which OT-I are ignorant of E.G7.
FIGURE 5. Peptide-pulsed APC induce anti-tumor response by naive
OT-I cells. Normal C57BL/6 mice (A) or CD4-deficient mice (B) were
injected with E.G7 (1 10
6
). Five days later, the mice received naive OT-I
T cells (3 10
6
) and 1 day later peptide-pulsed APC (0.5 10
6
)as
indicated. The size of the tumor was recorded as described in Fig. 2. Data
shown contain five mice per group and are representative of two experi-
ments (A) and six mice per group (B).
FIGURE 6. Lysis of tumor cells from OT-I-treated mice by OT-I ef-
fector cells in vitro. Tumors were excised from treated mice, as designated,
grown in culture for at least 7 days, and labeled with
51
Cr to serve as targets
for OT-I CTL (as described in Materials and Methods).
51
Cr-labeled E.G7
and EL4 served as positive and negative control targets, respectively.
6735The Journal of Immunology
The value of using adoptive transfer of TCR- transgenic T cells
to mice bearing tumors transfected with a model Ag is that this
approach provides insight into the strength and duration of an anti-
tumor T cell response. So far, two major outcomes have been
observed in these types of studies. As we have seen for E.G7 as a
solid tumor, tumor-specific TCR-transgenic T cells have been re-
ported to be ignorant of L
d
-transfected AG104A fibrosarcoma (27)
and a glycoprotein of lymphocytic choriomeningitis virus after
transfection into either Lewis lung carcinoma or the MC57G fi-
brosarcoma (17, 18). In the latter case, T cell ignorance required
that the tumor be transplanted as a solid tumor fragment rather than
s.c. injection of a single-cell suspension. These observations and
our results suggest that T cell ignorance represents one important
reason for failed anti-tumor immunity. Our data indicate that im-
munological ignorance may pertain to tumor-specific T cells bear-
ing a high affinity for TCR, as OT-I T cells are extremely sensitive
to OVA-peptide and proliferate to as little as 1 pM of peptide in
vitro, a dose that is 1000-fold lower than that required to activate
the lymphocytic choriomeningitis virus-specific TCR-transgenic T
cells (17).
In other studies of the adoptive transfer of TCR-transgenic T
cells to Ag-transfected tumors, an initial transient anti-tumor re-
sponse was observed (14, 15, 28). In several cases, failed anti-
tumor immunity was shown to be due to anergy of the tumor-
specific T cells. This has been observed for both MHC class I- and
II-restricted TCR-transgenic T cells. Interestingly, in the case of
the MHC class I T cell response, OT-I T cells and E.G7 tumor cells
also served as the model system (15). In that study, the E.G7 cells
were injected into the peritoneal cavity 1 day after adoptive trans-
fer of the OT-I T cells, which led to initial activation and prolif-
eration of the OT-I T cells in vivo. Effective anti-tumor immunity
failed in part due to CTLA4-mediated down-regulation of endog-
enous CD4 helper activity (29). This finding markedly contrasts
with our result in which we found that OT-I T cells were ignorant
of E.G7 as a solid tumor. We have compared the E.G7 subline
maintained in our laboratory with that used by Srikant et al. (15)
and found that our subline failed to activate OT-I after injection
i.p. whereas the E.G7 subline used by Shrikant and coworkers
activated OT-I when present as a solid tumor. This indicates that
the different pattern of results is unlikely to be due to differences
in experimental protocols and/or responses to a systemic vs solid
tumor. Furthermore, both sublines of E.G7 produce similar levels
of OVA. Therefore, it is most likely that these two cell lines ex-
press some intrinsic undefined difference, as the cells were inde-
pendently passaged for a considerable period of time.
Although in our study naive OT-I T cells failed to inhibit the
growth of E.G7, anti-tumor immune responses were elicited by
OT-I effector cells, confirming that the OT-I T cells have sufficient
affinity to specifically attack the developing E.G7 solid tumor. Ef-
fector OT-I T cells were generated either ex vivo in culture or by
in vivo stimulation of the naive adoptively transferred OT-I T cells
with peptide-pulsed bone marrow-derived APC. This illustrates
that T cell ignorance can be overcome simply by proper Ag pre-
sentation of a tumor Ag. Furthermore, by using CD4-deficient
mice, we demonstrated that this anti-tumor activity was indepen-
dent of CD4 T cell help. Although many other studies clearly
indicate a need for CD4
T cells for anti-tumor responses (30–33),
our data, similar to that reported by Wick et al. (27), demonstrate
that direct activation of CD8 T cells can be sufficient for potent
anti-tumor immunity.
Several other studies have demonstrated that bone marrow-de-
rived APC effectively inhibit tumor growth (17, 34), perhaps by
activation of a population of ignorant tumor-specific T cells. It is
interesting to note that so far we have only “cured” E.G7 with OT-I
cells when the mice were immunized with OVA-pulsed APC.
These findings raise the prospect that effective anti-tumor immu-
nity may be facilitated by approaches that both increase the fre-
quency of tumor-specific T cells and induce activation of such T
cells by vaccination with tumor-Ag-containing APC.
Acknowledgments
We thank Dr. E. Codias for critically reading this manuscript,
P. Scibelli and T. Nguyen for technical assistance, and the Sylvester Com-
prehensive Cancer Center for support of the FACS facilities.
References
1. Boon, T., J. C. Cerottini, B. Van den Eynde, P. van der Bruggen, and A. Van Pel.
1994. Tumor antigens recognized by T lymphocytes. Annu. Rev. Immunol. 12:
337.
2. Pardoll, D. M. 1998. Cancer vaccines. Nat. Med. 4:525.
3. Chen, L., S. Ashe, W. A. Brady, I. Hellstrom, K. E. Hellstrom, J. A. Ledbetter,
P. McGowan, and P. S. Linsley. 1992. Costimulation of antitumor immunity by
the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell
71:1093.
4. Cohen, E. P., and T. S. Kim. 1994. Neoplastic cells that express low levels of
MHC class I determinants escape host immunity. Semin. Cancer Biol. 5:419.
5. Bretscher, P., and M. Cohn. 1970. A theory of self-nonself discrimination. Sci-
ence 169:1042.
6. Lafferty, K. J., and A. J. Cunningham. 1975. A new analysis of allogeneic in-
teractions. Aust. J. Exp. Biol. Med. Sci. 53:27.
7. Schwartz, R. H. 1990. A cell culture model for T lymphocyte clonal anergy.
Science 248:1349.
8. Ranges, G. E., I. S. Figari, T. Espevik, and M. A. Palladino, Jr. 1987. Inhibition
of cytotoxic T cell development by transforming growth factor
and reversal by
recombinant tumor necrosis factor
. J. Exp. Med. 166:991.
9. Becker, J. C., C. Czerny, and E. B. Brocker. 1994. Maintenance of clonal anergy
by endogenously produced IL-10. Int. Immunol. 6:1605.
10. Brodie, S. J., D. A. Lewinsohn, B. K. Patterson, D. Jiyamapa, J. Krieger,
L. Corey, P. D. Greenberg, and S. R. Riddell. 1999. In vivo migration and func-
tion of transferred HIV-1-specific cytotoxic T cells. Nat. Med. 5:34.
11. Yee, C., M. J. Gilbert, S. R. Riddell, V. G. Brichard, A. Fefer, J. A. Thompson,
T. Boon, and P. D. Greenberg. 1996. Isolation of tyrosinase-specific CD8
and
CD4
T cell clones from the peripheral blood of melanoma patients following in
vitro stimulation with recombinant vaccinia virus. J. Immunol. 157:4079.
12. Riddell, S. R., K. S. Watanabe, J. M. Goodrich, C. R. Li, M. E. Agha, and
P. D. Greenberg. 1992. Restoration of viral immunity in immunodeficient humans
by the adoptive transfer of T cell clones. Science 257:238.
13. Kearney, E. R., K. A. Pape, D. Y. Loh, and M. K. Jenkins. 1994. Visualization
of peptide-specific T cell immunity and peripheral tolerance induction in vivo.
Immunity 1:327.
14. Staveley-O’Carroll, K., E. Sotomayor, J. Montgomery, I. Borrello, L. Hwang,
S. Fein, D. Pardoll, and H. Levitsky. 1998. Induction of antigen-specific T cell
anergy: an early event in the course of tumor progression. Proc. Natl. Acad. Sci.
USA 95:1178.
15. Shrikant, P., and M. F. Mescher. 1999. Control of syngeneic tumor growth by
activation of CD8
T cells: efficacy is limited by migration away from the site
and induction of nonresponsiveness. J. Immunol. 162:2858.
16. Marzo, A. L., R. A. Lake, D. Lo, L. Sherman, A. McWilliam, D. Nelson,
B. W. S. Robinson, and B. Scott. 1999. Tumor antigens are constitutively pre-
sented in the draining lymph nodes. J. Immunol. 162:5838.
17. Hermans, I. F., A. Daish, J. Yang, D. S. Ritchie, and F. Ronchese. 1998. Antigen
expressed on tumor cells fails to elicit an immune response, even in the presence
of increased numbers of tumor-specific cytotoxic T lymphocyte precursors. Can-
cer Res. 58:3909.
18. Ochsenbein, A. F., P. Klenerman, U. Karrer, B. Ludewig, M. Pericin,
H. Hengartner, and R. M. Zinkernagel. 1999. Immune surveillance against a solid
tumor fails because of immunological ignorance. Proc. Natl. Acad. Sci. USA
96:2233.
19. Hogquist, K., C. Stephen, W. Heath, L. Jane, M. Beven, and F. Carbone. 1994.
T cell receptor antagonist peptides induce positive selection. Cell 76:17.
20. Moore, M., F. Carbone, and M. Beven. 1988. Introduction of soluble protein into
the class I pathway of antigen processing and presentation. Cell 54:777.
21. Carbone, F., S. Sterry, J. Butler, S. Rodda, and M. Moore. 1992. T cell receptor
-chain pairing determines the specificity of residue 262 within the K
b
-restricted,
ovalbumin
257–264
determinant. Int. Immunol. 4:861.
22. Lyons, A. B., and C. R. Parish. 1994. Determination of lymphocyte division by
flow cytometry. J. Immunol. Methods 171:131.
23. Liu, B., E. Podack, J. Allison, and T. Malek. 1996. Generation of primary tumor-
specific CTL in vitro to immunogenic and poorly immunogenic mouse tumors.
J. Immunol. 156:1117.
24. Kurts, C., W. R. Heath, F. R. Carbone, J. Allison, J. F. Miller, and H. Kosaka.
1996. Constitutive class I-restricted exogenous presentation of self antigens in
vivo. J. Exp. Med. 184:923.
25. Kurts, C., J. F. Miller, R. M. Subramaniam, F. R. Carbone, and W. R. Heath.
1998. Major histocompatibility complex class I-restricted cross-presentation is
6736 REVERSAL OF TUMOR-SPECIFIC T CELL IGNORANCE
biased toward high dose antigens and those released during cellular destruction.
J. Exp. Med. 188:409.
26. Kurts, C., R. M. Sutherland, G. Davey, M. Li, A. M. Lew, E. Blanas,
F. R. Carbone, J. F. A. P. Miller, and W. R. Heath. 1999. CD8 T cell ignorance
or tolerance to islet antigens depends on antigen dose. Proc. Natl. Acad. Sci. USA
96:12703.
27. Wick, M., P. Dubey, H. Koeppen, C. Siegel, P. Fields, L. Chen, J. Bluestone, and
H. Schreiber. 1997. Antigenic cancer cells grow progressively in immune hosts
without evidence for T cell exhaustion or systemic anergy. J. Exp. Med. 186:229.
28. Prevost-Blondel, A., C. Zimmermann, C. Stemmer, P. Kulmburg, F. Rosenthal,
and H. Pircher. 1998. Tumor-infiltrating lymphocytes exhibiting high ex vivo
cytolytic activity fail to prevent murine melanoma tumor growth in vivo. J. Im-
munol. 161:2187.
29. Shrikant, P., A. Khoruts, and M. Mescher. 1999. CTLA-4 blockade reverses
CD8
T cell tolerance to tumor by a CD4
T cell- and IL-2-dependent mecha
-
nism. Immunity 11:483.
30. Marzo, A. L., R. A. Lake, B. W. S. Robinson, and B. Scott. 1999. T-cell receptor
transgenic analysis of tumor-specific CD8 and CD4 responses in the eradication
of solid tumors. Cancer Res. 59:1071.
31. Hung, K., R. Hayashi, A. Lafond-Walker, C. Lowenstein, D. Pardoll, and
H. Levitsky. 1998. The central role of CD4
T cells in the antitumor immune
response. J. Exp. Med. 188:2357.
32. Nishimura, T., K. Iwakabe, M. Sekimoto, Y. Ohmi, T. Yahata, M. Nakui, T. Sato,
S. Habu, H. Tashiro, M. Sato, and A. Ohta. 1999. Distinct role of antigen-specific
T helper type 1 (Th1) and Th2 cells in tumor eradication in vivo. J. Exp. Med.
190:617.
33. Zitvogel, L., J. I. Mayordomo, T. Tjandrawan, A. B. DeLeo, M. R. Clarke,
M. T. Lotze, and W. J. Storkus. 1996. Therapy of murine tumors with tumor
peptide-pulsed dendritic cells: dependence on T cells, B7 costimulation, and T
helper cell 1-associated cytokines. J. Exp. Med. 183:87.
34. Gilboa, E., S. K. Nair, and H. K. Lyerly. 1998. Immunotherapy of cancer with
dendritic-cell-based vaccines. Cancer Immunol. Immunother. 46:82.
6737The Journal of Immunology
... Cells (1-2 × 10 6 ) were cultured in 24 well flat bottom plates with complete RPMI 1640 medium (29) containing PMA (50 ng/ml), ionomycin (1 μM) and brefeldin A (Biolegend) for 4 hr. Cells were harvested, washed, and stained for cell surface markers and then fixed, permeabilized, and stained to detect IL-2, IL-17, and IFNγ by flow cytometry. ...
Article
Full-text available
Resting central Tregs (cTregs) and activated effector Tregs (eTregs) are required for self-tolerance, but the heterogeneity and relationships within and between phenotypically distinct subsets of cTregs and eTregs are poorly understood. By extensive immune profiling and deep sequencing of TCR-β V regions, two subsets of cTregs, based on expression of Ly-6C, and three subsets of eTregs, based on distinctive expression of CD62L, CD69, and CD103, were identified. Ly-6C(+)cTregs exhibited lower basal activation, expressed on average lower affinity TCRs, and less efficiently developed into eTregs when compared with Ly-6C(-)cTregs. The dominant TCR Vβs of Ly-6C(+)cTregs were shared by eTregs at a low frequency. A single TCR clonotype was also identified that was largely restricted to Ly-6C(+)cTregs, even under conditions that promoted the development of eTregs. Collectively, these findings indicate that some Ly-6C(+)cTregs may persist as a lymphoid-specific subset, with minimal potential to develop into highly activated eTregs, whereas other cTregs readily develop into eTregs. In contrast, subsets of CD62L(lo)eTregs showed higher clonal expansion and were more highly interrelated than cTreg subsets based on their TCR-β repertoires, but exhibited varied immune profiles. The CD62L(lo)CD69(-)CD103(-)eTreg subset displayed properties of a transitional intermediate between cTregs and more activated eTreg subsets. Thus, eTreg subsets appear to exhibit substantial flexibility, most likely in response to environmental cues, to adopt defined immune profiles that are expected to optimize suppression of autoreactive T cells.
... The specific cytotoxicity of CD8 1 T cells was assayed as described previously 26,27 with minor modifications. Briefly, splenocytes from OT-I mice were primed with 10 mg/ml OVA 257-264 peptide as target cells. ...
Article
The Fas/FasL system transmits intracellular apoptotic signaling, inducing cell apoptosis. However, Fas signaling also exerts non-apoptotic functions in addition to inducing tumor cell apoptosis. For example, Fas signaling induces lung cancer tumor cells to produce prostaglandin E2 (PGE2) and recruit myeloid-derived suppressor cells (MDSCs). Activated cytotoxic T lymphocytes (CTLs) induce and express high levels of FasL, but the effects of Fas activation initiated by FasL in CTLs on apoptosis-resistant tumor cells remain largely unclear. We purified activated CD8(+) T cells from OT-1 mice, evaluated the regulatory effects of Fas activation on tumor cell escape and investigated the relevant mechanisms. We found that CTLs induced tumor cells to secrete PGE2 and increase tumor cell-mediated chemoattraction of MDSCs via Fas signaling, which was favorable to tumor growth. Our results indicate that CTLs may participate in the tumor immune evasion process. To the best of our knowledge, this is a novel mechanism by which CTLs play a role in tumor escape. Our findings implicate a strategy to enhance the antitumor immune response via reduction of negative immune responses to tumors promoted by CTLs through Fas signaling.Cellular & Molecular Immunology advance online publication, 28 April 2014; doi:10.1038/cmi.2014.21.
... In particular, the treatment of patients with advanced non-small cell lung cancer (NSCLC) is frequently complicated by co-morbid conditions and older age. 1 Thus, tumor vaccines may be ideal in this population due to their favorable toxicity profile. 2 Unfortunately, tumor-associated antigen (TAA) vaccination alone is usually insufficient to induce innate immunity, likely due to host immune incompetence and tumor-related immune suppression. 3 Therefore, strategies to induce or deregulate co-stimulatory protein interactions have been investigated. In particular, dendritic cells (DC) are the most potent antigen presenting cells (APC) that express co-stimulatory molecules. ...
Article
We created a vaccine in which irradiated allogeneic lung adenocarcinoma cells are combined with a bystander K562 cell line transfected with hCD40L and hGM-CSF. By recruiting and activating dendritic cells, we hypothesized that the vaccine would induce tumor regression in metastatic lung adenocarcinoma. Intradermal vaccine was given q14 days×3, followed by monthly ×3. Cyclophosphamide (300 mg/m IV) was administered before the first and fourth vaccines to deplete regulatory T cells. All-trans retinoic acid was given (150/mg/m/d) after the first and fourth vaccines to enhance dendritic cell differentiation. Twenty-four participants were accrued at a single institution from October 2006 to June 2008, with a median age 64 years and median of 4 previous lines of systemic therapy. A total of 101 vaccines were administered. Common toxicities were headache (54%) and site reaction (38%). No radiologic responses were observed. Median overall survival was 7.9 months and median progression-free survival was 1.7 months. Of 14 patients evaluable for immunological study, 5 had peptide-induced CD8 T-cell activation after vaccination. Overall, vaccine administration was feasible in an extensively pretreated population of metastatic lung cancer. Despite a suggestion of clinical activity in the subset with immune response, the trial did not meet the primary endpoint of inducing radiologic tumor regression.
Article
Ovarian cancer continues to be a difficult medical issue that affects millions of individuals worldwide. Important platforms for cancer immunotherapy include checkpoint inhibitors, chimeric antigen receptor T cells, bispecific antibodies, cancer vaccines, and other cell-based treatments. To avoid numerous infectious illnesses, conventional vaccinations based on synthetic peptides, recombinant subunit vaccines, and live attenuated and inactivated pathogens are frequently utilized. Vaccine manufacturing processes, however, are not entirely safe and carry a significant danger of contaminating living microorganisms. As a result, the creation of substitute vaccinations is required for both viral and noninfectious illnesses, including cancer. Recently, there has been testing of nucleic acid vaccines, or NAVs, as a cancer therapeutic. Tumor antigens (TAs) are genetically encoded by DNA and mRNA vaccines, which the host uses to trigger immune responses against ovarian cancer cells that exhibit the TAs. Despite being straightforward, safe, and easy to produce, NAVs are not currently thought to be an ideal replacement for peptide vaccines. Some obstacles to this strategy include selecting the appropriate therapeutic agents (TAs), Cell Biochem Funct. 2024;42:e3978. wileyonlinelibrary.com/journal/cbf
Article
The cytokine interleukin-2 (IL-2) is critical for the functions of regulatory T cells (Tregs). The contribution of polymorphisms in the gene encoding the IL-2 receptor α subunit (IL2RA), which are associated with type 1 diabetes, is difficult to determine because autoimmunity depends on variations in multiple genes, where the contribution of any one gene product is small. We investigated the mechanisms whereby a modest reduction in IL-2R signaling selectively in T lymphocytes influenced the development of diabetes in the NOD mouse model. The sensitivity of IL-2R signaling was reduced by about two- to threefold in Tregs from mice that coexpressed wild-type IL-2Rβ and a mutant subunit (IL-2RβY3) with reduced signaling (designated NOD-Y3). Male and female NOD-Y3 mice exhibited accelerated diabetes onset due to intrinsic effects on multiple activities in Tregs. Bone marrow chimera and adoptive transfer experiments demonstrated that IL-2RβY3 Tregs resulted in impaired homeostasis of lymphoid-residing central Tregs and inefficient development of highly activated effector Tregs and that they were less suppressive. Pancreatic IL-2RβY3 Tregs showed impaired development into IL-10–secreting effector Tregs. The pancreatic lymph nodes and pancreases of NOD-Y3 mice had increased numbers of antigen-experienced CD4⁺ effector T cells, which was largely due to impaired Tregs, because adoptively transferred pancreatic autoantigen–specific CD4⁺ Foxp3⁻ T cells from NOD-Y3 mice did not accelerate diabetes in NOD.SCID recipients. Our study indicates that the primary defect associated with chronic, mildly reduced IL-2R signaling is due to impaired Tregs that cannot effectively produce and maintain highly functional tissue-seeking effector Treg subsets.
Chapter
Recognition of an antigen by specific T lymphocytes carrying the appropriate antigen receptor does not always lead to activation or inactivation. Many studies have found that such lymphocytes could remain in resting status after exposure to the antigen, a phenomenon described as “ignorance” or “indifference.” Immunological ignorance, although first introduced more than 10 years ago, remains a poorly understood phenomenon in the field of lymphocyte biology. Experimental data indicates that many tumor antigens are ignored by tumor-specific T cells in tumor-bearing animals and in cancer patients. In this chapter, we summarize the current status of research and the methods that may be utilized to overcome T-cell ignorance with the goal of achieving more effective antitumor T-cell responses.
Article
Objective: To study the role of TH cell in the activation of memory T cells and shifting to functional tumor killer cells. Methods: Adoptive transfers SIINFEKL specific CTL to RAG-/- mice and then immunized them with SIINFEKL to generate ovalbumin specific memory CTL. C57BL/6 mice were adoptive transferred with memory CTL immunized with T-helper epitopes of ovalbumin and then challenged with ovalbumin specific tumor cells. Results: With SIINFEKL immunization, specific CTL proliferated and acquired memory CTL phenotypes; OVA specific TH cells could help the host to generate more specific CTL but this effect is not strong enough to get tumor inhibition. Memory CTL needs TH cells to completely regress the development of tumor formation. Conclusion: Long lasting antitumor effect needs both antigen specific memory CTL and specific TH cells and so the design of tumor vaccine should include specific CTL and TH peptides.
Article
Naïve T cells in the draining lymph node (DLN) do not immediately respond to antigenic tissues or antigenic cancers in the periphery. Rather, the conditions under which naïve T cells encounter antigen in the DLN can result in the distinct immunological states of ignorance, tolerance or immunity. Recent work suggests that these immunological states are determined by the level of antigen expressed by peripheral tissues and the maturation stage of the dendritic cell presenting the antigen. When antigens are expressed at levels that are sufficient to be cross-presented by mature dendritic cells in the DLN, naïve T cells can respond to self antigens or tumor antigens to induce a state of autoimmunity or tumor immunity, respectively. Exploiting these conditions to target unique tumor antigens will enable us to develop better cancer immunotherapies.
Article
Several recent immunotherapy agents have exhibited exceptional activity, and their eventual approval for use in lung cancer appears plausible. The immune checkpoint proteins, such as the B7 superfamily, are becoming increasingly relevant targets for therapeutic inhibition. Tumor vaccines hold the potential to deliver durable responses that are specific for tumor antigen, with favorable adverse effect profiles. Several vaccine trials are accruing more patients than any previous lung cancer trials and are designed to select a specific population based on a predefined, scientifically justified biomarker. These emerging immune treatments may hold great potential for the systemic treatment of lung cancers.
Article
During homeostatic expansion, peripheral T cells proliferate in response to lymphopenia, in the absence of cognate antigen or costimulatory signals. Host CD8 reconstitution following hematopoietic cell transplantation (HCT) involves the de novo-generation of T cells in addition to the homeostatic expansion of mature donor T cells present in the graft and donor lymphocyte infusion as well as host T cells that survive conditioning. Although it is well appreciated that CD8 homeostatic expansion contributes to host CD8 reconstitution following HCT, the factors governing the extent of the contribution by donor CD8 homeostatic expansion to host reconstitution have not been precisely and systematically examined. The ex vivo generation of memory CD8 T cells specific for an epitope of the immunodominant minor histocompatibility antigen H60 was demonstrated. By adapting a previously described culture system designed to generate large numbers of transgenic memory CD8 T cells, memory CD8 cells (TM) specific for a physiological antigen were elicited from a heterogeneous population of CD8 T cells. The ex vivo-generated antigen-specific memory CD8 cells were then expanded under conditions of lymphopenia in ablatively conditioned syngeneic transplant recipients and found to persist greater than 2 months post-transplant. These findings support the notion that the transplantation of small numbers ex vivo-generated memory CD8 cells, specific for a physiologically relevant antigen, can help restore host immune function following HCT. Transplant conditions were found to modulate the contribution of homeostatically expanded donor CD8 TM to the host CD8 compartment. Varying the conditioning intensity, timing of infusion, and infusion dose affected the kinetics of expansion as well as the homeostatic set-point. In my HCT model, delayed infusions of 3 weeks and transplantation of small numbers of donor CD8 T cells resulted in significant contributions to host CD8 T cell compartment. These findings could be applied clinically to enhance the effectiveness of clinical immunotherapy in restoring host immune function post-transplant. Moreover, transplantation of varying doses of donor CD8 TM demonstrated there was maximal donor contribution to host CD8 reconstitution. Finally, the homeostatic expansion, persistence, and function of transgenic memory and naïve CD8 populations were investigated following transplantation into ablatively conditioned syngeneic recipients. Both donor CD8 populations underwent 2 weeks of expansion post-transplant. The naïve CD8 population reproducibly achieved higher homeostatic numbers than the memory CD8 population. By one month post-HCT, the transplanted naïve CD8 populations also exhibited a memory CD8 phenotype. Moreover, the CD8 TN→TM population mediated an effective response to a primary challenge, comparable to the antigen-experienced memory CD8 population. Thus, regardless of the state of differentiation of the donor CD8 population at the time of transplant, donor CD8 homeostatic expansion contributes a memory CD8 cells population, able to mediating effective immune responses, to host reconstitution.
Article
The identification of tumor-associated Ags recognized by CD8+ CTL and prevention of tumor outgrowth by adoptive transfer of these CTL demonstrates that CD8+ T cells play a major role in antitumor immunity. We have generated B16.F10 melanoma cells that express the glycoprotein epitope amino acid 33-41 (GP33) of the lymphocytic choriomeningitis virus (LCMV) to examine antitumor CD8+ T cell response in C57BL/6 mice immune to LCMV and in mice transgenic for the LCMV GP33-specific P14 TCR (P14 TCR mice). We find that B16.F10GP33 tumor cells grew in syngeneic C57BL/6 mice without inducing T cell tolerance. LCMV infection or adoptive transfer of LCMV-specific effector T cells delayed but did not prevent growth of preestablished tumors in these mice. However, B16.F10GP33 tumor cells were rejected in mice immune to LCMV and in mice treated with LCMV-specific effector T cells on the same day as the tumor. Surprisingly, B16.F10GP33 tumor cells grew in P14 TCR transgenic mice despite an abundance of tumor-associated Ag-specific CD8+ T cells. In these mice, freshly isolated tumor-infiltrating lymphocytes exhibited an activated phenotype and displayed high GP33-specific cytolytic activity when assessed ex vivo. Thus, B16.F10GP33 melanoma cells are able to initiate, but not to sustain, a GP33-specific CTL response sufficient to clear the tumor enduringly.
Article
Transplantation experiments have demonstrated that most mouse tumors express antigens that can constitute targets for rejection responses mediated by syngeneic T lymphocytes. For human tumors, autologous cultures mixing tumor cells and blood lymphocytes or tumor-infiltrating lymphocytes have produced CD8(+) and CD4(+) cytolytic T cell (CTL) clones that recognize tumor cells specifically. Attempts to identify the target antigens by biochemical fractionation of tumor cells up to now have failed, with the important exception of the identification of underglycosylated mucins present on breast and pancreatic carcinomas. Gene transfection approaches have proved more successful. A gene family named MAGE codes for antigens recognized by autologous CTL on a melanoma tumor. These genes are not expressed in normal tissues except for testis. They are expressed in many tumors of several histological types. Differentiation antigens coded by genes such as tyrosinase are also recognized on human melanoma by autologous CTL. The identification of human tumor rejection antigens opens new possibilities for systematic approaches to the specific immune therapy of cancer.
Article
The immunoregulatory effects of transforming growth factor beta (TGF-beta) and recombinant murine tumor necrosis factor alpha (rMuTNF-alpha) on CTL generation and activity were examined. The results demonstrate that TGF-beta, in a dose-dependent manner, inhibited CTL generation but not CTL activity. The inhibitory effects were detected only when TGF-beta was added within the first 48 h of the MLC. Little activity was seen when it was added thereafter, including the addition of TGF-beta to the cytotoxicity assay. The production of TNF-alpha, which occurs during early phases of the MLC and which is inhibited in the presence of TGF-beta, appears to have an important regulatory role, as altering the levels of TNF-alpha in an MLC can significantly influence CTL development. The inhibitory effects of TGF-beta on the MLC can be significantly reversed by the addition of rMuTNF-alpha to the cultures. These results demonstrate that TGF-beta can inhibit MLC and subsequent CTL generation at early stages of the reaction, and such inhibition may involve the suppression of TNF-alpha production.
Article
Ovalbumin-specific, Kb-restricted T cells recognize the minimal fully active synthetic peptide ovalbumin (OVA)257–264. This sequence coincides with the eight residue, allele-speclflc peptide binding motif previously predicted from direct sequencing of naturally occurring Kb-associated peptides (Falk, K., Rotzscke, O., Stevanovic, S., Jung, G., and Rammensee, H.-G., Nature 351: 290, 1990). T cell recognition of a panel of analogs with single residue substitutions between the two putative Kb anchor residues at OVA261 and OVA264 suggested that at least one residue, Glu at position 262, is involved in TCR interaction. OVA-specific cytotoxic T lymphocytes (CTL) derived from TCR β-chaln transgenic mice, where the β-chain originates from an OVA-specific, Kb-restricted CTL B3, showed that differences in TCR a-chaln pairing determined the specificity for OVA residue 262. These data support the notion that residue 262 of the OVA T cell determinant, corresponding to position 6 within the Kb-binding motif, represents a contact site for TCR. This residue interacts directly with the TCR α-chain or with a site on the TCR β-chaln whose conformation is affected by TCR α-chaln pairing.
Article
Allogeneic reactions have conventionally been considered as typical immune responses by one population of cells to antigens present on the other. This view is inadequate, since it does not explain many features of these reactions, among which are: (1) reactivity is much higher between different strains within a species than between species, in spite of the much greater antigenic disparity in the second case; (2) a very high proportion of cells may respond to allogeneic stimuli; (3) major histocompatibility differences are not essential for vigorous allogeneic reactions; (4) the responding population need not be immunologically competent to respond to antigens of the stimulating population; (5) the stimulating population must be both metabolically active and immunocompetent.
Article
Interaction of the B7 molecule on antigen-presenting cells with its receptors CD28 and CTLA-4 on T cells provides costimulatory signals for T cell activation. We have studied the effects of B7 on antitumor immunity to a murine melanoma that expresses a rejection antigen associated with the E7 gene product of human papillomavirus 16. While this E7+ tumor grows progressively in immunocompetent hosts, cotransfection of its cells with B7 led to tumor regression by a B7-dependent immune response mediated by CD8+ cytolytic T lymphocytes. The immune response induced by E7+B7+ tumor cells also caused regression of E7+B7- tumors at distant sites and was curative for established E7+B7- micrometastases. Our findings suggest that increasing T cell costimulation through the CD28 and CTLA-4 receptors may have therapeutic usefulness for generating immunity against tumors expressing viral antigens.
Article
The adoptive transfer of antigen-specific T cells to establish immunity is an effective therapy for viral infections and tumors in animal models. The application of this approach to human disease would require the isolation and in vitro expansion of human antigen-specific T cells and evidence that such T cells persist and function in vivo after transfer. Cytomegalovirus-specific CD8+ cytotoxic T cell (CTL) clones could be isolated from bone marrow donors, propagated in vitro, and adoptively transferred to immunodeficient bone marrow transplant recipients. No toxicity developed and the clones provided persistent reconstitution of CD8+ cytomegalovirus-specific CTL responses.