ArticlePDF Available

Dendritic cell subsets in the peritoneal fluid and peripheral blood of women suffering from ovarian cancer

Authors:
  • Medical University in Lublin; al. Racławickie 1; 20-059 Lublin; Poland

Abstract and Figures

Evaluation of immature myeloid and lymphoid dendritic cells (DCs) in the peritoneal fluid (PF) and peripheral blood (PB) mononuclears of women with ovarian carcinoma (n = 47) and benign ovarian tumors (n = 37). Mononuclear cells were isolated from PF and PB, stained with monoclonal antibodies (mAbs) against DC antigens (anti-BDCA-1, anti-BDCA-2), and estimated using flow cytometry. The percentage of PF myeloid DC (MDC) in mononuclears was significantly lower in patients with ovarian cancer in comparison to the group of nonmalignant ovarian tumors (0.65% and 6.95%). The percentage of PF lymphoid DCs (LDCs) was higher in patients with ovarian cancer than in the reference group (0.64% and 0.09%). The percentage of PB MDCs and LDCs did not differ significantly between studied groups. In women suffering from ovarian cancer the percentage of both MDCs and LDCs was higher in the PF than in the PB. In the reference group the percentage of MDCs was higher but that of LDCs was lower in the PF than in the PB. In women with ovarian cancer, PF MDCs/LDCs ratio was lower in comparison to patients with serous cystadenoma. In PB the ratio of MDCs to LDCs did not differ significantly between studied groups. We concluded that MDCs population may be affected by the presence of malignant disease. LDC subsets may have influence on the local immune response in the PF of women with malignant tumors of the ovary. (c) 2008 Clinical Cytometry Society.
Content may be subject to copyright.
Dendritic Cell Subsets in the Peritoneal Fluid
and Peripheral Blood of Women Suffering
from Ovarian Cancer
I. Wertel,
1
*G. Polak,
1
W. Bednarek,
1
B. Barczyn
´ski,
1
J. Rolin
´ski,
2
and J. Kotarski
1
1
1st Department of Gynaecology, University School of Medicine, Lublin, Poland
2
Department of Clinical Immunology, University School of Medicine, Lublin, Poland
Background: Evaluation of immature myeloid and lymphoid dendritic cells (DCs) in the peritoneal fluid
(PF) and peripheral blood (PB) mononuclears of women with ovarian carcinoma (n547) and benign
ovarian tumors (n537).
Methods: Mononuclear cells were isolated from PF and PB, stained with monoclonal antibodies
(mAbs) against DC antigens (anti-BDCA-1, anti-BDCA-2), and estimated using flow cytometry.
Results: The percentage of PF myeloid DC (MDC) in mononuclears was significantly lower in patients
with ovarian cancer in comparison to the group of nonmalignant ovarian tumors (0.65% and 6.95%).
The percentage of PF lymphoid DCs (LDCs) was higher in patients with ovarian cancer than in the refer-
ence group (0.64% and 0.09%). The percentage of PB MDCs and LDCs did not differ significantly
between studied groups. In women suffering from ovarian cancer the percentage of both MDCs and LDCs
was higher in the PF than in the PB. In the reference group the percentage of MDCs was higher but that
of LDCs was lower in the PF than in the PB. In women with ovarian cancer, PF MDCs/LDCs ratio was
lower in comparison to patients with serous cystadenoma. In PB the ratio of MDCs to LDCs did not differ
significantly between studied groups.
Conclusions: We concluded that MDCs population may be affected by the presence of malignant dis-
ease. LDC subsets may have influence on the local immune response in the PF of women with malignant
tumors of the ovary. q2008 Clinical Cytometry Society
Key terms: myeloid DCs; lymphoid DCs; peritoneal fluid; cystadenocarcinoma
How to cite this article: Wertel I, Polak G, Bednarek W, Barczyn
´ski B, Rolin
´ski J, Kotarski J. Dendritic cell subsets
in the peritoneal fluid and peripheral blood of women suffering from ovarian cancer. Cytometry Part B 2008;
74B: 251–258.
INTRODUCTION
Epithelial ovarian carcinoma (EOC) is the main cause
of death in women with gynecological malignancies. At
the time of diagnosis, 75% of patients with EOC have
advanced intraperitoneal disease (1). Standard treatment
includes surgery, combined chemotherapy or/and radia-
tion, with 90% of the patients developing a recurrence
(2). The 5-year survival rate in these patients, according
to the different data, does not exceed 15% (1).
It has been well documented that multiple mechanisms
may be involved in the development and progression of a
tumor, including a defect in the immune system at the
induction and/or effector phases of antitumor immune
responses, such as antigen presentation in the absence of
major histocompatibility complex (MHC) Class II mole-
cules or costimulatory molecules. The majority of tumor
antigens (Ags) are nonmutated self-antigens to which the
immune system is tolerant. As a result, tumor Ags may
not be effectively presented to the immune system, or T
cells responding to tumor Ags may be anergized (3,4).
One of the most promising strategies to correct such
*Correspondence to: I. Wertel, 1st Department of Gynaecology, Uni-
versity School of Medicine, ul. Staszica 16, 20-081 Lublin, Poland.
E-mail: iwonawertel@gmail.com
Grant sponsor: State Committee for Scientific Research (Warsaw,
Poland); Grant number: 2 PO5E 120 27.
Received 22 October 2007; Accepted 20 January 2008
Published online 26 February 2008 in Wiley InterScience (www.
interscience.wiley.com).
DOI: 10.1002/cyto.b.20410
Cytometry Part B (Clinical Cytometry) 74B:251–258 (2008)
q2008 Clinical Cytometry Society
defects is the enhancement of tumor antigen presentation
with the help of dendritic cells (DCs), the most powerful
inducers of tumor-specific (CD8
1
) cytotoxic T-lymphocyte
responses. Unlike other antigen-presenting cells (APCs),
such as B cells and macrophages, DCs are capable of ini-
tiating not only secondary immune responses but also pri-
mary immune responses (such as activation of the naive
T cells) that are directed against specific antigens (5–8).
The DC system represents a heterogeneous group of
APC differing at the level of precursor cells, factors influ-
encing growth and maturation, phenotype, and APC
function (5,6,8). Circulating DC precursors home to
lymphoid and nonlymphoid tissues where they reside as
immature cells. At this stage, DCs are well equipped to
acquire antigens but nevertheless express low levels of
the requisite MHC and costimulatory molecules needed
for T-lymphocyte stimulation. Following antigen engulf-
ment and processing, DCs migrate to secondary lymph-
oid organs where they mature and become APCs that
are able to select and activate naive Ag-specific T cells
and induce an Ag-specific immune response (5,8).
Morphologically and phenotypically distinct DCs,
which are present at many different anatomical sites, are
derived from two lineages: myeloid or lymphoid (9,10).
Myeloid DCs (DC-1) are a major subpopulation of human
peripheral blood (PB) DCs, which are CD4
1
, Lin
2
,
CD11c
bright
, CD123
dim
, CD45RO
1
, and CD2
1
. They
express myeloid markers (CD13, CD33) as well as Fc
receptors (CD32, CD64, FceRI). Myeloid DCs (MDCs)
also express the blood DC antigen-BDCA-1 (CD1c) that
is specific for immature PB MDCs (6,11).
Lymphoid DCs (DC-2) have been described recently in
human PB (12), lymphoid tissue (13), and in the perito-
neal fluid (PF) (14,15). DC-2 cells express a specific
BDCA-2 (CD303) marker. Immunophenotyping of BDCA-
2
1
DC characterizes these cells as being CD4
1
, Lin
2
,
CD11c
2
, CD123
bright
, CD45RA
1
, CD2
2
, and expressing
neither myeloid lineage markers nor Fc receptors (11).
Almand et al. (16) detected that in the PB of patients
with breast, head and neck, and lung cancers the num-
ber of DCs was dramatically reduced. Similar results
were found by Yanagimoto et al. (17) in patients with
pancreatic cancer. The decrease of DCs in the PB from
these patients closely correlated with the stage and dura-
tion of the disease (16). However, there is a lack of
detailed information relating to the presence and a role
of DC subsets in the microenvironment of nonmalignant
and malignant body fluids.
The purpose of our study was to establish if the imma-
ture MDCs and lymphoid DCs (LDCs) are present in the
PF and PB of patients with ovarian cancer.
METHODS
Patients
Of 84 women, 47 patients, aged 30–85 years, were
found to have ovarian epithelial cancer. In the study
group, the grade was I in one case, II in 18 cases, and
III in 28 cases. The levels of preoperative Ca-125 marker
ranged from 45.71 to 11,179.00 with the median
1,747.72 U/ml. The patient profiles are presented in Ta-
ble 1. None of the women received chemotherapy
before operation.
Because sterilization is forbidden by Polish law, there
was no possibility to obtain PF from healthy, fertile
women. Thus we decided to choose the reference group
of the 37 patients with serous cystadenoma. The study
was approved by the Lublin University School of Medi-
cine Ethics Committee.
Cell Preparation
PF and PB were taken into heparinized tubes (sodium
heparin). In the noncancer group, all visible PF was aspi-
rated during surgical procedure from the anterior and
posterior cul-de-sacs, under direct vision to avoid blood
contamination. All women had venous blood samples
collected before the surgical procedure. PF and PB
mononuclear cells were isolated by density gradient cen-
trifugation on Lymphoprep (Nycomed, Norway) for
25 min at 600gat room temperature. Interphase cells
were collected and washed twice in phosphate-buffered
saline (PBS). The cells were counted in a Neubauer
chamber. The results were expressed as the number of
mononuclear cells per milliliter of PF and PB.
The cell surface antigens were determined on fresh
cells at the time of sample submission. Isolated cells
(1 310
6
) were incubated for 20 min at 48C with mono-
clonal antibodies (mAbs) specified against DC surface
antigens and washed twice in PBS containing 0.2 mM
ethylene-diaminetetraacetic acid and 0.5% bovine serum
albumin. The following directly conjugated mAbs were
used: anti-BDCA-1 (CD1c) FITC, anti-BDCA-2 (CD303)
FITC (Miltenyi Biotec, Bergisch Gladbach, Germany), and
anti-CD19 CyChrome, anti-CD123 PE (Pharmingen, San
Diego, CA). Mouse anti-human IgG
2a
isotype control was
used for anti-CD1c staining. Mouse anti-human IgG
1
iso-
type control was used for anti-BDCA-2 staining.
Flow Cytometric Analysis
Flow cytometric analysis of stained samples was per-
formed on FacsCanto instrument (Becton Dickinson, San
Jose, CA). A total of 300,000 events were acquired and an-
alyzed using FacsDiva software. Cell debris and dead cells
were excluded from the analysis based on scatter signals.
In our study, we identified immature MDCs as BDCA-1
(CD1c)
1
CD19
2
cells. BDCA-1 (CD1c) marker is also
expressed on a subpopulation of CD19
1
small resting B
lymphocytes. Therefore, the mononuclear cell analysis
Table 1
Clinical Characteristic of the Tumors
Histologic type Number of patients
Serous cystadenocarcinoma 21
Undifferentiated carcinoma 14
Mucinous cystadenocarcinoma 8
Clear cell carcinoma 4
Serous cystadenoma 37
252 WERTEL ET AL.
Cytometry Part B: Clinical Cytometry
region was analyzed for BDCA-1 (CD1c) and CD19 stain-
ing and BDCA-1 (CD1c)
1
B cells were excluded from
CD1c
1
DCs by counterstaining for CD19. Next, the
mononuclear cell analysis region was analyzed for BDCA-
2 and CD123 antigens. BDCA-2
1
CD123
1
cells were
counted as immature LDCs as described by Dzionek
et al. (11). The identification of DCs by flow cytometry
in the PF is presented in Figure 1.
We expressed the results as a percentage of MDC and
LDC in mononuclear cells. In addition, we calculated the
MDCs/LDCs ratio (M/L ratio) and the concentration of
DCs per milliliter of PB and PF.
Statistical Analysis
Statistical analysis was performed using nonparametric
tests. All data were presented as medians with the inter-
quartile ranges (25th to 75th). Wilcoxon paired test was
used to compare the results of PF and PB. Mann–Whit-
ney U test was applied to the results of statistical com-
parison between the studied groups. Statistical compari-
sons among more than two groups were performed
using Kruskal–Wallis test for independent samples.
Spearman’s rank test was used to assess the relationship
between concentrations of mononuclear cells and con-
centrations of DCs. Pvalue less than 0.05 was consid-
ered statistically significant.
RESULTS
The concentration of mononuclears and DC subsets in
the PF and PB of patients with ovarian tumors are pre-
sented in Figure 2.
Concentration of Mononuclears and DCs in the PF and PB
of Women with Ovarian Cancer and Serous Cystadenoma
The concentration of mononuclear cells was signifi-
cantly higher in the PF of patients with ovarian cancer
(P<0.001) than in women with serous cystadenoma.
The concentration of MDCs was significantly lower in
the PF of patients with ovarian cancer (P<0.001) than
in women with benign tumors (Figure 2). In contrary,
the concentration of LDCs was higher (P<0.001) in
the PF of patients with malignant disease than in the ref-
erence group. The concentration of mononuclears,
MDCs, and LDCs in the PB did not differ significantly
between studied groups (Figure 2).
The Percentage of DCs in the PF and PB of Women with
Ovarian Cancer
The percentage of MDC and LDC subsets in mononu-
clears was significantly higher in the PF than in the PB
(Figure 3).
The MDC to LDC ratio in patients with ovarian cancer
did not differ significantly between PF (median, 1.0;
interquartile range, 0.45–2.46) and PB (median, 1.0;
interquartile range, 0.60–2.03).
The Percentage of DCs in the PF and PB
of Women with Serous Cystadenoma
The percentage of MDCs in the PF was 6.95% (inter-
quartile range, 3.56–13.32%) and was significantly higher
than in the PB (0.23%; interquartile range, 0.15–0.26%).
However, the percentage of LDCs was significantly
higher in the PB (0.20%; interquartile range, 0.13–0.27%)
FIG. 1. The identification of PF dendritic cells by flow cytometry
(P1). The mononuclear cell analysis region applied to light scatters.
The P1-gated events were analyzed for BDCA-1 (CD1c) and CD19
staining, and BDCA-11CD192cells were counted as immature MDCs
(P2). The P1-gated events were then analyzed for BDCA-2 and CD123,
and BDCA-21CD1231cells were counted as LDCs (P3).
253DENDRITIC CELLS IN OVARIAN CANCER PATIENTS
Cytometry Part B: Clinical Cytometry
compared with PF (0.09%; interquartile range, 0.05–
0.17%).
The MDC to LDC ratio in women with benign ovarian
tumors was significantly higher in the PF (median, 71.2;
interquartile range, 33.12–129.41) than in the PB (me-
dian, 1.0; interquartile range, 0.71–1.37).
The Percentage of DCs in the PF and PB of Women with
Ovarian Cancer and Benign Tumors
The percentages of PF mononuclear cells that were
identified as MDC and LDC in women with ovarian
tumors are presented in Figures 3a and 3b.
The percentage of MDCs was significantly lower in
the PF of patients with ovarian cancer (0.65%; interquar-
tile range, 0.24–1.36%) than in women with benign
tumors (6.95%; interquartile range, 3.56–13.32%). In con-
trary, the percentage of LDCs was higher in the PF of
patients with malignant disease (0.64%; interquartile
range, 0.29–1.43%) than in the reference group (0.09%;
interquartile range, 0.05–0.17%). We also found that the
ratio of MDCs to LDCs in the PF was different between
women with malignant (1.0%; interquartile range, 0.45–
2.46) and benign (71.2; interquartile range, 33.12–
129.41) ovarian tumors.
The percentage of MDCs and LDCs and the ratio of
MDCs to LDCs in the PB did not differ significantly
between studied groups (1.0; interquartile range, 0.60–
2.03 vs. 1.0; interquartile range, 0.71–1.37).
FIG. 2. The concentration (310
6
per ml) of mononuclears (a) and dendritic cells (b,c) in the peritoneal fluid (PF) and peripheral blood (PB) of
patients with epithelial ovarian cancer (EOC) and serous cystadenoma.
254 WERTEL ET AL.
Cytometry Part B: Clinical Cytometry
The Percentage of DC in Patients with Different Histologic
Type of Ovarian Cancer
The percentages of PF and PB MDC and LDC in
women grouped by histologic type of ovarian cancer are
presented in Figure 4.
The percentage of MDC and LDC in patients with se-
rous carcinoma (sCa), undifferentiated carcinoma
(udCa), and mucinous cystadenocarcinoma (mCa) was
significantly higher (P<0.05) in the PF than in the PB.
The percentage of MDC and LDC in patients with clear
cell carcinoma (ccCa) did not differ significantly
between PF and PB. The MDC to LDC ratio in patients
with different histologic type of ovarian cancer did not
differ significantly between PF and PB.
The percentage of PB and PF MDCs and LDCs did not
differ significantly among patients with sCa, udCa, mCa,
and ccCa (Fig. 4).
The percentage of PF MDCs in patients with sCa,
udCa, mCa, and ccCa was significantly lower but that of
LDCs was significantly higher in comparison to patients
with serous cystadenoma (Fig. 4).
The MDC to LDC ratio in the PF of patients with sCa
(1.0; interquartile range, 0.45–2.46), udCa (1.02; inter-
quartile range, 0.46–1.56), mCa (1.32; interquartile
range, 0.63–2.03), and ccCa (1.32; interquartile range,
0.04–5.07) was significantly lower than in women with
benign ovarian tumors (71.2; interquartile range, 33.12–
129.41).
The percentage of MDC and LDC and the ratio of
MDCs to LDCs in the PB did not differ significantly
between women with different histologic types of ovar-
ian cancer and the reference group.
Correlation Between Concentration of Mononuclear Cells
and DC Subsets
There was a positive correlation between concentra-
tions of PF and also PB mononuclear cells and both MDC
and LDC in patients with malignant and benign ovarian
tumors. The statistical data are detailed in Table 2.
DISCUSSION
Most cases of ovarian malignancies are advanced at
the time of diagnosis. Therefore, ovarian cancer is one
of the leading causes of death among gynecologic malig-
nancies with a 5-year survival rate of about 15% (1). The
early clinical development of ovarian neoplasm is associ-
ated with nonrecognition of cancer by the immune sys-
tem. The fact that ovarian tumors, which differ so signifi-
cantly from the normal ovarian tissue, are not recog-
nized as foreign and not eliminated remains unclear. It
should be appreciated that the resistance of neoplastic
tissue can reflect either antigenic similarity of neoplastic
and normal tissue or aberrant immune response, includ-
ing nonrecognition of foreign cancer antigens by APCs
(3,4).
DC are the most potent antigen-presenting cells play-
ing a key role in the induction of protective immune
responses and maintenance of immunological memory.
Their exceptional ability to instruct naive T cells to initi-
ate immune responses is critically beneficial for the host
defense against neoplastic cells (6). Furthermore, a large
body of literature demonstrates a close relationship
between the presence of DCs within various malignant
tumors and prognosis (18–21). On the other hand, sev-
eral investigators have described the defective function
of DCs in human suffering from cancer, such as the lack
of expression of costimulatory molecules, consistent
with the phenotype of immature and nonactivated DCs
(16,22–24).
Ovarian cancer differs from others malignancies of
genital tract because of the spread throughout the peri-
toneal cavity. Serous ovarian neoplasms spread along the
peritoneum even in the case of relatively small tumors
of the ovary. It has been suggested that immune altera-
tion may be responsible for this phenomenon. There-
fore, our study was undertaken not only to investigate
the PB DCs but also to examine DCs in the local PF
immune system of women with ovarian cancer.
For identification of MDC and LDC subsets we chose
relatively novel markers: BDCA-1 and BDCA-2. With
regard to the presented, currently insurmountable diffi-
culties in obtaining normal PF, we selected the PF from
FIG. 3. The percentage of PF and PB MDC (a) and LDC (b)inthe
mononuclears of patients with epithelial ovarian cancer (EOC) and se-
rous cystadenoma (S.C.).
255DENDRITIC CELLS IN OVARIAN CANCER PATIENTS
Cytometry Part B: Clinical Cytometry
women with serous cystadenoma as a reference fluid
developing in the absence of neoplastic process.
Our results show that PF environment in women with
both malignant and nonmalignant ovarian tumors con-
tains considerably more DCs than PB. This observation
appears to support our previous hypothesis that the PB
DCs are specifically recruited into peritoneal cavity (14).
The reason for the accumulation of DCs in the PF has
not been clearly explained yet. It has been demonstrated
that many tumor types attract macrophages and DC
through the direct or indirect production of chemoat-
tractant factors (25,26). APC recruitment is expected to
contribute to the development of antitumoral immune
response, and such an effect has been demonstrated in a
number of animal studies. However, a number of studies
have demonstrated that the infiltration of tumors by APC
contributed to their aggressive phenotype by supplying
factors necessary for tumor cell proliferation and inva-
siveness, such as growth and angiogenic factors, as well
as proteolytic enzymes (25,26). Experimental studies
confirmed that MDCs, contrary to LDCs, are highly mi-
gratory cells with remarkable capacity for transmigrating
FIG. 4. The percentages of PB (a) and PF (b) MDC and LDC in women grouped by histologic type of ovarian cancer and in the reference group, se-
rous cystadenoma (S.C.).
Table 2
Correlation Between Concentration of Mononuclear Cells and Myeloid (M) and Lymphoid (L) DC Subsets
in the Peritioneal Fluid (PF) and Peripheral Blood (PB) of Patients
Patients Correlation between
RSpearman rank
correlation coefficient t(N-2) P
Ovarian cancer (n547) PF mononuclear cells and MDC 0.745 6.988 <0.00l
PF mononuclear cells and LDC 0.597 4.656 <0.001
PB mononuclear cells and MDC 0.668 5.607 <0.001
PB mononuclear cells and LDC 0.591 4.577 <0.001
Serous Cystadenoma (n537) PF mononuclear cells and MDC 0.701 5.648 <0.001
PF mononuclear cells and LDC 0.481 3.159 <0.05
PB mononuclear cells and MDC 0.762 5.893 <0.001
PB mononuclear cells and LDC 0.700 4.913 <0.001
256 WERTEL ET AL.
Cytometry Part B: Clinical Cytometry
across endothelium in response to chemotactic agents
without requiring endothelial support (27). But migra-
tion of DCs from blood is not the only explanation for
finding high concentration of DCs in PF. The other possi-
bilities may be: accumulation of DCs in the peritoneal
cavity by the breakdown of their journey to the lymph-
oid tissues, local generation from precursors, or nonphy-
siological proliferation of immature DCs.
The comparison of DC subsets between studied
groups demonstrate that the concentration and the per-
centage of PF MDCs in mononuclear cells was signifi-
cantly lower in patients with ovarian cancer in compari-
son to the group of nonmalignant ovarian tumors. These
results suggest that PF MDCs population may be affected
by the presence of the malignant disease and might con-
tribute to diminished acquired immune responses
observed in these women.
In contrast, the concentration and the percentage of PF
LDCs was significantly higher in patients with ovarian
cancer than in the reference group. This fact may be im-
portant for understanding of the mechanism of tumor
immune escape, because LDCs are expected to induce
tolerance rather than immunity. In the study by Zou et al.
(28) it was shown that LDCs infiltrating ovarian carci-
noma inhibited tumor-specific immunity by suppressing T-
cell activation. The investigation by Curiel et al. (15)
shows comparable results in tumor ascites of women
with ovarian carcinomas. They demonstrated that numer-
ous functional LDCs accumulate in tumor ascites and in-
hibit antitumor immunity. The same authors found that
LDCs produce high levels of the angiogenic cytokines
(TNF-aand IL-8) and induce potent neovascularization in
vivo. Thus, tumors may manipulate DCs distribution and
alter DCs function to support tumor angiogenesis.
In addition, we have calculated MDCs/LDCs ratio
which expresses the equilibrium between presentation
of antigens for induction of an effective immune
response or stimulation of T-cell tolerance (29,30). We
found that in the PF of patients with ovarian cancer the
BDCA-1/BDCA-2 DCs ratio was significantly lower in
comparison to patients with benign disease. These data
may suggest that LDCs subsets have influence on the
local immune response in the PF of patients with malig-
nant disease. Interestingly, these cells markedly outnum-
bered MDCs in the PF but not in the PB. PF LDCs may
actively suppress the Ag-specific T-cell response and thus
could be involved in immunosuppression. However the
precise role of PF LDCs still needs to be evaluated.
Some studies documented a significant dysfunction of
type 1 T-cell responses in tumor-bearing hosts, suggest-
ing that tumor progression might be associated with a
preferential type 2 T-cell response (31). However, factors
which influence the Th2 predominance in malignant tu-
mor patients still remain enigmatic. The available data
indicate that BDCA-1 and BDCA-2 DCs were claimed to
stimulate Th1 and Th2 types of immune responses,
respectively (6,11).
In this study we have demonstrated a significant accu-
mulation of immature lymphoid (BDCA-2 positive ) DCs
in the malignant PF in comparison with PF from women
with benign disease. Therefore, we concluded that PF
LDCs in patients with ovarian cancer may favor Th2 lym-
phocyte differentiation and/or the induction of immuno-
logical tolerance which is now considered as one of the
important mechanisms of tumor escape from immune
system control.
Further studies on the DCs’ function are necessary for
the complete understanding of the influence of tumor
microenvironment on DCs and the importance of this
fact for circumventing the immune system by cancer. It
should be stressed that PF as an area where both
immune and tumors cells coexist is an excellent clinical
model for studying local immune response. Detailed
investigation on the role of defective DCs in antitumor
immunity may provide new insights into the develop-
ment of novel therapeutic strategies for malignancies.
LITERATURE CITED
1. Guppy AE, Nathan PD, Rustin GJ. Epithelial ovarian cancer: A
review of current management. Clin Oncol (R Coll Radiol) 2005;
17:399–411.
2. Yancik R. Ovarian cancer. Age contrasts in incidence, histology, dis-
ease stage at diagnosis, and mortality. Cancer 1993;71:517–523.
3. Chen L, Ashe S, Brady WA, Hellstrom I, Hellstrom KE, Ledbetter JA,
McGowan P, Linsley PS. Costimulation of antitumor immunity by
the B7 counterreceptor for the T lymphocyte molecules CD28 and
CTLA-4. Cell 1992;71:1093–1102.
4. Zheng P, Sarma S, Guo Y, Liu Y. Two mechanisms for tumor evasion
of preexisting cytotoxic T-cell responses: Lessons from recurrent
tumors. Cancer Res 1999;59:3461–3467.
5. Avigan D. Dendritic cells: Development, function and potential use
for cancer immunotherapy. Blood Rev 1999;13:51–64.
6. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ,
Pulendran B, Palucka K. Immunobiology of dendritic cells. Annu
Rev Immunol 2000;18:767–811.
7. Di Nicola M, Lemoli RM. Dendritic cells: Specialized antigen pre-
senting cells. Haematologica 2000;85:202–207.
8. Hart DNJ. Dendritic cells: Unique leukocyte populations which con-
trol the primary immune response. Blood 1997;90:3245–3287.
9. Olweus J, BitMansour A, Warnke R, Thompson PA, Carballido J,
Picker LJ, Lund-Johansen F. Dendritic cell ontogeny: A human den-
dritic cell lineage of myeloid origin. Proc Natl Acad Sci USA
1997;94:12551–12556.
10. Robinson SP, Patterson S, English N, Davies D, Knight SC, Reid CDL.
Human peripheral blood contains two distinct lineages of dendritic
cells. Eur J Immunol 1999;29:2769–2778.
11. Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, Buck
DW, Schmitz J. BDCA-2, BDCA-3, and BDCA-4: Three markers for
distinct subsets of dendritic cells in human peripheral blood.
J Immunol 2000;165:6037–6046.
12. O’Doherty U, Peng M, Gezelter S, Swiggard WJ, Betjes M, Bhardwaj
N, Steinman RM. Human blood contains two subsets of dendritic
cells, one immunologically mature and the other immature. Immu-
nology 1994;82:487–493.
13. Grouard G, Rissoan MC, Filgueira L, Durand I, Banchereau J, Liu YJ.
The enigmatic plasmacytoid T cells develop into dendritic cells with
interleukin (IL)-3 and CD40-ligand. J Exp Med 1997;17:1101–1111.
14. Wertel I, Kotarski J, Rolinski J, Bojarska-Junak A, Gogacz M. Evalua-
tion of myeloid and lymphoid dendritic cells in peritoneal fluid in
women with non-malignant ovarian tumors. Am J Reprod Immunol
2003;50:238–242.
15. Curiel TJ, Cheng P, Mottram P, Alvarez X, Moons L, Evdemon-Hogan
M, Wei S, Zou L, Kryczek I, Hoyle G, Lackner A, Carmeliet P, Zou
W. Dendritic cells subsets differentially regulate angiogenesis in
human ovarian cancer. Cancer Res 2004;64:5535–5538.
16. Almand B, Resser JR, Lindman B, Nadaf S, Clark JI, Kwon ED, Car-
bone DP, Gabrilovich DI. Clinical significance of defective dendritic
cell differentiation in cancer. Clin Cancer Res 2000;6:1755–1766.
17. Yanagimoto H, Takai S, Satoi S, Toyokawa H, Takahashi K, Terakawa
N, Kwon AH, Kamiyama Y. Impaired function of circulating dendri-
tic cells in patients with pancreatic cancer. Clin Immunol 2005;114:
52–60.
257DENDRITIC CELLS IN OVARIAN CANCER PATIENTS
Cytometry Part B: Clinical Cytometry
18. Wischatta M, Sprinzl GM, Gunkel AR, Hussl B, Romani N, Schrott-Fi-
scher A. Dendritic cells in selected head and neck tumors. Ann
Otol Rhinol Laryngol 2000;109:56–62.
19. Zehntner S, Townsend W, Parkes J, Schmidt C, Down M, Bell J, Mul-
ligan R, O’Rourke M, Ellem K, Thomas R. Tumor metastasis biopsy
as a surrogate marker of response to melanoma immunotherapy. Pa-
thology 1999;31:116–122.
20. Bethwaite PB, Holloway LJ, Thornton A, Delahunt B. Infiltration by
immunocompetent cells in early stage invasive carcinoma of the
uterine cervix: A prognostic study. Pathology 1996;28:321–327.
21. Eisenthal A, Polyvkin N, Bramante-Schreiber L, Misonznik F, Hassner
A, Lifschitz-Mercer B. Expression of dendritic cells in ovarian
tumors correlates with clinical outcome in patients with ovarian
cancer. Hum Pathol 2001;32:803–807.
22. Nestle FO, Burg G, Fah J, Wrone-Smith T, Nickoloff BJ. Humansun-
light-induced basal-cell-carcinoma-associated dendritic cells are defi-
cient in T cell co-stimulatory molecules and are impaired as antigen-
presenting cells. Am J Pathol 1997;150:641–651.
23. Staveley-O’Carroll K, Sotomayor E, Montgomery J, Borrello I, Hwang
L, Fein S, Pardoll D, Levitsky H. Induction of antigen-specific T cell
anergy: An early event in the course of tumor progression. Proc
Natl Acad Sci USA 1998;95:1178–1183.
24. Gabrilovich DI, Corak J, Ciernik IF, Kavanaugh D, Carbone DP.
Decreased antigen presentation by dendritic cells in patients with
breast cancer. Clin Cancer Res 1997;3:483–490.
25. Coussens LM, Werb Z. Inflammation and cancer. Nature
2002;420:860–867.
26. Vicari AP, Caux C. Chemokines in cancer. Cytokine Growth Factor
Rev 2002;13:143–154.
27. De la Rosa G, Longo N, Rodriguez-Fernandez JL, Puig-Kroger A,
Pineda A, Corbi AL, Sanchez-Mateos P. Migration of human blood
dendritic cells across endothelial cell monolayers: Adhesion mole-
cules and chemokines involved in subset-specific transmigration.
J Leukoc Biol 2003;73:639–649.
28. Zou W, Machelon V, Coulomb-L’Hermin A, Borvak J, Nome F, Isaeva
T, Wei S, Krzysiek R, Durand-Gasselin I, Gordon A, Pustilnik T,
Curiel DT, Galanaud P, Capron F, Emilie D, Curiel TJ. Stromal-derived
factor-1 in human tumors recruits and alters the function of plasma-
cytoid precursor dendritic cells. Nat Med 2001;7:1339–1346.
29. Dhodapkar MV, Steinman RM, Krasovsky J, Munz C, Bhardwaj N.
Antigen-specific inhibition of effector T cell function in humans after
injection of immature dendritic cells. J Exp Med 2001;193:233–238.
30. Kuwana M. Induction of anergic and regulator y T cells by plasmacy-
toid dendritic cells and other dendritic cell subsets. Hum Immunol
2002;63:1156–1163.
31. Santin AD, Bellone S, Palmieri M, Bossini B, Cane S, Bignotti E,
Roman JJ, Cannon MJ, Pecorelli S. Restoration of tumor specific
human leukocyte antigens class I-restricted cytotoxicity by dendritic
cell stimulation of tumor infiltrating lymphocytes in patients with
advanced ovarian cancer. Int J Gynecol Cancer 2004;14:64–76.
258 WERTEL ET AL.
Cytometry Part B: Clinical Cytometry
... However, melanoma and gastric cancer patients have shown an increased average of pDCs in circulation [255,298]. On the other hand, an accumulation of pDCs in malignant ascites with depletion in the blood has been described in patients with ovarian cancer with partial restoration reported after achieving complete remission through chemotherapy [239,299,300]. This decrease in circulating pDCs has also been described among different hematological malignancies [301][302][303] and in breast cancer at later stages [214,288]. ...
Article
Full-text available
Dendritic cells (DCs) are a unique myeloid cell lineage that play a central role in the priming of the adaptive immune response. As such, they are an attractive target for immune oncology based therapeutic approaches. However, targeting these cells has proven challenging with many studies proving inconclusive or of no benefit in a clinical trial setting. In this review, we highlight the known and unknown about this rare but powerful immune cell. As technologies have expanded our understanding of the complexity of DC development, subsets and response features, we are now left to apply this knowledge to the design of new therapeutic strategies in cancer. We propose that utilization of these technologies through a multiomics approach will allow for an improved directed targeting of DCs in a clinical trial setting. In addition, the DC research community should consider a consensus on subset nomenclature to distinguish new subsets from functional or phenotypic changes in response to their environment.
... Interestingly, Frigola and co-workers demonstrated that activated DCs are the source of sPD-L1. What is interesting is that in our earlier study we showed accumulation of myeloid and plasmacytoid DCs in the peritoneal fluid of OC patients [60]. These findings indicate that sPD-L1 is released by both the immune system and cancer cells [61]. ...
Article
Full-text available
The latest literature demonstrates the predominant role of the programmed cell death axis (PD-1/PD-L1/PD-L2) in ovarian cancer (OC) pathogenesis. However, data concerning this issue is ambiguous. Our research aimed to evaluate the clinical importance of PD-L1/PD-L2 expression in OC environments. We evaluated the role of PD-L1/PD-L2 in OC patients (n = 53). The analysis was performed via flow cytometry on myeloid (mDCs) and plasmacytoid dendritic cells (pDCs) and monocytes/macrophages (MO/MA) in peripheral blood, peritoneal fluid (PF), and tumor tissue (TT). The data were correlated with clinicopathological characteristics and prognosis of OC patients. The concentration of soluble PD-L1 (sPD-L1) and PD-1 in the plasma and PF were determined by ELISA. We established an accumulation of PD-L1 + /PD-L2 + mDCs, pDCs, and MA in the tumor microenvironment. We showed an elevated level of sPD-L1 in the PF of OC patients in comparison to plasma and healthy subjects. sPD-L1 levels in PF showed a positive relationship with Ca125 concentration. Moreover, we established an association between higher sPD-L1 levels in PF and shorter survival of OC patients. An accumulation of PD-L1 + /PD-L2 + mDCs, pDCs, and MA in the TT and high sPD-L1 levels in PF could represent the hallmark of immune regulation in OC patients.
... In ovarian cancer, several studies have shown an accumulation of pDC in malignant ascites fluid, with decreased blood concentration [115,117,118]. Ascites-originating pDC were shown to favor tumor angiogenesis via the production of TNFα and IL-8 [118]. ...
Article
Prostate cancer (PCa) is the most common non-cutaneous cancer among men and the second leading cause of male cancer deaths in the United States. With no effective cure for advanced disease, the survival rates of castration-resistant disease and metastatic disease remains poor. Treatment via hormonal manipulation, immunotherapy, and chemotherapy remain marginally effective, indicating the need for novel treatment strategies. Cytoreductive prostatectomy (CRP) has grown as a treatment modality for metastatic castration resistant prostate cancer (mCRPC) and an emerging body of literature has demonstrated its survival benefits. In this review, we hope to further explore immunologic changes after CRP and the resultant effects on oncologic outcomes. Conclusively, the data and technical considerations of CRS evolve, CRS may continue to expand treat various type of metastatic cancer. Still, there are little reports about immunological changed after CRP. However, based on technical improvement, CRP and combinational immunotherapy are developing treatments of metastatic disease.
... PDCs constitute a rare, yet critically important and highly specialized immune cell subpopulation whose main role in immune surveillance is rapid recognition of foreign pathogens via selectively expressed toll-like receptors and the immediate activation of both innate and acquired immune systems ( Figure 1A) [247]. Incessant stimulation of PDCs by self-DNA (a situation when PDCs, which do not normally react to inert DNA of organism's cells, become continually activated by the altered DNA of transformed cells) is a characteristic of a variety of neoplasms, including ovarian carcinomas [248][249][250]. Accumulation of PDCs within the epithelial ovarian tumor bulk promotes vasculogenesis [251,252] and immune tolerance [253], and it is associated with unfavorable disease prognosis [254]. ...
Article
Full-text available
Metastatic dissemination of epithelial ovarian cancer (EOC) predominantly occurs through direct cell shedding from the primary tumor into the intra-abdominal cavity that is filled with malignant ascitic effusions. Facilitated by the fluid flow, cells distribute throughout the cavity, broadly seed and invade through peritoneal lining, and resume secondary tumor growth in abdominal and pelvic organs. At all steps of this unique metastatic process, cancer cells exist within a multidimensional tumor microenvironment consisting of intraperitoneally residing cancer-reprogramed fibroblasts, adipose, immune, mesenchymal stem, mesothelial, and vascular cells that exert miscellaneous bioactive molecules into malignant ascites and contribute to EOC progression and metastasis via distinct molecular mechanisms and epigenetic dysregulation. This review outlines basic epigenetic mechanisms, including DNA methylation, histone modifications, chromatin remodeling, and non-coding RNA regulators, and summarizes current knowledge on reciprocal interactions between each participant of the EOC cellular milieu and tumor cells in the context of aberrant epigenetic crosstalk. Promising research directions and potential therapeutic strategies that may encompass epigenetic tailoring as a component of complex EOC treatment are discussed.
... The peritoneal cavity of OC patients is infiltrated with a variety of leukocyte populations, and the immune cell composition within the tumour microenvironment impacts upon disease progression and has been reported to correlate with clinical outcome [14,15]. DC are present in the OC environment [16,17] and as professional antigen-presenting cells (APC) they are thought to be pivotal for the initiation of tumour-specific immune responses because of their ability to take up and process tumour antigens and prime cytotoxic T cell (CTL) responses. However, the ability of DC to launch a potent anti-tumour immune response is dependent on their direct activation via pattern recognition receptors (PRR) such as TLR [18]. ...
Article
Full-text available
Dendritic cells (DC) have the potential to instigate a tumour-specific immune response, but their ability to prime naïve lymphocytes depends on their activation status. Thus, for tumour immunotherapy to be effective, the provision of appropriate DC activation stimuli such as Toll-like receptor (TLR) agonists is crucial in order to overcome immunosuppression associated with the tumour microenvironment. To address this, we investigated how ovarian carcinoma (OC)-associated ascites impedes activation of DC by TLR agonists. Our results show that ascites reduces the TLR-mediated up-regulation of CD86 and partially inhibits the production of the pro-inflammatory cytokines interleukin 6 (IL-6), IL-12 and tumour necrosis factor α (TNFα) in monocyte-derived DC from healthy controls. We further observe an impaired T cell stimulatory capacity of DC upon activation with TLR agonists in the presence of ascites, indicating that their functionality is affected by the immunosuppressive factors. We identify IL-10 and prostaglandin E2 (PGE2) as the pivotal immunosuppressive components in OC-associated ascites compromising TLR-mediated DC activation. Interestingly, IL-10 is present in both ascites from patients with malignant OC and in peritoneal fluid from patients with benign ovarian conditions and both fluids have similar ability to reduce TLR-mediated DC activation. However, depletion of IL-10 from ascites revealed that the presence of paracrine IL-10 is not crucial for ascites-mediated suppression of DC activation in response to TLR activation. Unlike IL-10, PGE2 is absent from peritoneal fluid of patients with benign conditions and selectively reduces TNFα induction in response to TLR-mediated activation in the presence of OC-associated ascites. Our study highlights PGE2 as an immunosuppressive component of the malignant OC microenvironment rendering PGE2 a potentially important target for immunotherapy in OC.
Article
Full-text available
Ovarian cancer, especially high-grade serous type, is the most lethal gynecological malignancy. The lack of screening programs and the scarcity of symptomatology result in the late diagnosis in about 75% of affected women. Despite very demanding and aggressive surgical treatment, multiple-line chemotherapy regimens and both approved and clinically tested targeted therapies, the overall survival of patients is still unsatisfactory and disappointing. Research studies have recently brought some more understanding of the molecular diversity of the ovarian cancer, its unique intraperitoneal biology, the role of cancer stem cells, and the complexity of tumor microenvironment. There is a growing body of evidence that individualization of the treatment adjusted to the molecular and biochemical signature of the tumor as well as to the medical status of the patient should replace or supplement the foregoing therapy. In this review, we have proposed the principles of the novel regimen of the therapy that we called the “DEPHENCE” system, and we have extensively discussed the results of the studies focused on the ovarian cancer stem cells, other components of cancer metastatic niche, and, finally, clinical trials targeting these two environments. Through this, we have tried to present the evolving landscape of treatment options and put flesh on the experimental approach to attack the high-grade serous ovarian cancer multidirectionally, corresponding to the “DEPHENCE” system postulates.
Article
A polycystic ovarian syndrome (PCOS) is the most common endocrine disorder affecting females . Furthermore, it is a heterogeneous disease with a variety of etiologies and outcomes. Patients frequently complain about infertility, irregular menstruation, acne, seborrheic dermatitis, hirsutism, and obesity. PCOS can be caused by hypothalamic-pituitary-ovarian axis dysfunction, heredity, or metabolic abnormalities. PCOS is characterized by chronic low-level inflammation, which includes an imbalance in pro-inflammatory factor secretion, endothelial cell dysfunction, and leukocytosis. PCOS is also distinguished by hormonal and immune dysregulation. During PCOS, immune cells and immune regulatory molecules play critical roles in maintaining metabolic homeostasis and regulating immune responses. Because of oligo/anovulation, patients with PCOS have low progesterone levels. Therefore, low progesterone levels in PCOS overstimulate the immune system, causing it to produce more estrogen, which leads to a variety of autoantibodies. This review aims to summarize the immune regulation involved in the pathogenesis of PCOS and pave the way for the development of better PCOS treatment options in the near future.
Chapter
Despite the nondisputable progress in the treatment of ovarian cancer, it still brings a serious challenge to doctors and poor prognosis to the majority of patients. The most important processes, which shape cancer development, are neo-angiogenesis and chronic inflammation. Mediators engaged in their regulation have a deep impact on the ovarian cancer proliferation, peritoneal spread, and distant metastases. The regulatory network, which contributes to cancer escape from host immunosurveillance, is very complex and still needs to be fully uncovered. However, it’s so far understanding has brought some improvements in ovarian cancer therapy. One of them is the introduction of Bevacizumab into routine treatment, based on its clinical efficacy. It is hoped that information presented in this chapter will help to summarize the present knowledge about immunology of ovarian cancer and to fertilize the reader’s mind with new ideas.
Article
Full-text available
Checkpoint blockade immunotherapy (CBT) can induce long-term clinical benefits in patients with advanced cancer; however, response rates to CBT vary by cancer type. Cancers of the skin, lung, and kidney are largely responsive to CBT, while cancers of the pancreas, ovary, breast, and metastatic lesions to the liver respond poorly. The impact of tissue-resident immune cells on antitumor immunity is an emerging area of investigation. Recent evidence indicates that antitumor immune responses and efficacy of CBT depend on the tissue site of the tumor lesion. As myeloid cells are predominantly tissue-resident and can shape tumor-reactive T cell responses, it is conceivable that tissue-specific differences in their function underlie the tissue-site-dependent variability in CBT responses. Understanding the roles of tissue-specific myeloid cells in antitumor immunity can open new avenues for treatment design. In this review, we discuss the roles of tissue-specific antigen-presenting cells (APCs) in governing antitumor immune responses, with a particular focus on the contributions of tissue-specific dendritic cells. Using the framework of the Cancer-Immunity Cycle, we examine the contributions of tissue-specific APC in CBT-sensitive and CBT-resistant carcinomas, highlight how these cells can be therapeutically modulated, and identify gaps in knowledge that remain to be addressed.
Article
Background The treatment options for patients with peritoneal carcinomatosis (PC) of gastrointestinal and pancreaticobiliary origins are limited. The virus-like particle, CMP-001, composed of the Qβ bacteriophage capsid protein encapsulating a CpG-A oligodeoxynucleotide, activates plasmacytoid dendritic cells (pDCs) and triggers interferon alpha (IFNα) release, leading to a cascade of anti-tumor immune effects.Methods To evaluate the ability of CMP-001 to trigger an immune response in patients with PC, peritoneal cells were isolated and stimulated ex vivo with CMP-001. Both IFNα release and percentage of pDC were quantified using enzyme-linked immunosorbent assay (ELISA) and flow cytometry, respectively. To evaluate the anti-tumor response in vivo, murine PC models were generated using mouse cancer cell lines (Panc02 and MC38) in immunocompetent mice treated with intraperitoneal CMP-001 or saline control. Survival was followed, and the immunophenotype of cells in the peritoneal tumor microenvironment was evaluated.ResultsThe pDCs accounted for 1% (range 0.1–3.9%; n = 17) of the isolated peritoneal cells. Ex vivo CMP-001 stimulation of the peritoneal cells released an average of 0.77 ng/ml of IFNα (range, 0–4700 pg/ml; n = 14). The IFNα concentration was proportional to the percentage of pDCs present in the peritoneal cell mixture (r = 0.6; p = 0.037). In murine PC models, intraperitoneal CMP-001 treatment elicited an anti-tumor immune response including an increase in chemokines (RANTES and MIP-1β), pro-inflammatory cytokines (IFNγ, interleukin 6 [IL-6], and IL-12), and peritoneal/tumor immune infiltration (CD4+/CD8+ T and natural killer [NK] cells). The CMP-001 treatment improved survival in both the Panc02 (median, 35 vs 28 days) and the MC38 (median: 57 vs 35 days) PC models (p < 0.05).Conclusions As a novel immunotherapeutic agent, CMP-001 may be effective for treating patients with PC.
Article
Full-text available
A subset of CD4+CD11c−CD3− blood cells was recently shown to develop into dendritic cells when cultured with monocyte conditioned medium. Here, we demonstrate that CD4+ CD11c−CD3− cells, isolated from tonsils, correspond to the so-called plasmacytoid T cells, an obscure cell type that has long been observed by pathologists within secondary lymphoid tissues. They express CD45RA, but not markers specific for known lymphoid- or myeloid-derived cell types. They undergo rapid apoptosis in culture, unless rescued by IL-3. Further addition of CD40-ligand results in their differentiation into dendritic cells that express low levels of myeloid antigens CD13 and CD33.
Article
Full-text available
Article
Full-text available
Dendritic cells (DCs) are antigen-presenting cells with a unique ability to induce primary immune responses. DCs capture and transfer information from the outside world to the cells of the adaptive immune system. DCs are not only critical for the induction of primary immune responses, but may also be important for the induction of immunological tolerance, as well as for the regulation of the type of T cell–mediated immune response. Although our understanding of DC biology is still in its infancy, we are now beginning to use DC-based immunotherapy protocols to elicit immunity against cancer and infectious diseases.
Article
Despite the large number of potentially cytotoxic tumor-infiltrating (TIL) and tumor-associated (TAL) lymphocytes accumulated in the peritoneal cavity ascitic fluid and tumor tissue, advanced ovarian cancer is a progressive disease, suggesting that TIL and TAL populations eventually become functionally suppressed in vivo . Dendritic cells (DC) are the most powerful professional antigen presenting cells known in humans and recently, ovarian tumor antigen pulsed DC have been shown to elicit tumor specific human leukocyte antigens (HLA)-class I-restricted cytotoxicity from the peripheral blood of advanced ovarian cancer patients. In this study, we have evaluated the potential of tumor antigen-pulsed fully mature DC stimulation in restoring tumor-specific cytotoxicity in anergic TIL populations from advanced ovarian cancer patients. In addition, we have compared tumor-specific T-cell responses induced by tumor antigen-loaded DC in TIL to those induced in TAL and peripheral blood lymphocytes (PBL). DC stimulation induced powerful cytotoxicity against autologous tumor target cells in TIL-derived CD8+ T-cells from all patients tested, while autologous Epstein–Barr virus (EBV)-transformed lymphoblastoid cell lines (LCL) were not lysed. Killing of autologous tumor cells was higher by CD8+ T-cells from TIL compared to PBL and TAL ( P < 0.01) and was more strongly inhibited by anti-HLA class I MAb ( P < 0.05 compared to PBL and TAL). Phenotypically, all cytotoxic T lymphocyte (CTL) populations were CD3+/CD8+, with variable levels of CD56 expression. Finally, although a marked Type 1 cytokine bias [ie, interferon-gamma/interleukin-4 (IFN-γ high /IL-4 low )] was observable in all DC-stimulated CD8+ T-cell populations, TIL derived CD8+ T-cells showed a higher percentage of IFN-γ positive cells compared to TAL and PBL. Taken together, these data show that tumor lysate-pulsed DC can consistently restore strong CD8+ CTL responses from TIL against autologous ovarian cancer cells. DC-stimulated TIL may represent a superior source of tumor-specific CTL for adoptive T-cell immunotherapy for advanced ovarian cancer.
Article
Background. Age comparisons for incidence, histology, disease stage at initial diagnosis, and mortality of more than 20,000 ovarian cancer patients diagnosed between 1973–1987 are the focus of this descriptive epidemiologic study. This paper highlights key issues and concerns regarding ovarian cancer in women 65 years and older as a frame of reference for the proceedings of the working conference, “Perspectives on Ovarian Cancer in Older-Aged Women,” co-sponsored by the National Institute on Aging, National Cancer Institute, and American Cancer Society held at the National Institutes of Health, November, 1991. Methods. Data are from the National Cancer Institute's Surveillance, Epidemiology, and End Results (SEER) Program and the National Center for Health Statistics. The SEER Program database represents approximately 9.6% of the U.S. population. Results. Ovarian cancer affects women in the age group 65 years and older more frequently than younger women. More than 48% of all ovarian cancers occur in women in this age group. Age-adjusted rates increase as age advances, peaking at 54.0 per 100,000 in the age group 75–79 years. Time trends also indicate increases in age-specific incidence rates. This malignancy takes its toll in mortality in women 65 years and older with 64% of all deaths due to this neoplasm (in 1989). Moreover, older women are more likely to be initially diagnosed with advanced disease. Conclusions. Important questions about ovarian cancer in older-aged women need urgent attention from the research community. New strategies for diagnostic leads have to be developed for older women.
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
Two subsets of dendritic cells, differing in T-cell stimulatory function, have been purified directly from human blood. Both subsets are positive for major histocompatibility complex (MHC) class II expression and negative for lineage-specific antigens (e.g. CD3, CD14, CD16, CD19 negative), but are separated by exploiting differences in expression of the beta 2-integrin, CD11c. The CD11c-negative subset is functionally immature, requiring monocyte-derived cytokines to develop into typical dendritic cells. The CD11c-positive subset has potent T-cell stimulating activity and expresses the activation antigen CD45RO, unlike its immature counterpart. However, these mature cells only develop typical dendritic morphology and high levels of MHC proteins and adhesins after a period of culture independent of exogenous cytokines. Although the freshly isolated mature dendritic cells resemble monocytes in cytospin preparations, the former lack CD14 and have a much stronger primary T-cell stimulatory capacity. We hypothesize that the CD11c-negative immature cells are marrow-derived precursors to tissue dendritic cells, such as epidermal Langerhans' cells, while the CD11c-positive cells are derived from tissues where they have been activated by antigen, and are en route to the spleen or lymph nodes to stimulate T-cell responses there.
Article
In various tumor types dentritic cell, infiltration and the presence of tumor-infiltrating lymphocytes have been associated with an improved clinical outcome. In the uterine cervix these immunocompetent cells have been associated with improved prognosis in high stage disease. The current study examines the significance of stromal and tumor T-lymphocyte infiltration together with S-100 positive dendritic cell infiltration in a series of 73 women with low stage (FIGO 1b) invasive squamous and adenosquamous cervical carcinoma. Thirty four percent of cases contained S-100 positive dendritic cells. These were under-represented in cases showing pelvic recurrence or distant disease (1 of 11 compared to 24 of 62 free of recurrence, P = 0.05) and over-represented in cases showing lymphatic/capillary space involvement (12 of 23 compared to 13 of 46 without vascular space invasion, P = 0.05). The women were followed up for an average of 5.2 years and the five-year survival for women whose tumors contained S-100 positive dendritic cells was 92% compared to 73% for negative cases (P = 0.04). There was a significant association between a low density of tumor infiltrating T-cells and risk of pelvic lymph node spread and subsequent local or distant disease control failure (P = 0.008). A five year survival advantage was seen with five or more CD 3 positive tumor infiltrating T-lymphocytes per high power field (90%) compared to a lower count (68%) (P = 0.04). A similar advantage could not be demonstrated for a high stromal infiltrate of T-cells. As yet neither the specific mechanisms that induce these cells to infiltrate some cervical carcinomas nor the nature of the immunological injury that the cells co-ordinate in tumor tissue are well understood.
Article
Immune surveillance of skin cancer involves the stimulation of effector T cells by tumor-derived antigens and antigen-presenting cells (APCs). An effective APC must not only display processed antigen in the context of MHC molecules but also express co-stimulatory molecules that are required to fully activate T cells. One of the most common cutaneous neoplasms is basal cell carcinoma. To investigate expression of the co-stimulatory molecules CD80 (B7-1) and CD86 (B7-2) on tumor-associated dendritic cells (TADCs), cryosections from basal cell carcinomas were immunostained. In basal cell carcinomas, only 1 to 2% of intratumor and 5 to 10% of peritumor APCs expressed CD80 or CD86. In contrast, biopsies of immunological/inflammatory dermatoses revealed that 38 to 73% of APCs expressed CD80 and CD86. To further evaluate their phenotype and function, TADCs were isolated from tissue samples of basal cell carcinomas; they were non-adherent to plastic, displayed a typical dendritic morphology, and expressed high levels of major histocompatibility class II molecules on their surface. When TADCs were compared with dendritic cells from blood for presentation of superantigens (staphylococcal enterotoxins A and B) to resting autologous T cells, TADCs were consistently weaker stimulators of T cell proliferation than blood dendritic cells. When analyzed by flow cytometry, TADCs expressed high levels of HLA-DR, but only 5 to 10% co-expressed CD80 or CD86. A 3-day culture in granulocyte/macrophage colony-stimulating factor-containing medium partially reconstituted the TADC expression of CD80 and CD86 as well as their immunostimulatory capacity. Thus, in this common skin cancer, although there are prominent collections of HLA-DR-positive APCs in and around tumor cells, the TADCs are deficient in important co-stimulatory molecules as well as being weak stimulators of T cell proliferation. The paucity of co-stimulatory molecule expression and functional activity of TADCs may explain why the local T lymphocytic infiltrate fails to become fully activated to eradicate adjacent tumor cells. From a clinical perspective, these findings suggest a novel immunotherapeutic strategy targeting T cell co-stimulatory molecules on professional APCs in cutaneous oncology.