ArticlePDF Available

Tumoricidal activity of TLR7/8-activated inflammatory dendritic cells

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

Abstract and Figures

Imiquimod (IMQ), a synthetic agonist to Toll-like receptor (TLR) 7, is being successfully used for the treatment of certain skin neoplasms, but the exact mechanisms by which this compound induces tumor regression are not yet understood. While treating basal cell carcinoma (BCC) patients with topical IMQ, we detected, by immunohistochemistry, sizable numbers of both myeloid dendritic cells (mDCs) and plasmacytoid DCs (pDCs) within the inflammatory infiltrate. Surprisingly, peritumoral mDCs stained positive for perforin and granzyme B, whereas infiltrating pDCs expressed tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). The biological relevance of this observation can be deduced from our further findings that peripheral blood-derived CD11c(+) mDCs acquired antiperforin and anti-granzyme B reactivity upon TLR7/8 stimulation and could use these molecules to effectively lyse major histocompatibility complex (MHC) class I(lo) cancer cell lines. The same activation protocol led pDCs to kill MHC class I-bearing Jurkat cells in a TRAIL-dependent fashion. While suggesting that mDCs and pDCs are directly involved in the IMQ-induced destruction of BCC lesions, our data also add a new facet to the functional spectrum of DCs, ascribing to them a major role not only in the initiation but also in the effector phase of the immune response.
Peripheral blood–derived DCs express lytic molecules after TLR7/8 stimulation. (A) Characterization of isolated peripheral blood–derived pDCs and mDCs as HLA-DR⁺BDCA-2⁺ and HLA-DR⁺CD11c⁺, respectively, and lineage-negative cell populations by flow cytometry. (B and C) Analysis of intracellular expression levels of perforin and granzyme B of isolated mDCs before and after culture with TLR7/8 agonists by flow cytometry. (B) Representative dot plots of mDCs show double-positive cells for perforin (FITC) and granzyme B (PE) after stimulation with TLR7, TLR8, and TLR7/8 agonists. (C) Quantitative analysis of granzyme B⁺ and perforin⁺ mDCs. Data are given as mean percentages of perforin- or granzyme B–positive mDCs ± SEM from three independent experiments. (D) Freshly isolated and TLR7/8-stimulated mDCs do not display membrane-bound TRAIL (A488). Representative histogram plots from three independent experiments show the anti-TRAIL reactivity (bold line) and the matching isotype control (dashed line). (E and F) Determination of intracellular expression levels of perforin and granzyme B on isolated pDCs before and after stimulation with TLR agonists. (E) Representative dot plots of pDCs show a decrease of granzyme B–positive cells after stimulation with TLR7 and TLR7/8 agonists. (F) Quantitative analysis of pDCs expressing perforin and granzyme B. Data are given as mean percentages of perforin- and granzyme B–positive cells ± SEM from three independent experiments. (G) TLR7/8-stimulated, but not freshly isolated, pDCs display membrane-bound TRAIL (A488). Representative histogram plots from three different experiments show the anti-TRAIL reactivity (bold line) and the matching isotype control (dashed line). Dead cells were excluded by gating out propidium iodide–positive cells in all experiments.
… 
This content is subject to copyright.
The Journal of Experimental Medicine
ARTICLE
JEM © The Rockefeller University Press $15.00
Vol. 204, No. 6, June 11, 2007 1441–1451 www.jem.org/cgi/doi/10.1084/jem.20070021
1441
Interest in imiquimod (IMQ)  rst came from the
observation that this imidazoquinoline exerts a
profound activity against viral acanthomas that
was originally explained by its IFN- inducing
capacity (1). When used topically for a prolonged
period of time, it can lead to the regression of
certain virus-induced (e.g., genital warts [2] and
molluscum contagiosum [3]) and other (e.g.,
basal cell carcinoma [BCC] [4, 5] and lentigo
maligna [6]) skin neoplasms. IMQ exerts its
biologic activity primarily by ligation of Toll-
like receptor (TLR) 7 (7) and, to a lesser extent,
TLR8, both of which have been identi ed as
natural receptors for single-stranded RNA
(8, 9). Cell stimulation via TLR7 and TLR8 leads
to downstream activation of NF-B and other
transcription factors (10, 11). Consequently,
several genes encoding mediators and e ector
molecules of the innate as well as the adap-
tive immune response are transcribed (12–14).
Because of their prominent expression of TLR7
and TLR8, plasmacytoid DCs (pDCs) and my-
eloid DCs (mDCs) (15, 16), respectively, are
therefore likely candidates for the initiation of
the IMQ-induced host defense reaction. Other
mechanisms explaining the antitumor activity
of IMQ are also under discussion. These include
(a) the reversal of CD4
regulatory T cell function
(17), (b) a TLR-independent immunostimu-
latory action of IMQ via adenosine receptor
signaling (18), (c) direct (19) and indirect, via
IFN- (20), IMQ-induced proapoptotic e ects
on tumor cells, and (d) an antiangiogenic ac-
tivity of IMQ, as shown in a mouse model of
angiogenesis (21, 22).
In a recent study, our group investigated
IMQ-induced tumor regression in mice and
found not only a good clinical response of the
tumors to the topically applied compound but
also a direct correlation between IMQ-induced
tumor regression and the density of DCs in the
peritumoral tissue (23). Not infrequently, cancer
cells in death were found in close contact with
DCs, which was compatible with a tumoricidal
property of the latter (24, 25). In this study, we
sought to determine whether similar phenome-
nona also occur during IMQ treatment of human
skin cancers and, if so, to unravel the mechanisms
responsible for IMQ-induced tumor regression.
Tumoricidal activity of TLR7/8-activated
in ammatory dendritic cells
Georg Stary,
1
Christine Bangert,
1
Martina Tauber,
2
Robert Strohal,
3
Tamara Kopp,
1
and Georg Stingl
1
1
Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna,
1090 Vienna, Austria
2
Department of Pathology and
3
Department of Dermatology, Federal Academic Hospital Feldkirch, 6800 Feldkirch, Austria
Imiquimod (IMQ), a synthetic agonist to Toll-like receptor (TLR) 7, is being successfully used
for the treatment of certain skin neoplasms, but the exact mechanisms by which this com-
pound induces tumor regression are not yet understood. While treating basal cell carcinoma
(BCC) patients with topical IMQ, we detected, by immunohistochemistry, sizable numbers of
both myeloid dendritic cells (mDCs) and plasmacytoid DCs (pDCs) within the in ammatory
in ltrate. Surprisingly, peritumoral mDCs stained positive for perforin and granzyme B,
whereas in ltrating pDCs expressed tumor necrosis factor–related apoptosis-inducing ligand
(TRAIL). The biological relevance of this observation can be deduced from our further  nd-
ings that peripheral blood–derived CD11c
mDCs acquired antiperforin and anti–granzyme
B reactivity upon TLR7/8 stimulation and could use these molecules to effectively lyse major
histocompatibility complex (MHC) class I
lo
cancer cell lines. The same activation protocol led
pDCs to kill MHC class I–bearing Jurkat cells in a TRAIL-dependent fashion. While suggesting
that mDCs and pDCs are directly involved in the IMQ-induced destruction of BCC lesions,
our data also add a new facet to the functional spectrum of DCs, ascribing to them a major
role not only in the initiation but also in the effector phase of the immune response.
CORRESPONDENCE
Georg Stingl:
georg.stingl@meduniwien.ac.at
Abbreviations used: APC, allo-
phycocyanin; BCC, basal cell
carcinoma; IMQ, imiquimod;
iNOS, inducible NO synthase;
mDC, myeloid DC; pDC,
plasmacytoid DC; TLR, Toll-
like receptor; TRAIL, TNF-
related apoptosis-inducing
ligand; TUNEL, Tdt-mediated
dUTP-biotin nick-end labeling.
T. Kopp and G. Stingl contributed equally to this study.
The online version of this article contains supplemental material.
1442 TUMORICIDAL FUNCTION OF ACTIVATED DENDRITIC CELLS | Stary et al.
RESULTS
Regression of BCC upon IMQ treatment
Seven patients with histopathologically con rmed super cial
BCC were treated with IMQ. After a treatment period of
6 wk, we observed a complete clinical (Fig. 1) and histopatho-
logical response in all patients. No signs of recurrence were
noted in any of the patients followed for at least 10 mo.
In accordance with a previous report (4), all patients de-
veloped, before tumor clearance, an in ammatory tissue re-
action at the site of IMQ application. It began as erythema
after 2–3 d of treatment; became erosive during the second
week (Fig. 1); appeared as crusting and, later, scaling plaques
after 3–4 wk; and resolved completely after cessation of IMQ
treatment. Systemic side e ects such as  u-like symptoms,
lymphadenopathy, myalgia, or changes in laboratory values
never occurred in any of our patients.
Emergence kinetics of leukocytic populations
in the peritumoral in ltrate upon IMQ treatment
Biopsies from BCCs were obtained before, during, and after
IMQ treatment and subjected to immuno uorescence analy-
sis using a broad panel of antibodies (Table I) to analyze, both
quantitatively and qualitatively, the composition and kinet-
ics of the IMQ-induced in ammatory in ltrate. In untreated
BCCs we found a sparse in ltrate, mainly consisting of T cells
of the helper phenotype (Table II). Upon 2 wk of IMQ treat-
ment, a dramatic increase of CD8
T cells and, to a much
lesser extent, of CD4
T cells was seen around tumor cell is-
lets (Table II). Fig. 2 (A and B) shows that, after 2 wk of topi-
cal IMQ treatment, BCC islets were surrounded and partly
in ltrated by dendritically shaped cells exhibiting either the
CD11c
/HLA-DR
mDC (Fig. 2 A) or the CD123
/HLA-
DR
pDC (Fig. 2 B) phenotype. This increase in in am-
matory-type mDCs and pDCs occurs at the expense of the
resident DC populations of normal human skin (Langerhans
cells and CD1c
dermal DCs; Table II). Other constituents
of the peritumoral in ltrate included (a) CD56
/CD94
NK
cells, (b) CD14
mononuclear phagocytes, and (c) CD15
/
HLA-DR
neutrophils (Table II). In contrast, B cell, eosinophil,
basophil, and mast cell counts displayed no major alterations
upon IMQ treatment. At the end of treatment, when tumor
clearance was achieved, the density of the various in ltrating
leukocyte subpopulations was comparable to the values before
IMQ application (Table II).
Detection of lytic molecules in the peritumoral tissue
upon IMQ treatment
Analyzing the mechanisms of IMQ-induced tumor regres-
sion, we searched, by Tdt-mediated dUTP-biotin nick-end
labeling (TUNEL) staining, for signs of apoptosis in IMQ-
treated BCC lesions. Although only a few TUNEL
tumor
cells were detected in untreated BCCs (Fig. 2 C), the decrease
Figure 1. Complete regression of super cial BCC after a 6-wk treat-
ment period with IMQ. IMQ topically applied  ve times a week for a period
of 6 wk led to a local in ammatory response, which resulted in a complete
clinical and histopathological tumor clearance in all patients treated. The
clinical pictures are representative for all patients (n 7) treated with IMQ.
Table I. List of mAbs used in this study
Antibody speci city Clone (source of antibodies) Isotype
Staining experiments
BDCA-2 (FITC) AC144 (Miltenyi Biotec) mIgG1
CD1a (FITC) HI149 (BD Biosciences) mIgG1
CD1c (pur., biot.) M241 (Ancell) mIgG1
CD3 (FITC, PerCP) SK-7 (BD Biosciences) mIgG1
CD4 (pur.) SK-3 (BD Biosciences) mIgG1
CD8 (pur.) C8/144B (DakoCytomation) mIgG1
CD8 (allophycocyanin [APC]) B9.11 (Immunotech Coulter) mIgG1
CD11c (FITC) BU15 (Serotec) mIgG1
CD11c (PE, PE-Cy5) B-ly6 (BD Biosciences) mIgG1
CD14 (pur., FITC, PerCP) MP9 (BD Biosciences) mIgG2b
CD15 (FITC) 80H5 (Immunotech Coulter) mIgM
CD19 (pur., PerCP) SJ25C1 (BD Biosciences) mIgG1
CD45RA (pur.) L48 (BD Biosciences) mIgG1
CD56 (biot., PE-Cy5) B159 (BD Biosciences) mIgG1
CD83 (FITC) H15a (Immunotech Coulter) mIgG2b
CD94 (pur.) HP-3D9 (BD Biosciences) mIgG1
CD117 (c-kit, pur.) 95C3 (An Der Grub) mIgG1
CD123 (biot.) 9F5 (BD Biosciences) mIgG1
CD203c (PE) 97A6 (Immunotech Coulter) mIgG1
CD207 (pur.) DCGM4 (Immunotech Coulter) mIgG1
Fas ligand (pur.) NOK-1 (BD Biosciences) mIgG1
Granzyme B (pur.) GrB-7 (Monosan) mIgG2a
Granzyme B (PE) GB-11 (PeliCluster) mIgG1
HLA-DR (pur., FITC, APC) L243 (BD Biosciences) mIgG2a
IFN- 225.2C (Chromaprobe) mIgG2b
iNOS (FITC) 6 (BD Biosciences) mIgG2a
MBP (pur.) BMK13 (Chemicon) mIgG1
Pancytokeratin (pur.) AE1 and AE3 (BioGenex) mIgG1
Perforin (pur.) dG9 (Abcam) mIgG2b
Perforin (FITC) G9 (BD Biosciences) mIgG2b
TNF- (FITC) MAb11 (BD Biosciences) mIgG1
TRAIL (pur.) 75402 (R&D Systems) mIgG1
TRAIL (neutralizing; pur.) 75411 (R&D Systems) mIgG1
TRAIL R1 (pur.) 69036 (R&D Systems) mIgG1
Cell-sorting experiments
CD3 (pur.) UCHT1 (Immunotech Coulter) mIgG1
CD16 (pur.) 3G8 (Immunotech Coulter) mIgG1
CD19 (pur.) J4.119 (Immunotech Coulter) mIgG1
CD34 (pur.) 581 (Immunotech Coulter) mIgG1
CD41 (pur.) SZ22 (Immunotech Coulter) mIgG1
CD56 (pur.) C218 (Immunotech Coulter) mIgG1
CD235a (pur.) 11E4B-7-6 (Immunotech Coulter) mIgG1
biot., biotinylated; MBP, eosinophil major basic protein; pur., puri ed.
JEM VOL. 204, June 11, 2007 1443
ARTICLE
in pancytokeratin
BCC cells after 2 wk of therapy was ac-
companied by a concomitant increase of TUNEL
tumor
cells (Fig. 2 D). One may therefore conclude that BCC re-
gression upon IMQ treatment is at least partly accomplished
by apoptosis of tumor cells.
Based on the observation of IFN-–induced tumor re-
gression (26) and tumor cell apoptosis (27), we searched for
the presence of IFN-–producing cells in IMQ-treated BCC
biopsies and found 30% of all pDCs, but no other leuko-
cytes, to exhibit anti–IFN- staining (Fig. 2 E). It is therefore
not unreasonable to assume that IMQ-activated pDCs con-
tribute to BCC apoptosis by IFN- production.
In addition to IFN-–induced apoptotic events, perforin/
granzyme-based and death receptor ligand–mediated cancer
cell lysis are the immune system’s major tumoricidal e ector
mechanisms. When we analyzed BCC biopsies before, during,
and after IMQ treatment for the expression of lytic mole-
cules, we could hardly detect any expression of Fas ligand
(Fig. 2 F). In contrast, we observed a considerable up-regulation
of perforin, granzyme B, and TNF-related apoptosis-inducing
ligand (TRAIL) on in ltrating cells after 2 wk of IMQ treat-
ment when compared with the low baseline levels of un-
treated BCCs (Fig. 2 F).
The majority of anti-TRAIL reactivity was observed on
T cells (Fig. 3 A), but CD11c
mDCs, CD123
pDCs, and
CD14
mononuclear phagocytes also expressed this molecule
(Fig. 3, A and B). The biologic importance of this  nding is
underscored by the further observation that tumor cells of
untreated (Fig. 3 C) and IMQ-treated (Fig. 3 D) BCC lesions
were positive for TRAIL receptor 1 (TRAIL R1) and sur-
rounded by a dense rim of CD123
cells (Fig. 3 D). Anti-
TRAIL R2, R3, and R4 stainings yielded negative results
(not depicted).
Somewhat to our surprise, we found that only a few T cells
and NK cells displayed antiperforin and anti–granzyme B
staining and that CD3
CD56
HLA-DR
cells were the main
source of these lytic molecules (Fig. 4, A and C). Additional
phenotyping revealed that these cells were CD11c
(Fig.
4, B and D) and lacked lineage markers such as CD3, CD56,
CD14, CD15, or CD19, as well as CD1a and CD207, thus
corresponding to mDCs. Similar to what has been observed
in untreated psoriatic skin lesions (28), CD11c
/HLA-DR
DCs in IMQ-treated BCC lesions were also found to re-
act with antibodies against inducible NO synthase (iNOS)
and TNF- (Fig. 4 E). At least phenotypically, these cells
therefore corresponded to TNF-– and iNOS- producing
DCs, an mDC population possibly involved in innate immune
defense against bacteria (29). In contrast to in ltrating pDCs
(Fig. S1 A, available at http://www.jem.org/cgi/content/full/
jem.20070021/DC1), the vast majority of CD11c
/HLA-
DR
DCs displayed the maturation marker CD83 (Fig. S1 B).
As it was quite unexpected not to  nd cytotoxic T cells
Table II. Number of leukocytes occurring in BCCs before, after 2 wk, and at the end of IMQ treatment
Cell type Antigens Epidermis/dermis Untreated BCCs 2 wk IMQ 6 wk IMQ
T helper cells
CD4
/CD3
epidermis (cells/mm)
dermis (cells/mm
2
)
1.8 2
98.2 64.4
3.3 3.3
164 60.6
1.4 1.8
99 56.8
Cytotoxic T cells
CD8
/CD3
epidermis (cells/mm)
dermis (cells/mm
2
)
1.4 0.9
55.7 31.1
5.5 2.6
236.6 61.4
3.73.4
63.135.3
NK cells
CD56
/CD94
epidermis (cells/mm)
dermis (cells/mm
2
)
0
3 2.2
0.7 1.3
46.8 19
0
10.1 10.7
Langerhans cells
CD207
/CD1a
epidermis (cells/mm)
dermis (cells/mm
2
)
9 4.2
6.2 3.6
3.6 1.7
6.7 4.9
8.3 5.2
5.1 4.7
CD11c
mDCs CD11c
/HLA-DR
epidermis (cells/mm)
dermis (cells/mm
2
)
0
25.5 4.2
0
58.4 14.6
0
22.9 4.8
CD1c
mDCs CD1c
/HLA-DR
epidermis (cells/mm)
dermis (cells/mm
2
)
0.7 1.3
55.2 29.7
0.3 0.5
16.3 7.4
0.3 0.4
24.1 12.7
Phagocytes
CD14
epidermis (cells/mm)
dermis (cells/mm
2
)
1.4 3.8
21.2 15.2
2.4 3.6
47.6 6.1
1.3 3
35.1 12.7
pDCs
CD123
/ CD45RA
epidermis (cells/mm)
dermis (cells/mm
2
)
0.2 0.4
9.9 12.1
0
54.7 6.5
0
17.1 8.8
Neutrophils
CD15
/HLA-DR
epidermis (cells/mm)
dermis (cells/mm
2
)
0
7.8 8.9
0
62.1 14.3
0
15.4 13
Eosinophils
MBP
epidermis (cells/mm)
dermis (cells/mm
2
)
0
0
0
2.1 3.3
0
0.5 0.9
Basophils
CD203c
epidermis (cells/mm)
dermis (cells/mm
2
)
0
0
0
0
0
0
Mast cells
cKit
epidermis (cells/mm)
dermis (cells/mm
2
)
0
14.3 13.5
0
13.3 2.9
0
10.4 11.7
B cells
CD19
epidermis (cells/mm)
dermis (cells/mm
2
)
0
3.2 4.6
0
2.8 5.6
0
1.1 1.5
Cells were visualized using the indicated markers and were evaluated by immuno uorescence analysis. Numbers in bold indicate means SE of the mean of cells counted in
BCC lesions before, after 2 wk, and after 6 wk of IMQ treatment. MBP, eosinophil major basic protein.
1444 TUMORICIDAL FUNCTION OF ACTIVATED DENDRITIC CELLS | Stary et al.
and/or NK cells, but instead to find DCs containing per-
forin and granzyme B, we compared immuno uorescence
triple stainings of IMQ-treated BCCs with those of allergic
contact dermatitis, a disease in which perforin- and gran-
zyme B–containing T cells have been previously described
(30). In contrast to IMQ-treated BCC lesions in which most
perforin- and granzyme B–positive cells were CD3
as
well as CD8
, antiperforin and anti–granzyme B reactivity
in allergic contact dermatitis was mainly restricted to T cells
(Fig. S1, C–F).
Detection of perforin and granzyme B in mDCs
upon TLR7 and TLR8 stimulation and of TRAIL
on pDCs upon TLR7 stimulation
To determine the biological signi cance of anti-TRAIL and
antiperforin/anti–granzyme B staining of DCs in IMQ-
treated BCC lesions, we decided, for quantitative reasons, to
work with DCs isolated from peripheral blood rather than
from these skin lesions. Using the protocol described in
Materials and methods, we routinely obtained mDC and
pDC populations of 93–99% (mean 97%) purity. By FACS
analysis, these cells were HLA-DR
, expressed either high
levels of CD11c or BDCA-2, and lacked the lineage markers
CD3, CD56, CD14, and CD19 (Fig. 5 A). None of the lytic
molecules searched for were detected on freshly isolated
mDCs. In contrast, stimulation of puri ed peripheral blood–
derived mDCs with TLR7 and TLR8 ligands, but not with
the vehicle control, led to the intracellular expression of per-
forin and granzyme B in 10–15% of these cells (Fig. 5, B
and C). Expression of TRAIL was not detectable on mDCs
after stimulation with TLR7/8 ligands (Fig. 5 D).
Unstimulated pDCs abundantly contained granzyme B
within their cytoplasm (Fig. 5 E). After 12 h of stimulation
with ligands to TLR7 and TLR7/8, but not with the vehicle
control or the TLR8 ligand alone, the intracellular expres-
sion of granzyme B in pDCs had decreased. (Fig. 5, E and F).
This is consistent with the previous  nding of pDCs express-
ing TLR7 but not TLR8 (15). In contrast to mDCs, TLR7/8
Figure 2. Induction of tumor cell apoptosis is accompanied by
peritumoral accumulation of DCs and up-regulation of lytic
molecules on in ammatory cells. (A and B) Immuno uorescence triple
labeling of cryostat sections of IMQ-treated BCC lesions with antipan-
cytokeratin (TRITC), anti–HLA-DR (FITC), and either (A) anti-CD11c (APC) or
(B) anti-CD123 (Cy5) revealed a close proximity of both DC populations
with tumor cells. Arrows indicate double-positive DCs. (C) Untreated
BCCs and (D) BCCs after 2 wk of IMQ treatment were stained with an
antipancytokeratin antibody (TRITC) and TUNEL staining (FITC). Note
the double-positive tumor cells occurring after 2 wk of IMQ treatment
(arrows). The pictures are representative for all evaluated biopsies. The
dotted lines point out the margins of BCCs. (E) Identi cation of IFN-
producing pDCs in 2-wk–treated BCCs by triple stainings with anti–
IFN- (TRITC), anti–HLA-DR (FITC), and anti-CD123 (Cy5) mAbs. The
arrow indicates a triple-positive cell. (F) Quantitative analysis of lytic
molecules expressed by in ammatory cells of the peritumoral in ltrate.
Immuno uorescence single stainings were performed using antiperforin,
anti–granzyme B, anti-Fas ligand, and anti-TRAIL antibodies (all TRITC).
Data are given as absolute numbers of positive cells SEM for the indi-
cated markers.
Figure 3. Upon IMQ treatment, TRAIL R1
BCC islets are sur-
rounded by TRAIL-expressing pDCs. (A) Detailed quantitative in situ
analysis of the expression pattern of TRAIL on leukocytes before, during,
and after IMQ treatment was performed by immuno uorescence double
stainings with an anti-TRAIL mAb and lineage markers in all patients
treated. The data are given as absolute numbers of double-positive
cells SEM. (B) A representative view of anti-CD123 (FITC), anti–HLA-DR
(APC), and anti-TRAIL (TRITC) triple stainings of a BCC lesion after 2 wk of
IMQ treatment identi es pDCs as TRAIL-expressing cells. Immuno uores-
cence triple labeling of (C) untreated and (D) IMQ-treated BCCs with
antipancytokeratin (TRITC), anti-TRAIL R1 (A488), and anti-CD123
(Cy5) reveals TRAIL R1
BCC cells (C and D) surrounded by CD123
cells (arrows; D).
JEM VOL. 204, June 11, 2007 1445
ARTICLE
agonists induced up-regulation of TRAIL on the surface of
pDCs (Fig. 5 G). Similar to the pretreatment situation, the
cell populations stimulated with TLR7/8 agonists for 12 h
failed to express CD3 and CD56, strongly arguing against the
possibility of the involvement of T cells and/or NK cells.
These  ndings indicate that, upon stimulation with TLR7/8
agonists, mDCs and pDCs from peripheral blood acquire the
phenotype of mDCs and pDCs found in the peritumoral tis-
sue of IMQ-treated BCC lesions.
mDCs release perforin and granzyme B upon
TLR7 and TLR8 stimulation
The cytotoxic e ect of the perforin–granzyme B pathway
is only functional if the cytotoxic proteins are degranulated
and come into contact with target cells. To reveal possible
cytotoxic functions of perforin- and granzyme B–expressing
TLR7/8-stimulated DCs, we analyzed perforin and gran-
zyme B levels in the supernatants of sorted DCs after TLR7
and TLR8 ligation by ELISA. The viability of DCs was
controlled by annexin V ( ow cytometry) and trypan blue
stainings (counting chamber) and exceeded 90% before and
after cultures. Supernatants of PMA/ionomycin-stimulated,
negatively selected T cells served as positive controls. We
observed a substantial release of perforin and granzyme B
Figure 4. IMQ treatment induces the up-regulation of perforin
and granzyme B on mDCs in the peritumoral in ltrate. A detailed
quantitative analysis of the expression pattern of (A) perforin and
(C) granzyme B on leukocytes before, during, and after IMQ treatment
was performed by immuno uorescence double stainings with antibodies
against the indicated lytic molecules and lineage markers in all seven
treated patients. The data are given as absolute numbers of double-positive
cells SEM. Representative pictures of immunofluorescence triple
stainings with anti-CD11c, anti–HLA-DR, and (B) antiperforin or (D) anti–
granzyme B mAbs of the peritumoral in ltrate after 2 wk of treatment
with IMQ. (E) Triple immuno uorescence labeling of BCCs upon IMQ
treatment with anti-CD11c (PE), anti–HLA-DR (APC), and anti-iNOS (FITC)
or anti–TNF- (FITC) mAbs identi es iNOS and TNF- to be produced by
CD11c
HLA-DR
cells (arrows).
Figure 5. Peripheral blood–derived DCs express lytic molecules
after TLR7/8 stimulation. (A) Characterization of isolated peripheral
blood–derived pDCs and mDCs as HLA-DR
BDCA-2
and HLA-
DR
CD11c
, respectively, and lineage-negative cell populations by  ow
cytometry. (B and C) Analysis of intracellular expression levels of perforin
and granzyme B of isolated mDCs before and after culture with TLR7/8
agonists by  ow cytometry. (B) Representative dot plots of mDCs show
double-positive cells for perforin (FITC) and granzyme B (PE) after stimu-
lation with TLR7, TLR8, and TLR7/8 agonists. (C) Quantitative analysis of
granzyme B
and perforin
mDCs. Data are given as mean percentages of
perforin- or granzyme B–positive mDCs SEM from three independent
experiments. (D) Freshly isolated and TLR7/8-stimulated mDCs do not
display membrane-bound TRAIL (A488). Representative histogram plots
from three independent experiments show the anti-TRAIL reactivity (bold
line) and the matching isotype control (dashed line). (E and F) Determina-
tion of intracellular expression levels of perforin and granzyme B on
isolated pDCs before and after stimulation with TLR agonists. (E) Repre-
sentative dot plots of pDCs show a decrease of granzyme B–positive cells
after stimulation with TLR7 and TLR7/8 agonists. (F) Quantitative analysis
of pDCs expressing perforin and granzyme B. Data are given as mean
percentages of perforin- and granzyme B–positive cells SEM from
three independent experiments. (G) TLR7/8-stimulated, but not freshly
isolated, pDCs display membrane-bound TRAIL (A488). Representative
histogram plots from three different experiments show the anti-TRAIL
reactivity (bold line) and the matching isotype control (dashed line).
Dead cells were excluded by gating out propidium iodide–positive cells in
all experiments.
1446 TUMORICIDAL FUNCTION OF ACTIVATED DENDRITIC CELLS | Stary et al.
after stimulation of mDCs with TLR7 and TLR8 ligands, in
contrast to the vehicle control (Fig. 6, A and B). We de-
tected granzyme B in the supernatants of pDCs after TLR7
and TLR7/8 ligation but not after TLR8 ligation alone or
vehicle control (Fig. 6 B), which was consistent with our
FACS data described in the previous section. Perforin could
not be detected at any setting in the supernatant of pDCs
(Fig. 6 A). These data demonstrate that mDCs and pDCs
can be activated to release lytic molecules in a biologically
relevant fashion.
TLR7/8-stimulated mDCs and pDCs become killer cells:
involvement of different cytotoxic pathways
To evaluate the functional signi cance of lytic molecules
expressed by DCs after TLR7 and TLR8 ligation, we per-
formed cytotoxicity assays with puri ed peripheral blood–de-
rived mDCs and pDCs as e ector cells, and perforin-sensitive
K562 (Fig. S2 A, available at http://www.jem.org/cgi/
content/full/jem.20070021/DC1) and TRAIL-sensitive Jurkat
cell lines (31) as target cells. Although freshly isolated mDCs
hardly displayed any cytotoxic activity against K562 and Jur-
kat cells, mDCs stimulated with TLR7/8 agonists e ectively
lysed K562 but not Jurkat cells (Fig. 7 A). We observed the
opposite e ect in pDCs, in that they exhibited substantial
cytotoxic activity against Jurkat and not K562 cells after
TLR7/8 stimulation, but not when unstimulated (Fig. 7 B).
This diverse killing behavior indicated distinct mechanisms
operative in DC-mediated cytotoxicity after stimulation with
TLR7/8 ligands. To investigate the di erent cytotoxic path-
ways in greater detail, we performed inhibition experiments
with (a) concanamycin A, a speci c inhibitor of the perforin-
based cytotoxic pathway (32), (b) a neutralizing anti-TRAIL
antibody to block TRAIL-mediated killing, and (c) a neutral-
izing anti-Fas ligand antibody to inhibit Fas-induced killing.
As shown in Fig. 7 C, coincubation of TLR7/8-stimulated
mDCs and K562 in the presence of concanamycin A led to
a complete disappearance of the speci c lysis, whereas anti-
TRAIL and anti-Fas ligand antibodies had no e ect on the
mDC-mediated cytotoxicity. On the other hand, preincuba-
tion of TLR7/8-stimulated pDCs with an inhibitory anti-
TRAIL antibody essentially abolished the pDC cytotoxicity
against Jurkat cells (Fig. 7 D); the speci city of this inhibition
is also documented by the failure of concanamycin A and
anti-Fas ligand antibodies to block the killing of Jurkat cells
by TLR7/8-activated pDCs. To exclude the theoretical pos-
sibility of contaminating NK/NKT cells exhibiting cytotoxic
Figure 6. Secretion of perforin and granzyme B by mDCs upon
TLR7 and TLR8 stimulation. (A) Perforin and (B) granzyme B protein
levels were determined by ELISA in media conditioned by puri ed blood–
derived mDCs and pDCs after 12 h of stimulation with TLR7 and TLR8
agonists. Media conditioned by 12-h PMA/ionomycin-stimulated T cells
were used as positive controls. Data are given as means SEM from two
independent experiments with 2 10
5
cells in each setting.
Figure 7. Tumoricidal activity of blood-derived mDCs and pDCs
after stimulation with TLR7/8 agonists. Unstimulated, TLR7/8-
stimulated, or vehicle-stimulated (A) mDCs and (B) pDCs were cultured
with K562 or Jurkat as target cells for a conventional 2-h europium-TDA
release assay at the indicated effector/target cell ratios. Data represent the
means of triplicate wells SEM from two independent experiments.
(C and D) For inhibition of different cytotoxicity-inducing pathways, 12-h
TLR7/8-stimulated mDCs and pDCs as effector cells were preincubated
with 100 nM concanamycin A for 120 min, 5 g/ml anti-TRAIL for
30 min, 5 g/ml anti-Fas ligand for 30 min, or 5 g/ml of an IgG1 isotype
for 30 min. Target cells were (C) K562 for TLR7/8-stimulated mDCs and
(D) Jurkat for TLR7/8-stimulated pDCs. Cytotoxicity was determined by a
2-h europium-TDA release assay at the indicated effector/target cell
ratios. Data represent the means of triplicate wells SEM from two
independent experiments.
JEM VOL. 204, June 11, 2007 1447
ARTICLE
activity within isolated DC populations, we performed cyto-
toxicity assays with e ector cells consisting of irrelevant  ller
cells (i.e., A431) containing titrated numbers of NK cells. In
a setting of 7% NK cells titrated into A431 as  ller cells, we
observed a 10% cytotoxic activity against K562 and Jurkat
(Fig. S2 B), which represented less than half of the mDC-
mediated killing and less than one third of pDC-mediated
cytotoxicity. In addition, NK cells killed K562 and Jurkat
cells with a comparable intensity and did not show selective
killing like mDCs and pDCs. As NK cells and T cells never
accounted for >1% within puri ed DC populations (Fig.
5 A), these experiments strongly argue against contaminating
cells being even partly responsible for the cytotoxic activity
of the DC populations.
D I S C U S S I O N
IMQ is now an established therapeutic option in the treat-
ment of certain skin neoplasms such as genital warts (2), ac-
tinic keratoses (33), and BCCs (4, 5). In BCCs, cure rates are
similar to those achieved with other therapeutic procedures
(34, 35). Our study con rms these observations.
After binding to and signaling through TLR7 (7), IMQ
leads to up-regulation of cytokines with growth-inhibitory
(e.g., IFN-) and/or proin ammatory (e.g., TNF-, IL-1,
and IL-6) properties (1, 36). TLR-independent mechanisms,
operating directly by induction of apoptosis (19) and/or indi-
rectly by adenosine receptor signaling (18), could also con-
tribute to the clinically observed tumor regression. Even
though the IMQ-induced regression pattern of skin cancers
shows no signs of T cell memory, evidence exists that CD8
T cells are abundantly present in the in ammatory in ltrate
of IMQ-treated skin cancers (12, 14). A role for NK cells has
also been claimed (37). To address the involvement of cellu-
lar cytotoxicity in IMQ-induced tumor regression, in this
study we searched for the expression of lytic molecules on/in
di erent cell populations forming this in ltrate.
As opposed to the pretreatment situation, we detected
sizable numbers of perforin/granzyme B–positive cells, as
well as TRAIL-expressing cells, after 2 wk of IMQ treat-
ment. Interestingly, we observed that the majority of CD8
T cells and CD56
NK cells, which together formed a major
portion of the IMQ-induced in ltrate, were largely devoid
of antiperforin and anti–granzyme B reactivity. The reasons
for this  nding are not entirely clear. One could argue that
CD8
T cells and NK cells are simply bystander cells within
the in ammatory in ltrate and are not concerned with cyto-
toxic e ector functions. It is also conceivable that CD8
T cells discharge their cytotoxic granules directly after migra-
tion into the peritumoral tissue; after 2 wk of IMQ treat-
ment, the time point we had chosen for taking the biopsies,
these granules would therefore no longer be detectable
within their cytoplasm. We cannot de nitively exclude this
possibility. The occurrence of staining artifacts is unlikely, as
we detected perforin- and granzyme B–positive T cells in
skin biopsies of allergic contact eczema (Fig. S1, C and E).
To our surprise, double and triple labeling of IMQ treated
BCC sections revealed perforin and granzyme B to be ex-
pressed predominantly by lineage-negative CD11c
/HLA-
DR
mDCs. On the other hand, TRAIL, a molecule directly
involved in tumor cell killing by binding to its corresponding
death receptors (38–40), was expressed on several cell types,
including pDCs. We sought to better characterize the mDC
subset expressing cytotoxic molecules in situ and found that
they coexpress TNF- and iNOS, two proin ammatory
mediators with antitumor activity (41, 42). Thus, these cells
corresponded to TNF-– and iNOS-producing DCs, which
were shown to originate from blood-derived monocytes (43)
and exhibit e ector functions in bacterial infections in mice
(29) and to correlate with disease activity in psoriasis in
humans (28).
We were not able to isolate DCs from IMQ-treated BCC
lesions in numbers that would have su ced to conduct
meaningful functional experiments. We therefore decided
to test the functional relevance of our immunostaining re-
sults in studies with peripheral blood–derived mDCs and
pDCs stimulated with TLR7 and TLR8 agonists. When
activated in vitro with such compounds, peripheral blood–
derived mDCs not only expressed and released perforin
and granzyme B but also exhibited substantial cytotoxicity
against MHC class I–negative (K562) but not MHC class
I–positive (Jurkat) tumor cell lines. This lytic activity was
abolished by pretreatment of e ector cells with concana-
mycin A, an inhibitor of perforin-mediated cytotoxicity.
To rule out the possibility that T cells and/or NK/NKT
cells would contaminate the mDC e ector cell population,
we analyzed puri ed DCs of the peripheral blood by FACS
stainings and by real-time PCR. Results obtained showed
that these cells lacked all lineage markers at the protein level
(Fig. 5 A) and were devoid of CD3 transcripts (unpub-
lished data). We also titrated CD56
cells into a population
of nonfunctional  ller cells and found that the lytic capacity
of this cell mixture was considerably lower than that of
puri ed TLR7/8-activated DCs, even when numbers of
CD56
NK cells were greater than the peak levels of all
lineage
CD11c
cells ever encountered in our mDC prep-
arations (Fig. S2 B). Evidence for a direct cytotoxic poten-
tial of mDCs has also been presented by other investigators,
but their  ndings di er from ours with regard to the acti-
vation status of the DCs and the type of lytic molecules
involved (24, 44–48).
The emergence of pDCs in the peritumoral tissue of
IMQ-treated skin cancers has been demonstrated in mice
(23) and in humans (49). We con rmed the occurrence of
IFN-–producing pDCs in the peritumoral in ltrate during
IMQ treatment. As IFN- induces cytotoxic molecules on
NK cells (50) and T cells (51), pDCs may indirectly contrib-
ute to the elimination of tumor cells. In this study, we asked
whether pDCs could directly acquire e ector functions via
up-regulation of cytotoxic molecules and, thus, play an active
role in tumor clearance. In contrast to mDCs, pDCs occur-
ring within the peritumoral in ltrate expressed neither perfo-
rin nor granzyme B. At  rst glance, this was rather surprising,
1448 TUMORICIDAL FUNCTION OF ACTIVATED DENDRITIC CELLS | Stary et al.
because unstimulated pDCs from peripheral blood clearly
displayed intracellular anti–granzyme B (Fig. 5 E) (52, 53).
Our further observation that pDC expression levels of gran-
zyme B are continuously decreasing upon TLR7 ligation may
be an explanation for the results of our in situ stainings. In any
event, granzyme B does not seem to contribute to the antitumor
activity of pDCs, as inhibition of the perforin/granzyme B–
mediated cytotoxic pathway of TLR7/8-stimulated pDCs
had no e ect on the cytotoxic activity of these cells. The
physiological role of granzyme B in pDCs therefore remains
to be established.
On the basis of our  ndings that pDCs occurring in the
IMQ-induced in ltrate expressed TRAIL and that TLR7-
stimulated peripheral blood–derived pDCs displayed TRAIL
on their surface and used this molecule for cytotoxic pur-
poses, we would like to propose that pDCs have the potential
to act as anticancer e ector cells. Our results con rm and
extend observations by others demonstrating the TRAIL-
mediated cytotoxicity of pDCs after stimulation by in uenza
virus or TLR agonists (25).
The biological importance of our  ndings was underlined
by the observation that IMQ-treated BCC cells, surrounded
by CD123
pDCs, are TRAIL 1 (DR4)–positive (Fig. 3 D)
and express only negligible amounts of MHC class I mole-
cules in situ (unpublished data). Even though we are aware of
the fact that TLR7/8-stimulated DCs from peripheral blood
are not necessarily equivalent to DCs that accumulate in the
skin in response to TLR activation, our data suggest that per-
forin/granzyme B–bearing cytotoxic mDCs and TRAIL-
expressing pDCs participate in IMQ-induced tumor regression
by MHC class I–independent and death receptor–dependent
killing of BCC cells, respectively. This does not exclude the
possibility that other e ector molecules (e.g., TNF-) are also
involved in this process.
Many questions remain to be resolved. To begin with, it
has to be determined whether all DCs or only subpopulations
thereof can be transformed into e ector cells upon appropri-
ate stimulation. Although we did not  nd NKG2D, CD94,
CD16, or CD56 to be expressed on cytotoxic mDCs or pDCs
before or after TLR7/8 stimulation (unpublished data), the
relationship of human DCs described in our study to the
newly described IFN-producing killer DCs in mice (54, 55)
has to be de ned. It also needs to be clari ed whether nat-
ural TLR7/8 ligands (viral single-stranded RNA) or stimuli
other than TLR7/8 agonists can endow DCs with lytic prop-
erties, which signaling cascades are involved in this process,
and whether tumor cells, by an as of yet unknown recog-
nition process, are triggering the release of cytotoxic gran-
ules by activated DCs. All of this information is required to
devise strategies for the use of cytotoxic DCs as tools in anti-
cancer immunotherapy.
Finally, it will be of great interest to determine whether DC
cytotoxicity ever occurs in the course of pathophysiological
DC-driven immune responses. If so, the attractive possibility
exists that DCs may use this e ector mechanism to down-
regulate the immune reactions that they initiate themselves.
MATERIALS AND METHODS
Patients and tissue material. Seven patients with histologically con rmed
super cial BCC (three females and four males; mean age 78.3 8.2) were
enrolled in the study after giving their written informed consent. BCCs were
located on the head (n 2), the neck (n 2), and the back (n 3). None
of the patients were immunosuppressed or had any dermatological disease
that could exacerbate upon IMQ (Aldara; 3M Pharmaceuticals) treatment.
BCCs included in this study were therapy naive and did not receive any
BCC-speci c therapy (e.g., photodynamic therapy, 5- uorouracil, or IFN-)
before this study.
Patients received a once daily,  ve-times-a-week dosing regimen of
IMQ 5% cream for 6 wk, according to the o cial treatment guidelines
approved for BCC. The cream was applied as a thin layer without occlusion
overnight exactly on the BCC sites. Safety and e cacy were clinically
monitored weekly and by blood examinations at weeks 3 and 6. After 6 wk
of treatment, response rates were determined by clinical examination
and histological analysis of a punch biopsy taken from formerly a ected
BCC sites.
For immuno uorescence analysis, 4-mm punch biopsies were taken un-
der local anesthesia (1% lidocaine) from untreated BCCs after 2 wk of IMQ
treatment, when the clinical signs of in ammation were strongest, and after
6 wk of IMQ therapy at the treatment endpoint. The biopsies were embedded
in optimum cutting tissue compound (Tissue-Tek; Sakura Finetek), snap-
frozen in liquid nitrogen, and stored at –80C until further processing.
Studies involving patient material were performed according to the
Declaration of Helsinki. Biopsies were routinely taken from every patient for
diagnostic purposes, followed by an informed consent about a possible scien-
ti c use of the data obtained as recommended by the local ethics committee
(Federal Academic Hospital Feldkirch).
Immuno uorescence staining. Single and multicolor immuno uores-
cence staining procedures were performed as previously described (56). The
mAbs used in this study and their sources are shown in Table I.
Apoptotic cells were detected using a kit from Roche Molecular Bio-
chemicals to visualize DNA breaks as TUNEL
, according to the manufac-
turer’s guidelines. After blocking with normal mouse serum for 20 min to
diminish background staining, sections were counterstained with a puri ed
antipancytokeratin mAb overnight at 4C. Puri ed antibody binding was
visualized by incubating the sections with rhodamine (TRITC)-conjugated
A niPure F(ab)2 fragment goat anti–mouse IgG (HL). The evaluation of
immuno uorescence results was performed as previously described (56). In
brief, biopsy specimens were read in a blinded fashion by two independent
investigators with a mean observer coe cient <10%. Labeled cells were
enumerated per visual  eld and expressed as the number of cells per millimeter/
basement membrane epidermis and millimeter
2
dermis SEM.
Cell preparations from peripheral blood. Peripheral blood samples
were purchased as bu y coats from the Vienna Red Cross Center. DCs were
isolated as follows: PBMCs were obtained by Ficoll-Plaque density gradient
centrifugation (Histopaque-1077; Sigma-Aldrich) and depleted of T cells,
B cells, NK cells, hemopoietic stem cells, monocytes, platelets, and erythro-
cytes by anti-CD3/CD19/CD56/CD34/CD14/CD16/CD41/CD235a
(3 g/ml each) immunolabeling and anti–mouse IgG immunomagnetic deple-
tion (MACS; Miltenyi Biotec). The remaining cell fraction was then divided
into mDCs and pDCs by positive selection using anti-CD1c and, thereafter,
anti-CD304 (BDCA-4) microbeads (MACS). The purity of isolated DC popu-
lations was 93–99%, as determined by  ow cytometric analysis (FACScan; BD
Biosciences) with either anti-CD11c/HLA-DR or anti-CD303 (BDCA-2)/
HLA-DR stainings. None of the other leukocyte subpopulations tested
individually (T cells, B cells, NK cells, and monocytes) accounted for > 1%
of the sorted cell population. T cells were prepared as a positive control
for the analysis of the perforin and granzyme B release by negative selec-
tion from PBMCs using anti-CD11c/CD14/CD16/CD19/CD34/CD41/
CD56/CD123/CD235a (3 g/ml each) immunolabeling and anti–mouse
IgG immunomagnetic depletion (MACS). NK cells used as positive controls
JEM VOL. 204, June 11, 2007 1449
ARTICLE
for cytotoxicity assays were separated by anti-CD56 (3 g/ml each) immu-
nolabeling and anti–mouse IgG immunomagnetic selection after T cell de-
pletion (MACS). The purity of T cells and NK cells was >99%, as determined
by FACS analysis.
DC stimulation with TLR agonists. Synthetic agonists to TLR7 (3M-
001), TLR8 (3M-002), TLR7/8 (3M-003), and an inactive small molecule
TLR7/8 analogue that served as a negative control (3M-006) were provided
by R.L. Miller (3M Pharmaceuticals, St. Paul, MN). Isolated mDCs and
pDCs were washed and resuspended in RPMI 1640 medium (Invitrogen)
supplemented with heat-inactivated 10% FCS (Invitrogen) and 1% penicillin/
streptomycin (Invitrogen) and cultured with 4 M 3M-001, 3M-002,
3M-003, or 3M-006 for 12 h at 2 10
6
cells/ml in 0.25 ml in 96-well plates
or 0.5 ml in 48-well plates. Viability of the cells was measured with trypan
blue and annexin V (BD Biosciences) before and after cultures, according to
the manufacturer’s instructions. Cells were analyzed by FACS stainings for
detection of lytic molecules or used as e ector cells in cytotoxicity assays.
Media conditioned by DC subsets were collected and stored at 20C for
further analysis.
Flow cytometry. To analyze lytic molecules on blood-derived DCs, we
performed intra- and extracellular FACS stainings of isolated DC popu-
lations before and after culture with TLR agonists. After staining of DC
populations with anti–BDCA-2 (Miltenyi Biotec) or anti-CD11c (BD Bio-
sciences) mAbs, cells were  xed and permeabilized with a cell permeabili-
zation kit (An Der Grub), according to the manufacturer’s instructions for
intracellular protein detection. Cells were incubated with antiperforin FITC
(BD Biosciences) and anti–granzyme B PE (PeliCluster; Sanquin) antibodies.
Surface TRAIL expression was visualized by staining with an anti-TRAIL
mAb (R&D Systems) after labeling the puri ed antibody with a protein
labeling kit (FluoReporter Oregon Green A488; Invitrogen), according to
the manufacturer’s instructions. Purity controls of DC populations before
and after culture were performed by triple stainings with antibodies against
HLA-DR FITC (BD Biosciences), BDCA-2 PE (Miltenyi Biotec), or
CD11c PE (BD Biosciences) and CD3 PerCp, CD14 PerCp, CD19 PerCp,
or CD56 PECy5.
After incubation with these mAbs, DCs were washed twice with ice-
cold PBS and subjected to  ow cytometric analysis using a  ow cytometer
(FACScan).
Measurement of perforin and granzyme B in the supernatants. Super-
natants were harvested after stimulation of isolated mDCs and pDCs with
TLR7, TLR8, or TLR7/8 agonists or vehicle control for 12 h and stored at
20C. Perforin and granzyme B levels were quanti ed by ELISA kits (Dia-
clone Research), according to the manufacturer’s instructions. Supernatants
of T cells stimulated with 50 ng/ml PMA (Sigma-Aldrich) and 1 g/ml
ionomycin (Sigma-Aldrich) for 12 h served as positive controls.
Cytotoxicity assays. The ability of DCs to exert cytotoxicity was assessed
in a conventional 2-h europium-TDA release assay (PerkinElmer), as previ-
ously described (57), according to the manufacture’s instructions. Data were
expressed as the percentage of cytotoxicity calculated by the following
formula: cytotoxicity (%) (experimental release – spontaneous release)/
(maximum release – spontaneous release) 100.
We used the chronic myelogenous leukemia cell line K562 (provided
by C. Wagner, Medical University of Vienna, Vienna, Austria) and the
acute T cell leukemia cell line Jurkat (provided by P. Meraner, Medical
University of Vienna, Vienna, Austria) as target cells. In a 96-well plate,
5 10
3
target cells per well were incubated with mDCs and pDCs in dif-
ferent e ector/target ratios (starting at 25:1) in triplicates. For inhibition of
TRAIL- and Fas ligand–dependent lysis, 5 g/ml of azide-free neutralizing
anti-TRAIL (clone 75411; R&D Systems) and 5 g/ml of anti–Fas ligand
(clone NOK-1; BD Biosciences), respectively, were added to e ector cells
30 min before the addition of target cells. A 30-min preculture of e ector
cells with 5 g/ml of an IgG1 isotype (Sigma-Aldrich) served as negative
control. For inhibition of the perforin-based cytotoxicity, e ector cells were
treated for 120 min with 100 nM concanamycin A (Sigma-Aldrich) before tar-
get cells were added.
Online supplemental material. Fig. S1 shows that mDCs, but not
pDCs, display CD83 in IMQ-treated BCC and that perforin/granzyme
B–containing T cells occur in lesions of allergic contact dermatitis, but not
IMQ-treated BCC. Fig. S2 depicts NK cell–mediated cytotoxicity. Online
supplemental material is available at http://www.jem.org/cgi/content/full/
jem.20070021/DC1.
We thank Richard L. Miller (3M Pharmaceuticals) for providing agonists to TLR7
and TLR8 and Drs. Dieter Maurer and Ethan Shevach for critically reading the
manuscript. We would also like to thank Sabine Altrichter for technical advice and
Andreas Ebner for preparing the  gures.
The authors have no con icting  nancial interests.
Submitted: 2 January 2007
Accepted: 9 May 2007
R E F E R E N C E S
1. Weeks, C.E., and S.J. Gibson. 1994. Induction of interferon and other
cytokines by imiquimod and its hydroxylated metabolite R-842 in hu-
man blood cells in vitro. J. Interferon Res. 14:81–85.
2. Garland, S.M., R. Waddell, A. Mindel, I.M. Denham, and J.C.
McCloskey. 2006. An open-label phase II pilot study investigating the
optimal duration of imiquimod 5% cream for the treatment of external
genital warts in women. Int. J. STD AIDS. 17:448–452.
3. Theos, A.U., R. Cummins, N.B. Silverberg, and A.S. Paller. 2004.
E ectiveness of imiquimod cream 5% for treating childhood molluscum
contagiosum in a double-blind, randomized pilot trial. Cutis. 74:134–
138, 141–142.
4. Gollnick, H., C.G. Barona, R.G. Frank, T. Ruzicka, M. Megahed, V.
Tebbs, M. Owens, and P. Stampone. 2005. Recurrence rate of super -
cial basal cell carcinoma following successful treatment with imiquimod
5% cream: interim 2-year results from an ongoing 5-year follow-up
study in Europe. Eur. J. Dermatol. 15:374–381.
5. Peris, K., E. Campione, T. Micantonio, G.C. Marulli, M.C. Fargnoli,
and S. Chimenti. 2005. Imiquimod treatment of super cial and nod-
ular basal cell carcinoma: 12-week open-label trial. Dermatol. Surg.
31:318–323.
6. Fleming, C.J., A.M. Bryden, A. Evans, R.S. Dawe, and S.H. Ibbotson.
2004. A pilot study of treatment of lentigo maligna with 5% imiquimod
cream. Br. J. Dermatol. 151:485–488.
7. Akira, S., and H. Hemmi. 2003. Recognition of pathogen-associated
molecular patterns by TLR family. Immunol. Lett. 85:85–95.
8. Diebold, S.S., T. Kaisho, H. Hemmi, S. Akira, and C. Reis e Sousa.
2004. Innate antiviral responses by means of TLR7-mediated recogni-
tion of single-stranded RNA. Science. 303:1529–1531.
9. Heil, F., H. Hemmi, H. Hochrein, F. Ampenberger, C. Kirschning, S.
Akira, G. Lipford, H. Wagner, and S. Bauer. 2004. Species-speci c rec-
ognition of single-stranded RNA via toll-like receptor 7 and 8. Science.
303:1526–1529.
10. Medzhitov, R., P. Preston-Hurlburt, and C.A. Janeway Jr. 1997. A
human homologue of the Drosophila Toll protein signals activation of
adaptive immunity. Nature. 388:394–397.
11. Akira, S., and K. Takeda. 2004. Toll-like receptor signalling. Nat. Rev.
Immunol. 4:499–511.
12. Michalopoulos, P., N. Yawalkar, M. Bronnimann, A. Kappeler, and
L.R. Braathen. 2004. Characterization of the cellular in ltrate during
successful topical treatment of lentigo maligna with imiquimod. Br. J.
Dermatol. 151:903–906.
13. Barnetson, R.S., A. Satchell, L. Zhuang, H.B. Slade, and G.M. Halliday.
2004. Imiquimod induced regression of clinically diagnosed super cial
basal cell carcinoma is associated with early in ltration by CD4 T cells
and dendritic cells. Clin. Exp. Dermatol. 29:639–643.
14. Smith, K.J., S. Hamza, and H. Skelton. 2004. Topical imidazoquinoline
therapy of cutaneous squamous cell carcinoma polarizes lymphoid and
1450 TUMORICIDAL FUNCTION OF ACTIVATED DENDRITIC CELLS | Stary et al.
monocyte/macrophage populations to a Th1 and M1 cytokine pattern.
Clin. Exp. Dermatol. 29:505–512.
15. Kadowaki, N., S. Ho, S. Antonenko, R.W. Malefyt, R.A. Kastelein, F.
Bazan, and Y.J. Liu. 2001. Subsets of human dendritic cell precursors
express di erent Toll-like receptors and respond to di erent microbial
antigens. J. Exp. Med. 194:863–869.
16. Jarrossay, D., G. Napolitani, M. Colonna, F. Sallusto, and A. Lanzavecchia.
2001. Specialization and complementarity in microbial molecule re-
cognition by human myeloid and plasmacytoid dendritic cells. Eur. J.
Immunol. 31:3388–3393.
17. Peng, G., Z. Guo, Y. Kiniwa, K.S. Voo, W. Peng, T. Fu, D.Y. Wang,
Y. Li, H.Y. Wang, and R.F. Wang. 2005. Toll-like receptor 8-mediated
reversal of CD4 regulatory T cell function. Science. 309:1380–1384.
18. Schön, M.P., M. Schön, and K.N. Klotz. 2006. The small antitumoral
immune response modi er imiquimod interacts with adenosine recep-
tor signaling in a TLR7- and TLR8-independent fashion. J. Invest.
Dermatol. 126:1338–1347.
19. Schön, M., A.B. Bong, C. Drewniok, J. Herz, C.C. Geilen, J.
Reifenberger, B. Benninghoff, H.B. Slade, H. Gollnick, and M.P.
Schön. 2003. Tumor-selective induction of apoptosis and the small-
molecule immune response modi er imiquimod. J. Natl. Cancer Inst.
95:1138–1149.
20. Grebenova, D., K. Kuzelova, O. Fuchs, P. Halada, V. Havlicek, I.
Marinov, and Z. Hrkal. 2004. Interferon-alpha suppresses proliferation
of chronic myelogenous leukemia cells K562 by extending cell cycle
S-phase without inducing apoptosis. Blood Cells Mol. Dis. 32:262–269.
21. Majewski, S., M. Marczak, B. Mlynarczyk, B. Benningho , and S.
Jablonska. 2005. Imiquimod is a strong inhibitor of tumor cell-induced
angiogenesis. Int. J. Dermatol. 44:14–19.
22. Li, V.W., W.W. Li, K.E. Talcott, and A.W. Zhai. 2005. Imiquimod as
an antiangiogenic agent. J. Drugs Dermatol. 4:708–717.
23. Palamara, F., S. Meindl, M. Holcmann, P. Lührs, G. Stingl, and M.
Sibilia. 2004. Identi cation and characterization of pDC-like cells in
normal mouse skin and melanomas treated with imiquimod. J. Immunol.
173:3051–3061.
24. Fanger, N.A., C.R. Maliszewski, K. Schooley, and T.S. Gri th.
1999. Human dendritic cells mediate cellular apoptosis via tumor ne-
crosis factor–related apoptosis-inducing ligand (TRAIL). J. Exp. Med.
190:1155–1164.
25. Chaperot, L., A. Blum, O. Manches, G. Lui, J. Angel, J.P. Molens, and
J. Plumas. 2006. Virus or TLR agonists induce TRAIL-mediated cyto-
toxic activity of plasmacytoid dendritic cells. J. Immunol. 176:248–255.
26. Tucker, S.B., J.W. Polasek, A.J. Perri, and E.A. Goldsmith. 2006. Long-
term follow-up of basal cell carcinomas treated with perilesional inter-
feron alfa 2b as monotherapy. J. Am. Acad. Dermatol. 54:1033–1038.
27. Thyrell, L., S. Erickson, B. Zhivotovsky, K. Pokrovskaja, O. Sangfelt, J.
Castro, S. Einhorn, and D. Grander. 2002. Mechanisms of Interferon-
alpha induced apoptosis in malignant cells. Oncogene. 21:1251–1262.
28. Lowes, M.A., F. Chamian, M.V. Abello, J. Fuentes-Duculan, S.L. Lin,
R. Nussbaum, I. Novitskaya, H. Carbonaro, I. Cardinale, T. Kikuchi,
et al. 2005. Increase in TNF-alpha and inducible nitric oxide synthase-
expressing dendritic cells in psoriasis and reduction with efalizumab
(anti-CD11a). Proc. Natl. Acad. Sci. USA. 102:19057–19062.
29. Serbina, N.V., T.P. Salazar-Mather, C.A. Biron, W.A. Kuziel, and
E.G.P. Am. 2003. TNF/iNOS-producing dendritic cells mediate in-
nate immune defense against bacterial infection. Immunity. 19:59–70.
30. Yawalkar, N., R.E. Hunger, C. Buri, S. Schmid, F. Egli, C.U. Brand,
C. Mueller, W.J. Pichler, and L.R. Braathen. 2001. A comparative
study of the expression of cytotoxic proteins in allergic contact dermati-
tis and psoriasis: spongiotic skin lesions in allergic contact dermatitis are
highly in ltrated by T cells expressing perforin and granzyme B. Am. J.
Pathol.
158:803–808.
31. Zauli, G., S. Sancilio, A. Cataldi, N. Sabatini, D. Bosco, and R. Di
Pietro. 2005. PI-3K/Akt and NF-kappaB/IkappaBalpha pathways are
activated in Jurkat T cells in response to TRAIL treatment. J. Cell.
Physiol. 202:900–911.
32. Kataoka, T., N. Shinohara, H. Takayama, K. Takaku, S. Kondo, S.
Yonehara, and K. Nagai. 1996. Concanamycin A, a powerful tool
for characterization and estimation of contribution of perforin- and
Fas-based lytic pathways in cell-mediated cytotoxicity. J. Immunol.
156:3678–3686.
33. Korman, N., R. Moy, M. Ling, R. Matheson, S. Smith, S. McKane,
and J.H. Lee. 2005. Dosing with 5% imiquimod cream 3 times per week
for the treatment of actinic keratosis: results of two phase 3, randomized,
double-blind, parallel-group, vehicle-controlled trials. Arch. Dermatol.
141:467–473.
34. Nikkels, A.F., C. Pierard-Franchimont, N. Nikkels-Tassoudji, R.
Bourguignon, and G.E. Pierard. 2005. Photodynamic therapy and imiqui-
mod immunotherapy for basal cell carcinomas. Acta Clin. Belg. 60:227–234.
35. Dixon, A.J. 2005. Multiple super cial basal cell carcinomata–topical im-
iquimod versus curette and cryotherapy. Aust. Fam. Physician. 34:49–52.
36. Sidky, Y.A., E.C. Borden, C.E. Weeks, M.J. Reiter, J.F. Hatcher, and
G.T. Bryan. 1992. Inhibition of murine tumor growth by an interferon-
inducing imidazoquinolinamine. Cancer Res. 52:3528–3533.
37. Sullivan, T.P., T. Dearaujo, V. Vincek, and B. Berman. 2003. Evaluation
of super cial basal cell carcinomas after treatment with imiquimod 5%
cream or vehicle for apoptosis and lymphocyte phenotyping. Dermatol.
Surg. 29:1181–1186.
38. Wiley, S.R., K. Schooley, P.J. Smolak, W.S. Din, C.P. Huang, J.K.
Nicholl, G.R. Sutherland, T.D. Smith, C. Rauch, C.A. Smith, et al.
1995. Identi cation and characterization of a new member of the TNF
family that induces apoptosis. Immunity. 3:673–682.
39. Schneider, P., M. Thome, K. Burns, J.L. Bodmer, K. Hofmann, T.
Kataoka, N. Holler, and J. Tschopp. 1997. TRAIL receptors 1 (DR4)
and 2 (DR5) signal FADD-dependent apoptosis and activate NF-kappaB.
Immunity. 7:831–836.
40. Wang, S., and W.S. El-Deiry. 2003. TRAIL and apoptosis induction by
TNF-family death receptors. Oncogene. 22:8628–8633.
41. Xu, W., L. Liu, and I.G. Charles. 2002. Microencapsulated iNOS-
expressing cells cause tumor suppression in mice. FASEB J. 16:213–215.
42. Mocellin, S., M. Provenzano, M. Lise, D. Nitti, and C.R. Rossi. 2003.
Increased TIA-1 gene expression in the tumor microenvironment after
locoregional administration of tumor necrosis factor-alpha to patients
with soft tissue limb sarcoma. Int. J. Cancer. 107:317–322.
43. Serbina, N.V., and E.G.P. Am. 2006. Monocyte emigration from bone
marrow during bacterial infection requires signals mediated by chemo-
kine receptor CCR2. Nat. Immunol. 7:311–317.
44. Janjic, B.M., G. Lu, A. Pimenov, T.L. Whiteside, W.J. Storkus, and
N.L. Vujanovic. 2002. Innate direct anticancer e ector function of hu-
man immature dendritic cells. I. Involvement of an apoptosis-inducing
pathway. J. Immunol. 168:1823–1830.
45. Schmitz, M., S. Zhao, Y. Deuse, K. Schakel, R. Wehner, H. Wohner, K.
Holig, F. Wienforth, A. Kiessling, M. Bornhauser, et al. 2005. Tumoricidal
potential of native blood dendritic cells: direct tumor cell killing and acti-
vation of NK cell-mediated cytotoxicity. J. Immunol. 174:4127–4134.
46. Lu, G., B.M. Janjic, J. Janjic, T.L. Whiteside, W.J. Storkus, and N.L.
Vujanovic. 2002. Innate direct anticancer e ector function of human
immature dendritic cells. II. Role of TNF, lymphotoxin-alpha(1)beta(2),
Fas ligand, and TNF-related apoptosis-inducing ligand. J. Immunol.
168:1831–1839.
47. Vanderheyde, N., E. Aksoy, Z. Amraoui, P. Vandenabeele, M.
Goldman, and F. Willems. 2001. Tumoricidal activity of monocyte-
derived dendritic cells: evidence for a caspase-8-dependent, Fas-associated
death domain-independent mechanism.
J. Immunol. 167:3565–3569.
48. Trinite, B., C. Chauvin, H. Peche, C. Voisine, M. Heslan, and R. Josien.
2005. Immature CD4- CD103 rat dendritic cells induce rapid caspase-
independent apoptosis-like cell death in various tumor and nontumor cells
and phagocytose their victims. J. Immunol. 175:2408–2417.
49. Urosevic, M., R. Dummer, C. Conrad, M. Beyeler, E. Laine, G. Burg,
and M. Gilliet. 2005. Disease-independent skin recruitment and activa-
tion of plasmacytoid predendritic cells following imiquimod treatment.
J. Natl. Cancer Inst. 97:1143–1153.
50. Sato, K., S. Hida, H. Takayanagi, T. Yokochi, N. Kayagaki, K. Takeda,
H. Yagita, K. Okumura, N. Tanaka, T. Taniguchi, and K. Ogasawara.
2001. Antiviral response by natural killer cells through TRAIL gene
induction by IFN-alpha/beta. Eur. J. Immunol. 31:3138–3146.
JEM VOL. 204, June 11, 2007 1451
ARTICLE
51. Kayagaki, N., N. Yamaguchi, M. Nakayama, H. Eto, K. Okumura, and
H. Yagita. 1999. Type I interferons (IFNs) regulate tumor necrosis factor–
related apoptosis-inducing ligand (TRAIL) expression on human T cells:
a novel mechanism for the antitumor e ects of type I IFNs. J. Exp. Med.
189:1451–1460.
52. Rissoan, M.C., T. Duhen, J.M. Bridon, N. Bendriss-Vermare, C.
Peronne, B. de Saint Vis, F. Briere, and E.E. Bates. 2002. Subtractive
hybridization reveals the expression of immunoglobulin-like transcript 7,
Eph-B1, granzyme B, and 3 novel transcripts in human plasmacytoid
dendritic cells. Blood. 100:3295–3303.
53. Santoro, A., A. Majorana, L. Roversi, F. Gentili, S. Marrelli, W. Vermi,
E. Bardellini, P. Sapelli, and F. Facchetti. 2005. Recruitment of den-
dritic cells in oral lichen planus. J. Pathol. 205:426–434.
54. Taieb, J., N. Chaput, C. Menard, L. Apetoh, E. Ullrich, M. Bonmort, M.
Pequignot, N. Casares, M. Terme, C. Flament, et al. 2006. A novel dendritic
cell subset involved in tumor immunosurveillance. Nat. Med. 12:214–219.
55. Chan, C.W., E. Crafton, H.N. Fan, J. Flook, K. Yoshimura, M. Skarica, D.
Brockstedt, T.W. Dubensky, M.F. Stins, L.L. Lanier, et al. 2006. Interferon-
producing killer dendritic cells provide a link between innate and adap-
tive immunity. Nat. Med. 12:207–213.
56. Stary, G., C. Bangert, G. Stingl, and T. Kopp. 2005. Dendritic cells in
atopic dermatitis: expression of FcepsilonRI on two distinct in ammation-
associated subsets. Int. Arch. Allergy Immunol. 138:278–290.
57. Blomberg, K., R. Hautala, J. Lovgren, V.M. Mukkala, C. Lindqvist,
and K. Akerman. 1996. Time-resolved  uorometric assay for natural
killer activity using target cells labelled with a  uorescence enhancing
ligand. J. Immunol. Methods. 193:199–206.
... 12,13 Although keratinocytes do not express TLR7, [14][15][16] the TLR7-mediated activation of plasmacytoid dendritic cells (pDCs) results in the induction of a strong inflammatory response. 17 Meanwhile, we recently showed that a coordinated expression of AA transporters and their function plays an essential role in the TLR-mediated activation of human pDCs. 18 Ultimately, the innate inflammatory response induced in IMQ-treated skin triggers the recruitment of effector T cells, among which CD8 + T cells are of special importance for achieving cancer control. ...
Article
Full-text available
T cells protect tissues from cancer. Although investigations in mice showed that amino acids (AA) critically regulate T cell immunity, this remains poorly understood in humans. Here, we describe the AA composition of interstitial fluids in keratinocyte-derived skin cancers (KDSCs) and study the effect of AA on T cells using models of primary human cells and tissues. Gln contributed to ∼15% of interstitial AAs and promoted interferon gamma (IFN-γ), but not granzyme B (GzB) expression, in CD8⁺ T cells. Furthermore, the Toll-like receptor 7 agonist imiquimod (IMQ), a common treatment for KDSCs, down-regulated the metabolic gatekeepers c-MYC and mTORC1, as well as the AA transporter ASCT2 and intracellular Gln, Asn, Ala, and Asp in T cells. Reduced proliferation and IFN-γ expression, yet increased GzB, paralleled IMQ effects on AA. Finally, Gln was sufficient to promote IFN-γ-production in IMQ-treated T cells. Our findings indicate that Gln metabolism can be harnessed for treating KDSCs.
... This compound is believed to activate immune cells as a Toll-like receptor-7 agonist. It exerts its antiviral effects by activating dendritic cells and inducing cytokines such as tumor necrosis factor-alpha (TNF-α), interferon-alpha (IFN-α), and interleukins (ILs) [9]. Multiple studies have shown that Imiquimod could be a potential conservative treatment for precursor cervical lesions by accelerating viral clearance [10][11][12]. ...
Article
Full-text available
Simple Summary There are publications on the use of Imiquimod in cervical intraepithelial neoplasia (CIN) and HPV clearance; however, the literature is not consistent about its efficacy. Moreover, in cervical precancers, surgical solutions are widely accepted therapies, despite their association with increased obstetrical complications, such as miscarriage and preterm birth. Therefore, a conservative solution is needed. Topical Imiquimod reduced CIN and enhanced HPV clearance, though surgical intervention conization was found to be more effective than Imiquimod treatment. Side effects were common, though mostly mild. Topical Imiquimod could be a valuable therapeutic option for CIN patients, especially for women who have future pregnancy desires. Imiquimod should be incorporated into guidelines as evidence shows it is effective and safe. Abstract Introduction: Topical Imiquimod is an immune response modifier approved for the off-label use of vulvar intraepithelial neoplasia. We conducted this systematic review and meta-analysis to investigate the efficacy and safety of Imiquimod in treating cervical intraepithelial neoplasia (CIN) and human papillomavirus (HPV)-positive patients. Methods: The study was prospectively registered (CRD420222870) and involved a comprehensive systematic search of five medical databases on 10 October 2022. We included articles that assessed the use of Imiquimod in cervical dysplasia and HPV-positive patients. Pooled proportions, risk ratios (RRs), and corresponding 95% confidence intervals (CIs) were calculated using a random effects model to generate summary estimates. Statistical heterogeneity was assessed using I² tested by the Cochran Q tests. Results: Eight articles reported on 398 patients who received Imiquimod out of 672 patients. Among CIN-2–3 patients, we observed a pooled regression rate of 61% (CI: 0.46–0.75; I²: 77%). When compared, Imiquimod was inferior to conization (RR: 0.62; CI: 0.42–0.92; I²: 64%). The HPV clearance rate in women who completed Imiquimod treatment was 60% (CI: 0.31–0.81; I²: 57%). The majority of side effects reported were mild to moderate in severity. Conclusions: Our findings indicate that topical Imiquimod is safe and effective in reducing cervical intraepithelial neoplasia and promoting HPV clearance. However, it was found to be inferior compared to conization. Imiquimod could be considered a potential medication for high-grade CIN patients and should be incorporated into guidelines for treating cervical dysplasia.
... Mechanistically, our findings suggest that PCM derived immunosuppressive cytokines as TGF-b and reduced glycolysis rewired fully differentiated pDCs towards a tolerogenic state (summarized by the graphical abstract in Figure 8). Properly activated pDCs drive melanoma regression, as shown by experimental model and clinical studies (95)(96)(97)(98). In PCM, pDC infiltration is heterogeneous, occurs early, and localizes at the invasive margin, the site where pDCs interact with CD8 + T cells (46,99,100). ...
Article
Full-text available
Introduction Plasmacytoid dendritic cells (pDCs) infiltrate a large set of human cancers. Interferon alpha (IFN-α) produced by pDCs induces growth arrest and apoptosis in tumor cells and modulates innate and adaptive immune cells involved in anti-cancer immunity. Moreover, effector molecules exert tumor cell killing. However, the activation state and clinical relevance of pDCs infiltration in cancer is still largely controversial. In Primary Cutaneous Melanoma (PCM), pDCs density decreases over disease progression and collapses in metastatic melanoma (MM). Moreover, the residual circulating pDC compartment is defective in IFN-α production. Methods The activation of tumor-associated pDCs was evaluated by in silico and microscopic analysis. The expression of human myxovirus resistant protein 1 (MxA), as surrogate of IFN-α production, and proximity ligation assay (PLA) to test dsDNA-cGAS activation were performed on human melanoma biopsies. Moreover, IFN-α and CXCL10 production by in vitro stimulated (i.e. with R848, CpG-A, ADU-S100) pDCs exposed to melanoma cell lines supernatants (SN-mel) was tested by intracellular flow cytometry and ELISA. We also performed a bulk RNA-sequencing on SN-mel-exposed pDCs, resting or stimulated with R848. Glycolytic rate assay was performed on SN-mel-exposed pDCs using the Seahorse XFe24 Extracellular Flux Analyzer. Results Based on a set of microscopic, functional and in silico analyses, we demonstrated that the melanoma milieu directly impairs IFN-α and CXCL10 production by pDCs via TLR-7/9 and cGAS-STING signaling pathways. Melanoma-derived immunosuppressive cytokines and a metabolic drift represent relevant mechanisms enforcing pDC-mediated melanoma escape. Discussion These findings propose a new window of intervention for novel immunotherapy approaches to amplify the antitumor innate immune response in cutaneous melanoma (CM).
... The properties of pDCs in tumors were controversial. Many studies have suggested that PDCs may enhance an antitumor immune response by using TLR7 and TLR9 agonists [17][18][19]. However, others believe that pDCs may exhibit a pro-tumorigenic or immunoregulatory feature in tumor [20,21]. ...
Article
Full-text available
To the best of the author’s knowledge, studies of mature plasmacytoid dendritic cell proliferation associated with T lymphoblastic lymphoma were extremely rare in the literature. Here, we report a patient who underwent both mature plasmacytoid dendritic cell proliferation and T lymphoblastic lymphoma. With the findings of lymph node biopsy taken from the right cervical and inguinal regions, we identified eye-catching mature plasmacytoid dendritic cells that were considered to be responsible for this lesion at the beginning, until the immunostaining of Ki67 and TDT showed a small group of positive cells hiding in these plasmacytoid dendritic cells. A bone marrow biopsy was also performed on this patient. Microscopically, the hematopoietic tissue was almost completely replaced by lymphoblastoid cells with condensed chromatin, inconspicuous nucleoli and scanty cytoplasm, which were basically the same as those seen in the lymph nodes in morphology. However, there was no sign of plasmacytoid dendritic cells or Langerhans cells in the bone marrow biopsy. With the help of bone marrow biopsy, our final diagnosis of the lymph node was T lymphoblastic lymphoma coexisting with mature plasmacytoid dendritic cell proliferation. Although accumulations of plasmacytoid dendritic cells may occur in some infections or reactive lymphadenopathy, the presence of extensive nodules or infiltration of plasmacytoid dendritic cells strongly reminds the pathologist to carefully evaluate the bone marrow or peripheral blood status of the patient to exclude a hidden myeloid or other neoplasm.
... In addition, PDCs may infiltrate a variety of malignancies, such as melanoma, squamous cell carcinoma, basal cell carcinoma, and breast carcinoma. Many studies have suggested that PDCs may enhance an antitumor immune response by using TLR7 and TLR9 agonists [10][11][12]. ...
Preprint
Full-text available
To the best of the author’s knowledge, studies of mature plasmacytoid dendritic cell proliferation associated with T lymphoblastic lymphoma were extremely rare in the literature. Here, we report a patient who underwent both mature plasmacytoid dendritic cell proliferation and T lymphoblastic lymphoma. With the findings of lymph node biopsy taken from the right cervical and inguinal regions, we identified eye-catching mature plasmacytoid dendritic cells that were considered to be responsible for this lesion at the beginning, until the immunostaining of Ki67 and TDT showed a small group of positive cells hiding in these plasmacytoid dendritic cells. Bone marrow biopsy was also performed in this patient. Microscopically, the hematopoietic tissue was almost completely replaced by lymphoblastoid cells with condensed chromatin, inconspicuous nucleoli and scanty cytoplasm, which were basically the same as those seen in the lymph nodes in morphology. However, there was no sign of plasmacytoid dendritic cells or Langerhans cells in the bone marrow biopsy. With the help of bone marrow biopsy, our final diagnosis of the lymph node was T lymphoblastic lymphoma coexisting with mature plasmacytoid dendritic cell proliferation . Although accumulations of plasmacytoid dendritic cells may occur in some infections or reactive lymphadenopathy, the presence of extensive nodules or infiltration of plasmacytoid dendritic cells strongly reminds the pathologist to carefully evaluate the bone marrow or peripheral blood status of the patient to exclude a hidden myeloid or other neoplasm.
Chapter
The immune system serves as the fundamental defense mechanism of the human body, playing a crucial role in preventing tumorigenesis and managing the pathological progression of cancer. Despite constant pressure from the immune system and the tumor microenvironment, cancer cells can transform into aggressive, abnormal malignant cells and develop immunity-suppressing capabilities, eventually leading to mutant progression or immune evasion. The immune system is a highly organized and intricate network of cells that is both dynamic and complex. Immune cells are motile and migrate to specific organs in a context-specific manner, where they must come into close proximity with other cells to exchange information and function appropriately. These interactions give rise to diverse immune responses in different situations. In cancer, the intra-organ movement or infiltration of active immune cells into the tumor tissue is a decisive factor that determines immune outcome (Gardner A, Ruffell B, Trends Immunol. 37:855–865, 2016; Kohli K et al., Cancer Gene Ther. 29:10–21, 2022). Over the past decade, significant progress has been made in cancer immunotherapy, which has successfully cured certain types of cancer in some patients across several different cancer types. A key factor in this success is understanding the mechanisms of immune modulation and effectively directing cytotoxic T cell responses against tumors.
Article
Full-text available
Plasmacytoid dendritic cells (pDCs) are a pioneer cell type that produces type I interferon (IFN-I) and promotes antiviral immune responses. However, they are tolerogenic and, when recruited to the tumor microenvironment (TME), play complex roles that have long been a research focus. The interactions between pDCs and other components of the TME, whether direct or indirect, can either promote or hinder tumor development; consequently, pDCs are an intriguing target for therapeutic intervention. This review provides a comprehensive overview of pDC crosstalk in the TME, including crosstalk with various cell types, biochemical factors, and microorganisms. An in-depth understanding of pDC crosstalk in TME should facilitate the development of novel pDC-based therapeutic methods.
Article
The important role of the immune system in the surveillance and control of keratinocyte cancers (KCs), namely squamous and basal cell carcinomas, is increasingly appreciated, as new immunotherapies have recently become available. As the field of immunotherapy is rapidly evolving, this review synthesizes key concepts and highlights important cellular components within the immune system responsible for attacking KCs. We review the most current data on the epidemiology, risk factors, and immunotherapy management for KCs. Patients will seek advice from dermatologists to help explain why immunotherapies work for KCs and whether they might be appropriate for different clinical scenarios. Collaboration with medical colleagues across different disciplines to evaluate KCs for response to immunotherapy and early recognition of immune-related adverse events will help to optimize patient outcomes.
Article
Full-text available
Dendritic cells (DCs) are characterized by their unique capacity for primary T cell activation, providing the opportunity for DC-based cancer vaccination protocols. Novel findings reveal that besides their role as potent inducers of tumor-specific T cells, human DCs display additional antitumor effects. Most of these data were obtained with monocyte-derived DCs, whereas studies investigating native blood DCs are limited. In the present study, we analyze the tumoricidal capacity of M-DC8(+) DCs, which represent a major subpopulation of human blood DCs. We demonstrate that IFN-gamma-stimulated M-DC8(+) DCs lyse different tumor cell lines but not normal cells. In addition, we show that tumor cells markedly enhance the production of TNF-alpha by M-DC8(+) DCs via cell-to-cell contact and that this molecule essentially contributes to the killing activity of M-DC8(+) DCs. Furthermore, we illustrate the ability of M-DC8(+) DCs to promote proliferation, IFN-gamma production, and tumor-directed cytotoxicity of NK cells. The M-DC8(+) DC-mediated enhancement of the tumoricidal potential of NK cells is mainly dependent on cell-to-cell contact. These results reveal that, in addition to their crucial role in activating tumor-specific T cells, blood DCs exhibit direct tumor cell killing and enhance the tumoricidal activity of NK cells. These findings point to the pivotal role of DCs in triggering innate and adaptive immune responses against tumors.
Article
Full-text available
Toll-like receptors (TLRs) are ancient microbial pattern recognition receptors highly conserved from Drosophila to humans. To investigate if subsets of human dendritic cell precursors (pre-DC), including monocytes (pre-DC1), plasmacytoid DC precursors (pre-DC2), and CD11c+ immature DCs (imDCs) are developed to recognize different microbes or microbial antigens, we studied their TLR expression and responses to microbial antigens. We demonstrate that whereas monocytes preferentially express TLR 1, 2, 4, 5, and 8, plasmacytoid pre-DC strongly express TLR 7 and 9. In accordance with these TLR expression profiles, monocytes respond to the known microbial ligands for TLR2 (peptidoglycan [PGN], lipoteichoic acid) and TLR4 (lipopolysaccharide), by producing tumor necrosis factor (TNF)-α and interleukin (IL)-6. In contrast, plasmacytoid pre-DCs only respond to the microbial TLR9-ligand, CpG-ODNs (oligodeoxynucleotides [ODNs] containing unmethylated CpG motifs), by producing IFN-α. CD11c+ imDCs preferentially express TLR 1, 2, and 3 and respond to TLR 2-ligand PGN by producing large amounts of TNF-α, and to viral double-stranded RNA-like molecule poly I:C, by producing IFN-α and IL-12. The expression of distinct sets of TLRs and the corresponding difference in reactivity to microbial molecules among subsets of pre-DCs and imDCs support the concept that they have developed through distinct evolutionary pathways to recognize different microbial antigens.
Article
We examined the effects of interferon-alpha (IFN-α) treatment on the growth, cell cycle, proliferation, and apoptotic parameters as well as adhesive properties and proteome of chronic myelogenous leukemia (CML)-derived K562 cells. IFN-α treatment (200 to 600 U/ml, 24 to 72 h) suppressed growth and caused accumulation of K562 cells in the S-phase of cell cycle (increase in S-phase cells by up to 52% in comparison with the untreated controls) at the expenses of cells in G1-phase. No transition of cells to G0-phase occurred as followed from Ki-67 protein determination. Although the level of chimeric gene product, BCR-ABL mRNA coding for BCR-ABL protein with anti-apoptotic properties, decreased by 30%, apoptosis was not triggered as judged from Annexin-V, APO2.7, and TUNEL assays. Adhesion of K562 cells to fibronectin-coated surfaces increased by up to 52% as determined by calcein assay. The proteomic analysis (2-D electrophoresis in combination with mass spectrometry, MALDI-MS) revealed a single protein, ubiquitine cross-reactive protein (UBCR), whose level markedly increased due to IFN-α treatment. The ubiquitination-like directed degradation processes may thus play a role in the mechanism of IFN-α antiproliferative effects.
Article
Recent reports indicate that cytotoxic T cells are critically involved in contact hypersensitivity reactions in animals. In this study we sought to investigate the in vivo expression of cytotoxic granule proteins in the elicitation phase of allergic contact dermatitis in humans. Skin biopsy specimens were obtained from patients with allergic contact dermatitis (n = 8) and psoriasis (n = 6) and from controls with normal skin (n = 6). Expression of perforin and granzyme B was investigated by in situ hybridization and immunohistochemistry. In contrast to normal skin and psoriasis, a significant enhancement of perforin and granzyme B gene expression and immunoreactivity was observed in the mononuclear cell infiltrate of allergic contact dermatitis. Immunoreactivity for perforin and granzyme B was mainly found in the cytoplasm of lymphocytic cells, which were located in the dense perivascular infiltrate as well as at sites of marked spongiosis in the epidermis. Double immunostaining revealed that both CD4+ and CD8+ T cells are capable of expressing perforin and granzyme B. In conclusion, our data suggest that T-cell-mediated mechanisms involving cytotoxic granule proteins may elicit epidermal cell injury in vivo and thereby strongly contribute to the development of allergic contact dermatitis in humans.
Article
TRAIL induces apoptosis through two closely related receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5). Here we show that TRAIL-R1 can associate with TRAIL-R2, suggesting that TRAIL may signal through heteroreceptor signaling complexes. Both TRAIL receptors bind the adaptor molecules FADD and TRADD, and both death signals are interrupted by a dominant negative form of FADD and by the FLICE-inhibitory protein FLIP. The recruitment of TRADD may explain the potent activation of NF-κB observed by TRAIL receptors. Thus, TRAIL receptors can signal both death and gene transcription, functions reminiscent of those of TNFR1 and TRAMP, two other members of the death receptor family.
Article
The low-molecular-weight imidazoquinolinamine derivative, 1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine (imiquimod, previously described as R-837), induced alpha-interferon (IFN-alpha) in mice. IFN induction was identified at oral doses as low as 3 mg/kg. The 10% lethal dose for daily treatment with imiquimod was 200 mg/kg. Oral treatment with 30 mg/kg imiquimod once every three days significantly inhibited MC-26 colon carcinoma. Delay of treatment from day 1 to day 5, when tumors were easily palpable, did not reduce benefits. Ten daily treatments were slightly more effective than five. However, delivery of the same total dose of imiquimod either once every day for 20 days, once every 4 days, once every 7 days, or once every 10 days inhibited tumor growth to the same level. The antitumor effects of imiquimod were significantly abrogated by an antiserum to murine IFN-alpha, suggesting that the antitumor effect was to a substantial extent mediated by IFN induction. Imiquimod also significantly reduced the number of lung colonies in mice inoculated i.v. with MC-26 tumor cells. Combination of treatment with imiquimod and cyclophosphamide was significantly (P less than 0.01) better than treatment with either drug alone. Combination treatment with cyclophosphamide led to cures in some of the mice inoculated either s.c. or i.v. with MC-26 cells. Treatment with imiquimod also inhibited the growth of RIF-1 sarcoma and Lewis lung carcinoma but was ineffective for P388 leukemia. Imiquimod is an oral IFN-alpha inducer with antitumor effectiveness for transplantable murine tumors.
Article
The low-molecular-weight immunomodulator drug candidate, imiquimod (R-837), and its hydroxylated metabolite R-842, induce interferon-alpha (IFN-alpha) in human blood cells in vitro when tested in concentrations of 0.5 microgram/ml or more. The amounts of IFN-alpha found increased with time from 2-6 h of incubation up to 24-48 h, and were dependent on cell number and drug concentration. These two chemicals yielded more IFN-alpha in human peripheral blood mononuclear cell (PBMC) cultures than other known inducers tested in parallel. They also induced detectable amounts of interleukin (IL)-1, IL-6, IL-8, and tumor necrosis factor-alpha in human PBMC cultures in vitro.
Article
Perforin- and Fas-based cytolytic pathways are two major mechanisms of cell-mediated cytotoxicity. Recently, we have shown that an inhibitor of vacuolar type H+-ATPase, concanamycin A (CMA), inhibits perforin-based cytotoxic activity, mostly due to accelerated degradation of perforin by an increase in the pH of lytic granules. Here we show that CMA failed to inhibit the cytolytic activity of CD4+ CTL clone and perforin-deficient CD8+ CTL clone, which exclusively mediate Fas-based cytotoxicity, although CMA inhibited acidification and induced drastic vacuolation of cytoplasmic granules in these clones. In a wide range of alloantigen-specific CTL, a significant amount of the lysis of Con A blasts from normal mice and of Fas-positive tumor cells remained unaffected even in excess concentrations of CMA. However, CMA almost completely inhibited the lysis of Con A blasts from lpr mice and of Fas low expressing or negative tumor cells. Cytolysis by alloantigen-specific CD8+ CTL derived from gld mice was completely prevented by CMA. Furthermore, CMA-insensitive cytolysis exerted by CD8+ CTL clone was completely inhibitable by soluble Fas molecules. Thus, these data clearly indicate not only that CMA-insensitive cytolysis mediated by alloantigen-specific CTL is Fas dependent, but also that CMA is a selective inhibitor to block only the perforin-based killing pathway. In contrast, brefeldin A blocked the Fas-based cytotoxicity, but only marginally reduced the perforin-based cytotoxicity. Moreover, CMA and brefeldin A in combination completely abrogated all cytolytic activity of alloantigen-specific CTL. Taken together, these results reveal that CTL mainly exert perforin-based cytotoxicity and complementary Fas-based cytotoxicity, and that CMA is a powerful tool to clarify the contributions of the two distinct cytolytic pathways.
Article
A time-resolved fluorometric assay for the measurement of natural killer cell activity against target cells labelled with the acetoxymethyl ester of the fluorescence enhancing ligand 2,2':6',2"-terpyridine-6,6"-dicarboxylic acid (TDA) is described. The hydrophobic esterified form of TDA (bis(acetoxymethyl) 2,2':6',2"-terpyridine-6,6"-dicarboxylate, BATDA) diffuses readily through the cell membrane of viable cells. BATDA is hydrolysed by intracellular esterases resulting in accumulation of membrane impermeable TDA inside the target cells. After incubation of labelled K-562 cells with effector cells the TDA released from lysed cells into the supernatant is chelated with Eu3+. The natural killer cell activity is then quantified by measuring the intense fluorescence of the EuTDA chelates formed. Target cells are rapidly labelled when incubated with BATDA, TDA is released from target cells faster than 51Cr, the spontaneous release permits a short-term release assay to be set up and the detection of EuTDA is fast (5 min/96 well plate). Furthermore, this non-radioactive method permits the use of complex culture media since, in contrast to methods based on prompt fluorometry, the problem with autofluorescence can be avoided by the use of time-resolved fluorometry.