ArticlePDF Available

Abstract and Figures

The human breast tumor microenvironment can display features of T helper type 2 (Th2) inflammation, and Th2 inflammation can promote tumor development. However, the molecular and cellular mechanisms contributing to Th2 inflammation in breast tumors remain unclear. Here, we show that human breast cancer cells produce thymic stromal lymphopoietin (TSLP). Breast tumor supernatants, in a TSLP-dependent manner, induce expression of OX40L on dendritic cells (DCs). OX40L(+) DCs are found in primary breast tumor infiltrates. OX40L(+) DCs drive development of inflammatory Th2 cells producing interleukin-13 and tumor necrosis factor in vitro. Antibodies neutralizing TSLP or OX40L inhibit breast tumor growth and interleukin-13 production in a xenograft model. Thus, breast cancer cell-derived TSLP contributes to the inflammatory Th2 microenvironment conducive to breast tumor development by inducing OX40L expression on DCs.
Inflammatory Th2 in breast cancer immune environment. (A) Cytokine profiles as determined by Luminex in supernatants of human breast tumor fragments stimulated for 16 h with PMA and ionomycin. Numbers on the x-axis indicate the number of tissue samples from different patients tested. (B) Cytokine profiles as determined by Luminex in supernatants of tumor fragments (T) and surrounding tissue (ST) from the same patient after PMA and ionomycin stimulation. Cytokine concentration values of IL-2, IL-4, and IL-13 from T and ST samples were plotted and analyzed using linear regression to determine the level of correlation between cytokine concentration in T and ST samples. (C) Cytokine profiles as determined by Luminex in supernatants of tumor fragments after PMA and ionomycin stimulation. Cytokine concentration values of TNF and IL-13 and of TNF and IL-4 were plotted and analyzed using nonparametric Spearman correlation to determine the level of correlation of two cytokines concentration in tumor samples. (D, top) Single-cell suspensions from tumor samples were stimulated for 5 h with PMA and ionomycin. Cytokine production was measured by flow cytometry. Dot plots are gated on CD3⁺CD4⁺ T cells. (top right dot plot) Blue indicates gate on CD3⁺CD4⁺IL-13⁺ T cells that coexpress IFN-γ and TNF. Representative of four different patients from whom we have been able to obtain sufficient numbers of cells for 10-color analysis (patient nos. 148, 155, 164, and 169). Bottom, percentages of CD4⁺ T cells expressing IL-4 and IL-13 in tumor infiltrates and surrounding tissue (ST) were analyzed by flow cytometry. Dotted lines indicate paired samples from the same patient (n = 7, Wilcoxon matched-pairs ranked test). Single points indicate the percentage of cytokine expressing cells in tumor samples analyzed by flow cytometry for which we did not obtain sufficient number of cells from surrounding tissue to allow the analysis. (E) Frozen tissue sections from the same patient as in D were analyzed by immunofluorescence. Triple staining with anti-CD3-FITC (green), anti–IL-13–Texas red (red), and DAPI nuclear staining (blue). Bar, 90 µm.
… 
OX40L in breast cancer immune environment. (A) Immunofluorescence of primary breast tumor with indicated antibodies. Bar, 180 µm. Representative of 57/60 tumors analyzed. (B) Flow cytometry analysis of single-cell suspensions of primary breast tumors and surrounding tissue. Dot plots are gated on CD14neg nonlymphocytes. OX40L expression is analyzed on HLA-DRhighCD11chigh DCs. Graph summarizes percentages of OX40L-expressing DCs in tumor infiltrates and surrounding tissue (ST) analyzed by flow cytometry. Dotted lines indicate paired samples from the same patient (Wilcoxon matched-pairs ranked test). Single points indicate the percentage of OX40L⁺ DCs in tumor samples for which we did not obtain sufficient number of cells from surrounding tissue to allow the analysis. (C and D) mDCs were exposed to media alone, to supernatant of breast cancer cell lines (1806 or Hs587T), or to sonicate of primary breast cancer tissue from patients (tumor 43). OX40L and CD83 were measured by flow cytometry. FMO, fluorescence minus one indicates controls where one staining fluorescence is omitted to set negative gate. (E and F) mDCs were exposed for 48 h to supernatants of breast cancer cells Hs578T, and then co-cultured with allogeneic naive CD4⁺ T cells in the presence of 40 µg/ml of anti-OX40L (Ik-5 clone) or isotype control antibody. After 1 wk, cells were collected and restimulated for 5 h with PMA/ionomycin for intracellular cytokine staining. Data in E are representative of four experiments. (F) Summary of the effect of blocking OX40L during T cell stimulation by tumor-activated DCs. Graph shows the proportion of IL-13–secreting cells induced by DCs activated with supernatants from breast cancer cell line Hs578T (left) or primary breast tumors (right, T15, T29, and T53).
… 
TSLP in breast cancer environment. (A) Luminex analysis of TSLP in supernatants of breast cancer cell lines after 24 h of culture in the presence of PMA and ionomycin. (B) Luminex analysis of TSLP levels in supernatants of primary breast tumors (from 44 patients) activated with PMA and ionomycin. (C) NOD/SCID/β2m−/− mice were irradiated the day before tumor implantation and 10 × 10⁶ MDA-MB-231 cells were implanted by subcutaneous injection. Tumors were harvested at 4 wk after implant. Frozen tissue sections were analyzed by immunofluorescence for expression of TSLP (red). Actively dividing cells were identified by expression of Ki67 (green). Bar, 45 µm. (D and E) Frozen tissue sections from primary breast tumors from patients (38 patient samples) were analyzed by immunofluorescence for expression of TSLP. Tissues were also stained for the expression of IL-13 and cytokeratin 19, as indicated, to confirm TSLP expression by cancer cells. Staining pattern is representative of 35 out of 38 analyzed tumor samples from different patients. Bars: (D) 180 µm; (E) 15 µm. (F) NOD/SCID/β2m−/− mice were sublethally irradiated and transplanted with human CD34⁺ HPCs by intravenous injection. 4 wk after HPC transplant, 5 × 10⁶ MDA-MB-231 breast cancer cells were implanted subcutaneously. Tumors at the site of implantation, as well as lungs and kidneys, were harvested at 3 mo after implant. Frozen tissue sections were analyzed by immunofluorescence for expression of TSLP (green) and cytokeratin (red). Staining pattern is representative of tumors from three different mice. Bar, 90 µm.
… 
This content is subject to copyright.
Article
The Rockefeller University Press $30.00
J. Exp. Med. Vol. 208 No. 3 479-490
www.jem.org/cgi/doi/10.1084/jem.20102131
479
There is accumulating evidence that inamma-
tion plays a key role in the initiation and pro-
gression of cancer (Grivennikov et al., 2010).
There are two types of inammation that have
opposing eects on tumors: (a) chronic inam-
mation, which promotes cancer cell survival
and metastasis (Coussens and Werb, 2002;
Condeelis and Pollard, 2006; Mantovani et al.,
2008), and (b) acute inammation, which can
trigger cancer cell destruction as illustrated by
regressions of bladder cancer after treatment
with microbial preparations (Rako-Nahoum
and Medzhitov, 2009). Although chronic in-
ammation is often linked with the presence of
type 2–polarized macrophages (M2), acute in-
ammation associated with cancer destruction
is linked with type 1–polarized macrophages
(M1). M1 macrophages are induced by the type
1 cytokine IFN-, whereas, M2 macrophages
are induced by the type 2 cytokines IL-4 and
IL-13 (Mantovani and Sica, 2010).
Type 2 cytokines can contribute to tumori-
genesis in several ways. For example, IL-13 pro-
duced by NKT cells induces myeloid cells to
CORRESPONDENCE
A. Karolina Palucka:
karolinp@baylorhealth.edu
Abbreviations used: HPC,
hematopoietic progenitor cell;
mDC, myeloid DC; NOD/
SCID/2m/, nonobese
diabetic/LtSz-scid/scid 2
microglobulin–decient; TSLP,
thymic stromal lymphopoietin.
A. Pedroza-Gonzalez’s present address is Dept. of
Gastroenterology and Hepatology, Erasmus MC,
Rotterdam, Netherlands.
T.-C. Wu’s present address is College of Life Sciences,
Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan.
C. Aspord’s present address is Institut National de la Santé
et de la Recherche Médicale U823, Immunobiology and
Immunotherapy of Cancers, 38701, La Tronche, France.
J. Banchereau’s present address is Homan-La Roche, Inc.,
Nutley, NJ.
Thymic stromal lymphopoietin fosters
human breast tumor growth by promoting
type 2 inammation
Alexander Pedroza-Gonzalez,1 Kangling Xu,1,2 Te-Chia Wu,1,2
Caroline Aspord,1 Sasha Tindle,1 Florentina Marches,1 Michael Gallegos,1
Elizabeth C. Burton,4 Daniel Savino,4 Toshiyuki Hori,5 Yuetsu Tanaka,6
Sandra Zurawski,1 Gerard Zurawski,1 Laura Bover,7 Yong-Jun Liu,7
Jacques Banchereau,1,8,9 and A. Karolina Palucka1,3,8,9
1Baylor Institute for Immunology Research, Baylor Research Institute, Dallas, TX 75204
2Department of Biomedical Studies, Baylor University, Waco, TX 76706
3Sammons Cancer Center, 4Baylor University Medical Center, Dallas, TX 75246
5Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Sakyoku, Kyoto 606-8507, Japan
6Department of Immunology, University of the Ryukyus, Okinawa 903-0215, Japan
7MD Anderson Cancer Center, Houston, TX 77030
8Department of Gene and Cell Medicine and 9Department of Medicine, Immunology Institute, Mount Sinai School of Medicine,
New York, NY 10029
The human breast tumor microenvironment can display features of T helper type 2 (Th2)
inammation, and Th2 inammation can promote tumor development. However, the
molecular and cellular mechanisms contributing to Th2 inammation in breast tumors
remain unclear. Here, we show that human breast cancer cells produce thymic stromal
lymphopoietin (TSLP). Breast tumor supernatants, in a TSLP-dependent manner, induce
expression of OX40L on dendritic cells (DCs). OX40L+ DCs are found in primary breast
tumor inltrates. OX40L+ DCs drive development of inammatory Th2 cells producing
interleukin-13 and tumor necrosis factor in vitro. Antibodies neutralizing TSLP or OX40L
inhibit breast tumor growth and interleukin-13 production in a xenograft model. Thus,
breast cancer cell–derived TSLP contributes to the inammatory Th2 microenvironment
conducive to breast tumor development by inducing OX40L expression on DCs.
© 2011 Pedroza-Gonzalez et al. This article is distributed under the terms of
an Attribution–Noncommercial–Share Alike–No Mirror Sites license for the
rst six months after the publication date (see http://www.rupress.org/terms).
After six months it is available under a Creative Commons License (Attribution–
Noncommercial–Share Alike 3.0 Unported license, as described at http://creative-
commons.org/licenses/by-nc-sa/3.0/).
The Journal of Experimental Medicine
480 TSLP–OX40L–IL-13 axis in human breast cancer | Pedroza-Gonzalez et al.
>410,000 will die from the disease (Coughlin and Ekwueme,
2009). Here, we show that inammatory Th2 cells that promote
tumor development are driven by breast cancer–derived TSLP,
which induces and maintains OX40L-expressing DCs in the
tumor microenvironment. Thus, TSLP, and down-stream mole-
cules, might represent novel potential therapeutic targets.
RESULTS
Inammatory Th2 cells in primary breast cancer tumors
Our earlier study using a pilot cohort of 19 samples of pri-
mary breast cancer tumors revealed the secretion, upon acti-
vation with PMA and ionomycin, of both type 1 (IFN-) and
type 2 (IL-4 and IL-13) cytokines (Aspord et al., 2007). The
current study extends the analysis to a total of 99 consecutive
samples (Table S1). Supernatants of activated tumor fragments
display high levels of IFN-, IL-2, IL-4, IL-13, and TNF
(Fig. 1 A and Table S1). Supernatants from tumor sites
contained signicantly higher levels of IL-2, type 2 (IL-4 and
IL-13), and inammatory (TNF) cytokines than those from
macroscopically uninvolved surrounding tissue (Fig. 1 B and
Table S1). Whereas a signicant correlation can be observed
between IL-2 levels in tumor and adjacent tissue (P = 0.02),
IL-4 and IL-13 levels are not correlated (P = 0.5), further
suggesting polarization of cytokine environment in breast
tumors (Fig. 1 B). IFN- did not correlate with other cyto-
kine levels (Table S1). However, levels of TNF were correlated
with those of IL-13 (P < 0.0001; r = 0.62; n = 98) and IL-4
(P = 0.0175; r = 0.31; n = 59; Fig. 1 C). Thus, this survey of
cytokine expression suggested Th2 polarization in the breast
tumor microenvironment.
To identify the cells producing these cytokines, single-cell
suspensions were prepared from tumors; activated for 5 h with
PMA and ionomycin; stained with antibodies against T cells
and cytokines; and analyzed by ow cytometry. Gated viable
CD4+CD3+ T cells expressed IL-13 (3.67%), most of them
coexpressing IFN- and TNF (Fig. 1 D and Fig. S1 A).
A small fraction of IL-13+CD4+ T cells coexpressed IL-4, but
none expressed IL-10 (Fig. 1 D). Such T cells have been re-
ferred to as inammatory Th2 cells that are involved in aller-
gic inammatory diseases (Liu et al., 2007). Flow cytometry
analysis of consecutive tumor inltrates (n = 22) shows the
overall increased percentages of IL-4– and IL-13–secreting
CD4+CD3+ T cells (P = 0.0313 and P = 0.0156, respectively)
when compared with adjacent tissue samples (Fig. 1 D and
Fig. S1 B). Thus, the dierence between tumor tissue and ad-
jacent tissue appears to be caused by both the increased num-
bers of inltrating T cells and enhanced polarization. The
analysis of frozen tissue sections further demonstrated that in-
ltrating T cells in primary breast cancer tumors express IL-13
(Fig. 1 E). Thus, breast cancer tumors are inltrated with in-
ammatory Th2 cells.
DCs inltrating breast cancer tumors express OX40 ligand
Because OX40 ligation drives the dierentiation of CD4+
T cells into inammatory Th2 (Ito et al., 2005), we analyzed
the presence of OX40L in primary breast cancer tumors.
make TGF-, which ultimately inhibits CTL functions
(Berzofsky and Terabe, 2008). Spontaneous autochthonous
breast carcinomas arising in Her-2/neu transgenic mice ap-
pear more quickly when the mice are depleted of T cells,
which is evidence of T cell–mediated immunosurveillance
slowing tumor growth (Park et al., 2008). This immuno-
surveillance could be further enhanced by blockade of IL-13,
which slowed the appearance of these autologous tumors
compared with control antibody-treated mice (Park et al.,
2008). A spontaneous mouse breast cancer model recently
highlighted the role of Th2 cells which facilitate the develop-
ment of lung metastasis through macrophage activation
(DeNardo et al., 2009). We identied CD4+ T cells secreting
IFN- and IL-13 in breast cancer tumors (Aspord et al.,
2007). We also found that breast cancer cells express IL-13 on
cell surface. Autocrine IL-13 has been shown to be important
in the pathophysiology of Hodgkin’s disease (Kapp et al.,
1999; Skinnider et al., 2001, 2002). IL-13 and IL-13R are fre-
quently expressed by Hodgkin’s and Reed-Sternberg cells
(Skinnider et al., 2001), and IL-13 stimulates their growth
(Kapp et al., 1999; Trieu et al., 2004). Similar to Hodgkin’s
cells (Skinnider et al., 2002), breast cancer cells express
pSTAT6 (Aspord et al., 2007), suggesting that IL-13 actually
delivers signals to cancer cells. However, the mechanisms un-
derlying the development of Th2 inammation in breast can-
cer are unknown.
Like many other features of the immune response, Th1/
Th2 polarization is regulated by DCs. In the steady state, non-
activated (immature) DCs present self-antigens to T cells,
which leads to tolerance (Hawiger et al., 2001; Steinman et al.,
2003). Once activated (mature), antigen-loaded DCs are
geared toward the launching of antigen-specic immunity
(Finkelman et al., 1996; Brimnes et al., 2003) leading to the
proliferation of T cells and their dierentiation into helper
and eector cells. DCs are composed of distinct subsets, in-
cluding myeloid DCs (mDCs) and plasmacytoid DCs (Caux
et al., 1997; Maldonado-López et al., 1999; Pulendran et al., 1999;
Luft et al., 2002; Dudziak et al., 2007; Klechevsky et al.,
2008). DCs are also endowed with functional plasticity, i.e.,
they respond dierentially to distinct activation signals
(Steinman and Banchereau, 2007). For example, IL-10–
polarized mDCs generate anergic CD8+ T cells that are un-
able to lyse tumors (Steinbrink et al., 1999), as well as CD4+
T cells with regulatory/suppressor function (Levings et al.,
2005). In contrast, thymic stromal lymphopoietin (TSLP)–
polarized mDCs are conditioned to express OX40 ligand
(OX40L) and to expand T cells producing type 2 cytokines
(Soumelis et al., 2002; Gilliet et al., 2003). Both the distinct DC
subsets and their distinct response to microenvironment contrib-
ute to the generation of unique adaptive immune responses.
Unraveling the mechanisms by which breast cancer po-
larizes the immune responses might oer novel therapeutic
options. This is important because despite declining mortality
rates, breast cancer ranks second among cancer-related deaths
in women. Worldwide, it is estimated that more than 1 million
women are diagnosed with breast cancer every year, and
JEM VOL. 208, March 14, 2011
Article
481
mean ± SE for surrounding tissue = 1.5 ± 0.8% [n = 7]
and for breast cancer tumors 11 ± 1.67% [n = 12], respec-
tively; Fig. 2 B). Thus, breast cancer tumors are inltrated
with OX40L+ mDCs.
Breast cancer tumors produce soluble factors that induce
functional OX40L expression on DCs
To identify the breast cancer tumor factors that induce
OX40L on mDCs, LINnegHLA-DR+ CD123 CD11c+
mDCs were sorted from blood of healthy volunteers and
Immunouorescence staining of frozen tissue sections of pri-
mary breast cancer tumors showed the expression, in 57 out
of 60 analyzed tumors, of OX40L by a majority of HLA-
DRhigh cells (Fig. 2 A). These OX40L+ cells are located in
peritumoral areas (Fig. 2 A). Flow cytometry analysis of
single-cell suspensions further conrmed the expression of
OX40L by a fraction of HLA-DRhigh CD14neg CD11chigh
mDCs (Fig. 2 B). Paired analysis demonstrated that the tumor
beds express higher percentages of OX40L+ mDCs than
the surrounding tissue (P = 0.0156; n = 7 paired samples;
Figure 1. Inammatory Th2 in breast cancer immune environment. (A) Cytokine proles as determined by Luminex in supernatants of human
breast tumor fragments stimulated for 16 h with PMA and ionomycin. Numbers on the x-axis indicate the number of tissue samples from different pa-
tients tested. (B) Cytokine proles as determined by Luminex in supernatants of tumor fragments (T) and surrounding tissue (ST) from the same patient
after PMA and ionomycin stimulation. Cytokine concentration values of IL-2, IL-4, and IL-13 from T and ST samples were plotted and analyzed using lin-
ear regression to determine the level of correlation between cytokine concentration in T and ST samples. (C) Cytokine proles as determined by Luminex
in supernatants of tumor fragments after PMA and ionomycin stimulation. Cytokine concentration values of TNF and IL-13 and of TNF and IL-4 were
plotted and analyzed using nonparametric Spearman correlation to determine the level of correlation of two cytokines concentration in tumor samples.
(D, top) Single-cell suspensions from tumor samples were stimulated for 5 h with PMA and ionomycin. Cytokine production was measured by ow
cytometry. Dot plots are gated on CD3+CD4+ T cells. (top right dot plot) Blue indicates gate on CD3+CD4+IL-13+ T cells that coexpress IFN- and TNF. Rep-
resentative of four different patients from whom we have been able to obtain sufcient numbers of cells for 10-color analysis (patient nos. 148, 155, 164,
and 169). Bottom, percentages of CD4+ T cells expressing IL-4 and IL-13 in tumor inltrates and surrounding tissue (ST) were analyzed by ow cytometry.
Dotted lines indicate paired samples from the same patient (n = 7, Wilcoxon matched-pairs ranked test). Single points indicate the percentage of cytokine
expressing cells in tumor samples analyzed by ow cytometry for which we did not obtain sufcient number of cells from surrounding tissue to allow the
analysis. (E) Frozen tissue sections from the same patient as in D were analyzed by immunouorescence. Triple staining with anti-CD3-FITC (green), anti–
IL-13–Texas red (red), and DAPI nuclear staining (blue). Bar, 90 µm.
482 TSLP–OX40L–IL-13 axis in human breast cancer | Pedroza-Gonzalez et al.
To test whether primary breast cancer tumors could also
regulate OX40L expression, fragments of primary tumors
were sonicated, centrifuged, ltered, and used in cultures with
blood mDCs. As illustrated in Fig. 2 D, mDCs acquired both
CD83 (a DC maturation marker) and OX40L. To determine
the impact of OX40L on the generation of inammatory Th2
responses in breast cancer, blood mDCs were rst exposed for
48 h to either TSLP or breast tumor–soluble fractions. Exposed
mDCs were then used to stimulate naive allogeneic CD4+
T cells with either the anti-OX40L antibody or a relevant
exposed to breast cancer supernatants. These were generated
from established breast cancer cell lines expanded in vitro
(Hs578T, MDA-MB-231, MDA-MB-468, MCF7, HCC-
1806, and T47D; Table S2) and breast cancer tumors estab-
lished in vivo by implanting breast cancer cell lines in
immunodecient mice (Aspord et al., 2007). As illustrated in
Fig. 2 C, mDCs exposed for 48 h to Hs578T and HCC-1806
supernatants expressed OX40L. Four of the ve breast cancer
cell lines, with the notable exception of T47D, induced
OX40L expression on mDCs (Fig. S1 C).
Figure 2. OX40L in breast cancer immune environment. (A) Immunouorescence of primary breast tumor with indicated antibodies. Bar, 180 µm.
Representative of 57/60 tumors analyzed. (B) Flow cytometry analysis of single-cell suspensions of primary breast tumors and surrounding tissue. Dot
plots are gated on CD14neg nonlymphocytes. OX40L expression is analyzed on HLA-DRhighCD11chigh DCs. Graph summarizes percentages of OX40L-expressing
DCs in tumor inltrates and surrounding tissue (ST) analyzed by ow cytometry. Dotted lines indicate paired samples from the same patient (Wilcoxon
matched-pairs ranked test). Single points indicate the percentage of OX40L+ DCs in tumor samples for which we did not obtain sufcient number of cells
from surrounding tissue to allow the analysis. (C and D) mDCs were exposed to media alone, to supernatant of breast cancer cell lines (1806 or Hs587T),
or to sonicate of primary breast cancer tissue from patients (tumor 43). OX40L and CD83 were measured by ow cytometry. FMO, uorescence minus one
indicates controls where one staining uorescence is omitted to set negative gate. (E and F) mDCs were exposed for 48 h to supernatants of breast cancer
cells Hs578T, and then co-cultured with allogeneic naive CD4+ T cells in the presence of 40 µg/ml of anti-OX40L (Ik-5 clone) or isotype control antibody.
After 1 wk, cells were collected and restimulated for 5 h with PMA/ionomycin for intracellular cytokine staining. Data in E are representative of four ex-
periments. (F) Summary of the effect of blocking OX40L during T cell stimulation by tumor-activated DCs. Graph shows the proportion of IL-13–secreting
cells induced by DCs activated with supernatants from breast cancer cell line Hs578T (left) or primary breast tumors (right, T15, T29, and T53).
JEM VOL. 208, March 14, 2011
Article
483
and secreted TSLP (Fig. 3 A). Supernatants of some pr imary
breast cancer tumors stimulated with PMA and ionomycin
displayed up to 300 pg/ml TSLP (Fig. 3 B). The expression of
TSLP by cancer cells was further analyzed using an anti-TSLP
antibody and immunouorescence of frozen breast cancer
tumors generated in the xenograft model (Aspord et al., 2007).
There, subcutaneous MDA-MB-231 tumors transplanted in
mice expressed TSLP (Fig. 3 C). The specicity of the staining
is demonstrated by pretreatment of the antibody with recom-
binant TSLP (Fig. S3).
Importantly, TSLP is expressed in 35 out of 38 analyzed
primary breast cancer tumors obtained from patients regard-
less of grade, histology, or stage of analyzed tumors. Fig. 3 D
illustrates the pattern of TSLP staining and coexpression with
isotype control. Blocking OX40L prevented the expansion of
IL13+CD4+ or TNF+CD4+ T cells by TSLP-primed mDCs
(>50% inhibition; Fig. S2 A), mDCs exposed to Hs578T
breast cancer cells (n = 4; median inhibition of IL13+CD4+
cells = 74%; range = 67–80%; Fig. 2, E and F), and mDCs ex-
posed to sonicates of randomly selected primary breast cancer
tumors (Fig. 2 F and Fig. S2 B). Thus, breast cancer cells pro-
duce factors that activate mDCs and induce them to express
OX40L and to elicit inammatory Th2 cells.
Breast cancer tumors express and secrete TSLP
OX40L can be induced on mDCs by TSLP, an IL-7–like cyto-
kine produced by epithelial cells (Liu et al., 2007; Ziegler
and Artis, 2010). All tested breast cancer cell lines expressed
Figure 3. TSLP in breast cancer environment. (A) Luminex analysis of TSLP in supernatants of breast cancer cell lines after 24 h of culture in the
presence of PMA and ionomycin. (B) Luminex analysis of TSLP levels in supernatants of primary breast tumors (from 44 patients) activated with PMA and
ionomycin. (C) NOD/SCID/2m/ mice were irradiated the day before tumor implantation and 10 × 106 MDA-MB-231 cells were implanted by subcuta-
neous injection. Tumors were harvested at 4 wk after implant. Frozen tissue sections were analyzed by immunouorescence for expression of TSLP (red).
Actively dividing cells were identied by expression of Ki67 (green). Bar, 45 µm. (D and E) Frozen tissue sections from primary breast tumors from patients
(38 patient samples) were analyzed by immunouorescence for expression of TSLP. Tissues were also stained for the expression of IL-13 and cytokeratin
19, as indicated, to conrm TSLP expression by cancer cells. Staining pattern is representative of 35 out of 38 analyzed tumor samples from different
patients. Bars: (D) 180 µm; (E) 15 µm. (F) NOD/SCID/2m/ mice were sublethally irradiated and transplanted with human CD34+ HPCs by intravenous
injection. 4 wk after HPC transplant, 5 × 106 MDA-MB-231 breast cancer cells were implanted subcutaneously. Tumors at the site of implantation, as well
as lungs and kidneys, were harvested at 3 mo after implant. Frozen tissue sections were analyzed by immunouorescence for expression of TSLP (green)
and cytokeratin (red). Staining pattern is representative of tumors from three different mice. Bar, 90 µm.
484 TSLP–OX40L–IL-13 axis in human breast cancer | Pedroza-Gonzalez et al.
is specic to epithelial cells as no staining can be found in
tumor-inltrating broblasts (Fig. S4 B). Furthermore, non-
malignant breast epithelia can also express TSLP (Fig. S4 C).
Thus, similar to normal skin or lung epithelium, breast
epithelial cells and breast cancer cells have the capacity to ex-
press, produce, and secrete TSLP.
cytokeratin 19–positive cells. It demonstrates that TSLP is ex-
pressed in the cytoplasm and the nucleus of breast cancer cells
that display IL-13 on their surface (Fig. 3 E). Importantly,
TSLP is also expressed in lung and kidney metastasis of MDA-
MB-231 tumors in humanized mice (Fig. 3 F) and in breast can-
cer tumor metastasis from patients (Fig. S4 A). TSLP expression
Figure 4. Blocking TSLP in vitro. (A) mDCs were incubated with supernatant of breast cancer cell line Hs578T in the presence or absence of 20 µg/ml
of anti-TSLP (AB 19024; rabbit IgG). OX40L expression was measured by ow cytometry after 48 h of incubation. (B) mDCs treated as in A were co-
cultured with naive allogeneic CD4+ T cells for 7 d. IL-13 production was measured by intracellular cytokine staining and ow cytometry after cells were
restimulated for 5 h with PMA and ionomycin. Data are representative of three experiments. (C) mDCs were incubated with soluble factors from sonicated
human breast tumors (T53, T60, and T97) in the presence or absence of 20 µg/ml of anti-TSLP (AB 19024; rabbit IgG). OX40L expression was measured by
ow cytometry after 48 h of incubation. (D) mDCs treated as in C were co-cultured with naive allogeneic CD4+ T cells for 7 d. IL-13 production was mea-
sured by intracellular cytokine staining and ow cytometry after cells were restimulated for 5 h with PMA and ionomycin. Representative of three pa-
tients tested. Blue dots represent IL-13+ T cells gated in the same sample. (E) Graph shows data from three independent experiments as described in A–D.
JEM VOL. 208, March 14, 2011
Article
485
whether this axis might actually contribute to breast cancer
tumor development. To address this question, humanized mice
were reconstituted with both Hs578T cells and T cells with
or without anti-OX40L–, anti-TSLPR–, and anti-TSLP–
neutralizing antibodies. As shown in Fig. 6 A, the administra-
tion of neutralizing anti-OX40L antibodies leads to signicant
inhibition of tumor development.
The administration of a neutralizing anti-TSLP antibody
also results in the inhibition of tumor development (Fig. 6 B).
TSLP blockade also leads to decreased secretion of IL-4 and
IL-13 by tumor inltrating T cells upon PMA and ionomycin
activation (Fig. 6 C). Finally, the administration of antibody
blocking TSLPR nearly completely blocks tumor develop-
ment driven by CD4+ T cells (Fig. 6 D). These results indicate
that the TSLP contributes to breast cancer pathogenesis.
DISCUSSION
Here, we show that breast cancer is inltrated with inamma-
tory Th2 cells and that such T cells are driven by OX40L on
DCs. Blocking OX40L in vitro prevents generation of these
CD4+ T cells without impact on IL-10–producing CD4+
T cells. Blocking OX40L in vivo partially prevents T cell–
dependent acceleration of breast cancer tumor development.
OX40L is not constitutively expressed, but can be induced on
DCs, macrophages, and B cells; e.g., upon CD40 engagement
or cytokine signals such as TSLP or IL-18, as well as upon
TLR stimulation (Ito et al., 2005; Croft et al., 2009). Thus, the
Anti-TSLP antibodies block the generation of inammatory
Th2 responses in vitro
To determine the impact of blocking TSLP on the generation
of inammatory Th2 responses in breast cancer, blood mDCs
were rst exposed for 48 h to either TSLP or breast tumor–
soluble fractions. Addition of anti-TSLP–neutralizing anti-
bodies to breast cancer tumor supernatants inhibited their
ability to induce OX40L on mDCs (Fig. 4 A). Such mDCs
displayed a diminished capacity to expand IL13+CD4+ or
TNF+CD4+ T cells (n = 3; median inhibition = 73%, range =
72–77%; Fig. 4, B and E). Likewise, adding anti-TSLP–
neutralizing antibody to sonicate of randomly selected primary
breast cancer tumors led to down-regulation of OX40L ex-
pression by mDCs (Fig. 4 C) and decreased expansion of
IL13+TNF+CD4+ T cells (Fig. 4, D and E). Finally, when
anti-TSLP receptor chain (TSLPR) antibody was added to
mDCs during their exposure to the supernatant of three dif-
ferent breast cancer cell lines (Hs578T, MDA-MB-231, and
MCF7; Fig. 5 A), resulting mDCs showed a much diminished
expansion of IL13+ CD4+ T cells (Fig. 5 B). Thus, TSLP is the
factor secreted by breast cancer cells that contributes to gen-
eration of inammatory Th2 responses.
Antibodies neutralizing TSLP–OX40L axis block tumor
development in vivo
Our results thus far suggest a role for the TSLP–OX40L axis
in generation of IL13+TNF+CD4+ T cells, but do not establish
Figure 5. Blocking TSLP-R in vitro. (A) mDCs were treated with anti-TSLP-R (clone AB81_85.1F11, mouse IgG1), media or control antibody during
activation with TSLP or with supernatant of one of the three different breast cancer cell lines (Hs578T, MDA-MB-231, and MCF7). mDCs were then co-
cultured with allogeneic naive CD4+ T cells. After 1 wk, cells were collected, restimulated for 5 h with PMA and ionomycin, and analyzed by ow cytom-
etry. (B) Analysis of different experiments showing the effect of blocking TSLP-R on the induction of IL-13 secreting cells as described in A.
486 TSLP–OX40L–IL-13 axis in human breast cancer | Pedroza-Gonzalez et al.
cells markedly in-
crease TSLP expres-
sion (Liu et al.,
2007). The TSLP-
activated DCs mi-
grate to the draining lymph nodes, prime CD4+ T cells via
OX40L to dierentiate into inammatory Th2 eector
and memory cells, and thus initiate the adaptive phase of
allergic immune responses. Interestingly, in breast cancer,
OX40L+ mDCs are present in the tumor. It remains to be
determined whether this reects their inability to migrate
from the tumor to draining lymph nodes. Indeed, such
an evasion mechanism has been documented recently in
human and mouse tumors showing an inhibition of DC
migration from tumors to tumor-draining lymph nodes
(Villablanca et al., 2010). This eect depends on tumor-
derived ligands of the liver X receptors (Villablanca et al.,
2010). It also remains to be determined whether these DCs
are able to prime Th2 immunity in situ in tertiary lymphoid
structures or whether their main role is to maintain the ac-
tivation and survival of Th2 cells at the tumor site. Their
ability to maintain Th2 cell phenotype and eector func-
tion is supported by our earlier studies showing that T cells
isolated from experimental breast tumors and transferred to
naive tumor-bearing humanized mice can promote tumor
development even at low numbers and upon single injec-
tion (Aspord et al., 2007).
presence of OX40L+ mDCs in breast tumors indicate sus-
tained activation of DCs in tumor environment. Indeed,
OX40L expression by DCs is driven by TSLP secreted from
breast cancer cells. Accordingly, TSLP expression can be found
in primary and metastatic tumors. Blocking TSLP reduces in-
ammation and partially inhibits tumor development.
Based on our results presented herein and in our earlier
studies, we propose a vicious circle of smoldering type 2 in-
ammation that perpetuates breast cancer and which is main-
tained by TSLP. There, breast cancer attracts DCs, possibly
through macrophage inammatory protein 3 (MIP3-;
Bell et al., 1999). Tumor-inltrating DCs are then exposed to
TSLP secreted by breast cancer cells, which triggers their
maturation and OX40L expression. This might explain the
aseptic mDC maturation that we found in breast cancer (Bell
et al., 1999; Aspord et al., 2007). OX40L+ mDCs induce
CD4+ T cells to secrete IL-13, as well as TNF. These inam-
matory CD4+ T cells contribute to tumor development in an
IL-13–dependent pathway (Aspord et al., 2007). Thus far,
TSLP represents the only factor that activates mDCs without
inducing them to produce Th1-polarizing cytokines (Liu
et al., 2007). Under normal physiological conditions, TSLP
appears to play a critical role in CD4+ T cell homeostasis in
the peripheral mucosa-associated lymphoid tissues and in the
positive selection and/or expansion of regulatory T (T reg)
cells in the thymus (Watanabe et al., 2005a,b). In inamma-
tory conditions such as atopic dermatitis and asthma, epithelial
Figure 6. Blocking OX40L-TSLP in vivo.
(A) NOD/SCID/2m/ mice were sublethally
irradiated and transplanted with human
CD34+ HPCs by intravenous injection. 4 wk
after HPC transplant, 10 × 106 Hs578T breast
cancer cells were implanted subcutaneously.
3, 6, and 9 d after, mice were reconstituted
with autologous total T cells together with
200 µg per injection of blocking anti-OX40L
or isotype control antibody (mouse Ig; red
arrows). PBS group was injected with tumor
cells but not with T cells (gray line). Tumor
size was measured at indicated times. Mean
values from three experiments representing
nine mice per each condition. Anti-OX40L and
isotype-treated cohorts were compared sta-
tistically. (B) NOD/SCID/2m/ mice were
irradiated and implanted with 10 × 106
Hs578T breast cancer cells together with 200 µg
per injection of neutralizing anti-TSLP
(rabbit), rabbit isotype control antibody, or
PBS. 3, 6, and 9 d after, mice were reconsti-
tuted with immature DCs and autologous
total T cells together with 200 µg per injection
of neutralizing anti-TSLP (rabbit), rabbit iso-
type control antibody or PBS. Representative
of three independent experiments with a total
of nine mice in TSLP blockade group. (C) Cyto-
kine secretion in single-cell suspensions from
tumors after 16-h restimulation with PMA
and ionomycin. (D) Same as in B, but mice
were injected with anti-TSLPR or isotype con-
trol at days 3, 6, and 9. Representative of two
independent experiments. n indicates number
of mice per cohort in this representative
experiment.
JEM VOL. 208, March 14, 2011
Article
487
Interestingly, studies in mice suggest the role of Th2 cyto-
kines in mammary gland development. Indeed, the dierenti-
ation of the luminal epithelial lineage appears to require
autocrine signaling by Th2 cytokines, as shown by reduced
dierentiation and alveolar morphogenesis in both Stat6 and
IL-4–/IL-13–decient mice during pregnancy (Khaled et al.,
2007). Yet, in the murine model of endogenous breast cancer
type 2, polarized CD4+ T cells are essential to the metastatic
process via secretion of IL-4, which induces macrophages to
secrete epidermal growth factor (DeNardo et al., 2009). Simi-
lar to endogenous mouse model, in our model of transplanted
metastatic human breast cancer IL-13 is derived from micro-
environment. IL-13 can exert pro-cancer activity in several
ways, including the triggering of TGF- secretion (Terabe
et al., 2000, 2003; Park et al., 2005; Shimamura et al., 2010).
Furthermore, IL-4 exposure of cancer cells leads to the up-
regulation of antiapoptotic pathways via mobilization of
STAT6 (Zhang et al., 2008). We have shown that STAT6 is
phosphorylated in primary breast cancer tumors (Aspord et al.,
2007). All these antiapoptotic pathways are likely to synergize
to promote the survival of cancer cells and facilitate metastasis.
Importantly, such a protective eect on cancer cell suscepti-
bility to apoptosis might increase their resistance to chemother-
apy (Todaro et al., 2008) and immune-mediated cytotoxicity
driven by Granzyme B (Sarin et al., 1997; Heibein et al.,
2000). Thus, the TSLP–OX40L–IL13 axis might oer a novel
therapeutic target.
MATERIALS AND METHODS
Isolation and culture of myeloid dendritic cells. DCs were pur ied
from buy coat of blood from healthy donors. In brief, DCs were enriched
from mononuclear cells by negative selection using a mixture of antibodies
against linage markers for CD3, CD14, CD16, CD19, CD56, and glycopho-
rin A (Dynabeads Human DC Enrichment kit; Invitrogen). Cells from nega-
tive fraction were immunolabeled with anti–human FITC-labeled lienage
cocktail (CD3, CD14, CD16, CD19, CD20, and CD56; BD); PE-labeled
CD123 (mIgG1; clone 9F5; BD), QR-labeled HLA-DR (mIgG2a; clone
HK14; Sigma-Aldrich) and APC-labeled CD11c (mIgG2b; clone S-HCL-3;
BD). DCs (lin, CD123, HLA-DR+, CD11c+) were sorted in a FACSAria
cytometer (BD). DCs were seeded at 100 × 103 cells/well in 200 µl of me-
dium (RPMI supplemented with 2 mM glutamine, 50 U/ml penicillin,
50 µg/ml streptomycin, 0.1 mM MEM nonessential amino acids, 10 mM
Hepes buer, 0.1 mM sodium pyruvate, and 10% of human AB serum). DCs
were cultured with medium alone or in the presence of 20 ng/ml of TSLP,
or dierent tumor derived products. After 48 h, DCs were harvested and
washed. The stimulated cells were stained for phenotype analysis or co-
cultured with allogeneic naive CD4+ T cells.
Immunouorescence. 6-µm-frozen sections from tissues were xed with
cold acetone for 5 min. The sections were labeled with 5 µg/ml of anti-
OX40L antibody (mouse IgG1, 8F4), followed by anti–mouse IgG conju-
gated to Texas red (Jackson ImmunoResearch Laboratories). For IL-13, tissue
was labeled with 10 µg/ml of anti–IL-13 (polyclonal goat IgG; AF-123-NA;
R&D System) followed with Texas red anti–goat IgG (Jackson Immuno-
Research Laboratories). TSLP was detected with 10 µg/ml of mouse anti-TSLP
antibody prepared in-house (mIgG1; clone 14C3.2E11). Cytokeratin 19 was
labeled with monoclonal antibody clone A53-BA2 (IgG2a; Abcam), followed
by Alexa Fluor 568 goat anti–mouse IgG2a (Invitrogen). The following di-
rect-labeled antibodies used were as follows: FITC anti–HLA-DR (mouse
IgG2a; L243; BD), FITC anti-CD11c (clone; KB 90; Dako), FITC anti-Ki67
Our results add another feature to the role of OX40L in
tumors. Indeed, several studies in mouse models of transplant-
able tumors suggested that engaging OX40 via an agonist anti-
body, OX40L.Fc, or transfected tumor cells and DCs appears
to promote antitumor eects (Weinberg et al., 2000; Morris
et al., 2001; Ali et al., 2004; Piconese et al., 2008). However,
Tnfsf4 (gene coding OX40L) is regulated by the mRNA
MIRN125B (Smirnov and Cheung, 2008), whose expression
is down-regulated in breast cancer (Iorio et al., 2005). Tnfsf4
is up-regulated in ataxia telangiectasia carriers and patients
(Smirnov and Cheung, 2008), who have been shown to have
an increased risk of breast cancer (Swift et al., 1987). Further-
more, a recent pilot study described the expression of OX40
and OX40L in >100 tissue samples from invasive ductal car-
cinomas, and suggested a possible association of OX40 ex-
pression and lymph node metastatic status (Xie et al., 2010).
OX40L signaling has several important features that might
help explain the results observed in our and other studies.
Thus, OX40L triggers Th2 polarization independent of IL-4,
promotes TNF production, and inhibits IL-10 production by
the developing Th2 cells, but only in the absence of IL-12. In
the presence of IL-12, OX40L signaling instead promotes the
development of Th1 cells that, like inammatory Th2 cells,
produce TNF but not IL-10 (Liu et al., 2007).
Interestingly, inammatory Th2 cells coexist within the
tumor immune environment alongside IL-10–secreting CD4+
T cells. Recent studies demonstrate a colocalization of cells
with the phenotype of T reg cells) with mature DCs in lym-
phoid inltrates in breast cancer (Gobert et al., 2009). A high
number of FoxP3+ T reg cells was associated with disease re-
lapse (Gobert et al., 2009). Thus, the niche nding and metas-
tasis formation might be facilitated by inammatory Th2
response, whereas in the established tumor a T reg cell re-
sponse might prevail. Interestingly, CCL22 (a macrophage-
derived chemokine) appears involved in the attraction of
T reg cells (Gobert et al., 2009), but also, in cooperation with
CCL17 (TARC), of inammatory Th2 cells (Liu et al., 2007).
How this ne balance between T reg cells and Th2 responses
is regulated will require further study.
Two key questions arise from our work: (1) what are the
mechanisms allowing TSLP release from cancer cells; and
(2) what is the impact of IL-13 (and IL-4) on cancer cells,
on the stroma, and on the immune inltrate? Our data show
that nonmalignant breast epithelia can express TSLP. This fur-
ther demonstrates that cancer cells can exploit pathways that
are present in the normal tissue to establish a microenviron-
ment facilitating tumor development. The mechanisms
regulating TSLP expression and secretion from cancer cells,
including a potential link with oncogenic events, remain to
be established. It will also be important to determine the
impact of TSLP and inammatory Th2 environment on the
stromal broblasts. Indeed, recent studies point to the critical
role of cross talk between cancer cells and broblasts in deter-
mining the type of microenvironment established by cancer
cells originating from dierent types of breast cancers (Camp
et al., 2011).
488 TSLP–OX40L–IL-13 axis in human breast cancer | Pedroza-Gonzalez et al.
followed by surface and intracellular staining. For blocking OX40L, tumor-
activated mDCs were co-cultured with naive CD4 T cells in the presence of
50 µg of anti-OX40L (Ik-5 clone) or control IgG2a isotype antibody. For
blocking TSLP, tumor-derived factors were preincubated with 20 µg/ml of
anti-TSLP antibody (rabbit; AB 19024) or normal rabbit IgG (R&D Sys-
tems) at room temperature for 30 min after DC activation. DCs were acti-
vated with the neutralized tumor-derived factors and nally co-cultured
with naive CD4 T cells for 7 d. For TSLPR blocking, DCs were preincubated
with anti-TSLP receptor antibody (clone AB81_85.1F11; mouse IgG1) for
3 min at room temperature.
Tumor-bear ing mice. Mice were humanized either by CD34+ hemato-
poietic progenitor cell (HPC) transplant or by co-administration of DCs and
T cells as described previously (Aspord et al., 2007; Institutional Animal Care
and Use Committee no. A01-005). CD34+HPCs were obtained from apher-
esis of adult healthy volunteers mobilized with G-CSF and puried as de-
scribed previously (Aspord et al., 2007). The CD34 fraction of apheresis was
Ficoll pur ied, and the PBMCs obtained were stored frozen and used as a
source of autologous T cells. 3 million CD34+ HPCs were transplanted intra-
venously into sublethally irradiated (12 cGy/g body weight of 137Cs irra-
diation) nonobese diabetic/LtSz-scid/scid 2 microglobulin–decient
(NOD/SCID/2m/) mice (Jackson ImmunoResearch Laboratories).
After 4 wk of engraftment, 10 million Hs578T breast cancer cells were har-
vested from cultures and injected subcutaneously into the anks of the mice.
Mice were reconstituted with 10 million CD4+ T cells and 10 million CD8+
T cells autologous to the grafted CD34+ HPCs. CD4+ and CD8+ T cells
were positively selected from thawed PBMCs using magnetic selection ac-
cording to the manufacturer’s instructions (Miltenyi Biotec). The purity was
routinely >90%. T cells were transferred at days 3, 6, and 9 after tumor im-
plantation. For experiments with NOD/SCID/2m/ mice, they were
sublethally irradiated the day before tumor implantation. Mice were then re-
constituted with 1 million monocyte-derived DCs (MDDCs) and autolo-
gous T cells as described in the previous paragraph. MDDCs were generated
from the adherent fraction of PBMCs by culturing with 100 ng/ml GM-
CSF (Immunex) and 10 ng/ml IL-4 (R&D Systems). Tumor size was moni-
tored every 2–3 d. Tumor volume (ellipsoid) was calculated as follows: ([short
diameter]2 × long diameter)/2.
Blocking in vivo experiments. For dierent experimental purposes,
tumor-bearing mice transferred with autologous T cells were injected intra-
tumorally with 200 µg of anti-OX40L (clone IK-5; mIgG2a) blocking anti-
body, 100 µg of anti-TSLP antibody (clone AB19024; rabbit IgG), 100 µg of
anti-IL-13 neutralizing antibody prepared in-house (clone 13G1.B2; mIgG1),
200 µg of anti-TSLPR neutralizing antibody (clone AB81_85.1F11; mouse
IgG1), or isotype control, respectively at days 3, 6, and 9 after tumor implan-
tation. For blocking TSLP, anti-TSLP antibody injection was also given at day
0 while the tumor was implanted.
Online supplemental material. Fig. S1 illustrates gating strategy for ow
cytometry analysis of tumor-inltrating lymphocytes and the accumulation
of lymphocytes in tumor beds as compared with surrounding tissue. Fig. S2
shows that mDCs exposed to TSLP or to soluble tumor factors in the pres-
ence of anti-OX40L are unable to drive the dierentiation of inammatory
Th2 cells. Fig. S3 shows the specicity of anti-TSLP antibody staining in fro-
zen tissue sections. Fig. S4 shows the expression of TSLP in breast cancer cells
and in nonmalignant breast tissue. Table S1 provides the pathological diagno-
sis information and Luminex data for cytokine levels of all 99 breast cancer
patients used for cytokine analysis. Table S2 describes the characteristics of
breast cancer cell lines studied. Online supplemental material is available at
http://www.jem.org/cg i/content/full/jem.20102131/DC1.
We are grateful to Albert Barnes, Sebastien Coquery, Elizabeth Kraus, Mark
Michnevitz, Gina Stella Garcia-Romo, Jenny Smith, and Lynnette Walters for help;
Dr. Joseph Fay for help with healthy volunteers; Cindy Samuelsen for continuous
support; and Drs. Sally M. Knox and Michael Grant at the Department of Surgery
(clone ki-67; Dako), and Alexa Fluor 488 anti-CD3 (mIgG1; UCHT1; BD).
Finally, sections were counterstained for 2 min with 3 µM of the nuclear stain
DAPI (in PBS; Invitrogen). To conr m specicity of TSLP staining, pr imary
anti-TSLP antibody was preincubated with 100 µg of recombinant human
TSLP (R&D Systems) for 30 min at room temperature before staining of
tissue sections that previously showed to be TSLP positive.
Flow cytometry analysis. Cell suspensions from human breast carcinoma
tissue and tumor or draining lymph nodes from humanized mice were used
for phenotypic characterization of leukocytes. Cell suspensions were ob-
tained by digestion with 2.5 mg/ml of collagenase D (Roche), and 200 U/ml
of DNase I (Sigma-Aldrich) for 30–60 min at 37°C. The anti–human anti-
bodies used were as follows: labeled FITC-labeled lineage cocktail (CD3,
CD14, CD16, CD19, CD20, and CD56; BD); PE-labeled OX40L (mIgG1;
clone Ik-1; BD); QR-labeled HLA-DR (mIgG2a; clone HK14; Sigma-
Aldrich); APC-labeled CD11c (mIgG2b; clone S-HCL-3; BD); PerCP-labeled
CD3 (mIgG1; clone SK7; BD); PECy7-labeled CD4 (mIgG1; clone SK3;
BD); APCCy7-labeled CD8 (mIgG1; clone SK1; BD); CFS-labeled IL-4
(mIgG1; clone 3007; R&D Systems); Pacic blue–labeled IL-10 (rat IgG1;
clone JES3-9D7; eBioscience); PE-labeled IL-13 (rat IgG1; clone JES10-5A2
BD); APC-labeled TNF (mIgG1; clone 6401.1111; BD); Alexa Fluor 700–
labeled IFN- (mIgG1; clone B27; BD). Cells were incubated with the
antibodies for 30 min at 4°C in the dark, and then washed three times and
xed with 1% paraformaldehyde to be analyzed in a FACSCalibur or LSR-II
cytometer (BD). For intracellular cytokines, cells were stained using BD
Cytox/Cytoperm xation/permeabilization kit according to the manufac-
turer’s directions.
Tumor factors preparation. Tumor factors were obtained from superna-
tant of established breast cancer cell lines cultured in vitro (Table S2; Soule
et al., 1973; Hackett et al., 1977; Lacroix and Leclercq, 2004; Neve et al.,
2006) or by sonication from tumor cell lines, human breast tumor tissue, or
tumors from humanized mice. In brief, cell lines were culture in medium
(RPMI supplemented with 2 mM glutamine, 50 U/ml penicillin, 50 µg/ml
streptomycin, 0.1 mM MEM nonessential amino acids, 10 mM Hepes buer,
0.1 mM sodium pyruvate, and 10% fetal calf serum), and when the cells
reached 90% of conuence fresh medium was added and the cells were left
in culture for an additional 48 h. For sonication, cells or tissues were placed
in PBS and disrupted for 30 s at 4°C, with the power output adjusted at 4.5
level of the 60 sonic dismembrator (Thermo Fisher Scientic). Cellular de-
bris was removed by centr ifugation, and the supernatant was collected and
stored at 80°C.
Cytokine analysis. Tumor samples from patients diagnosed with breast car-
cinoma (in situ, invasive duct, and/or mucinous carcinoma of the breast, as
well as lobular carcinoma) were obtained from the Baylor University Medical
Center Tissue Bank (Institutional Review Board no. 005–145). Tumors and
draining lymph nodes from mice implanted with breast cancer cell lines
H578T and MDA-MB-231 were also analyzed. Whole-tissue fragments (4 ×
4 × 4 mm, 0.02 g, approximately) were placed in culture medium with
50 ng/ml of PMA (Sigma-Aldrich) and 1 µg/ml of ionomycin (Sigma-Aldrich)
for 16 h. Cytokine production was analyzed in the culture super natant by
Luminex. For intracellular staining, cells were resuspended at a concentration
of 106 cells/ml in medium and activated for 5 h with PMA and ionomycin;
brefeldin A (GolgiPlug; BD) and monensin (GolgiStop; BD) were added for
the last 2.5 h.
DC–T cell co-cultures. Total CD4+ T cells were enriched from PBMCs of
healthy donors using magnetic depletion of other leukocytes (EasySep
Human CD4+ T Cell Enrichment kit; STEMCELL Technologies, Inc.).
Naive CD4+ T cells were sorted based on the expression of CD4+ CD27+ and
CD45RA+. Activated mDCs with medium, TSLP, or tumor-derived factors
were co-cultured with naive CD4+ T cells at a ratio of 1:5 for 7 d. After that,
the cells were washed and restimulated for 5 h with 50 ng/ml PMA and
1 µl/ml ionomycin. Brefeldin A and monensin were added for the last 2.5 h,
JEM VOL. 208, March 14, 2011
Article
489
TSLP and CD40L induce proallergic cytotoxic T cells. J. Exp. Med.
197:1059–1063. doi:10.1084/jem.20030240
Gobert, M., I. Treilleux, N. Bendriss-Vermare, T. Bachelot, S. Goddard-Leon,
V. Ar, C. Biota, A.C. Don, I. Durand, D. Olive, et al. 2009. Regulatory
T cells recruited through CCL22/CCR4 are selectively activated in
lymphoid inltrates surrounding primary breast tumors and lead to an
adverse clinical outcome. Cancer Res. 69:2000–2009. doi:10.1158/0008-
5472.CAN-08-2360
Grivennikov, S.I., F.R. Greten, and M. Karin. 2010. Immunity, inammation,
and cancer. Cell. 140:883–899. doi:10.1016/j.cell.2010.01.025
Hackett, A.J., H.S. Smith, E.L. Springer, R.B. Owens, W.A. Nelson-Rees,
J.L. Riggs, and M.B. Gardner. 1977. Two syngeneic cell lines from hu-
man breast tissue: the aneuploid mammary epithelial (Hs578T) and the
diploid myoepithelial (Hs578Bst) cell lines. J. Natl. Cancer Inst. 58:1795–
1806.
Hawiger, D., K. Inaba, Y. Dorsett, M. Guo, K. Mahnke, M. Rivera, J.V. Ravetch,
R.M. Steinman, and M.C. Nussenzweig. 2001. Dendritic cells induce
peripheral T cell unresponsiveness under steady state conditions in vivo.
J. Exp. Med. 194:769–779. doi:10.1084/jem.194.6.769
Heibein, J.A., I.S. Goping, M. Barry, M.J. Pinkoski, G.C. Shore, D.R. Green,
and R.C. Bleackley. 2000. Granzyme B-mediated cytochrome c release
is regulated by the Bcl-2 family members bid and Bax. J. Exp. Med.
192:1391–1402. doi:10.1084/jem.192.10.1391
Iorio, M.V., M. Ferracin, C.G. Liu, A. Veronese, R. Spizzo, S. Sabbioni, E.
Magri, M. Pedriali, M. Fabbri, M. Campiglio, et al. 2005. MicroRNA
gene expression deregulation in human breast cancer. Cancer Res.
65:7065–7070. doi:10.1158/0008-5472.CAN-05-1783
Ito, T., Y.H. Wang, O. Duramad, T. Hori, G.J. Delespesse, N. Watanabe, F.X.
Qin, Z. Yao, W. Cao, and Y.J. Liu. 2005. TSLP-activated dendritic cells
induce an inammatory T helper type 2 cell response through OX40
ligand. J. Exp. Med. 202:1213–1223. doi:10.1084/jem.20051135
Kapp, U., W.C. Yeh, B. Patterson, A.J. Elia, D. Kägi, A. Ho, A. Hessel, M.
Tipsword, A. Williams, C. Mirtsos, et al. 1999. Interleukin 13 is secreted
by and stimulates the growth of Hodgkin and Reed-Sternberg cells.
J. Exp. Med. 189:1939–1946. doi:10.1084/jem.189.12.1939
Khaled, W.T., E.K. Read, S.E. Nicholson, F.O. Baxter, A.J. Brennan, P.J. Came,
N. Sprigg, A.N. McKenzie, and C.J. Watson. 2007. The IL-4/IL-13/Stat6
signalling pathway promotes luminal mammary epithelial cell develop-
ment. Development. 134:2739–2750. doi:10.1242/dev.003194
Klechevsky, E., R. Morita, M. Liu, Y. Cao, S. Coquery, L. Thompson-Snipes, F.
Briere, D. Chaussabel, G. Zurawski, A.K. Palucka, et al. 2008. Functional
specializations of human epidermal Langerhans cells and CD14+
dermal dendritic cells. Immunity. 29:497–510. doi:10.1016/j.immuni
.2008.07.013
Lacroix, M., and G. Leclercq. 2004. Relevance of breast cancer cell lines as
models for breast tumours: an update. Breast Cancer Res. Treat. 83:249–
289. doi:10.1023/B:BREA.0000014042.54925.cc
Levings, M.K., S. Gregori, E. Tresoldi, S. Cazzaniga, C. Bonini, and M.G.
Roncarolo. 2005. Dierentiation of Tr1 cells by immature dendritic cells
requires IL-10 but not CD25+CD4+ Tr cells. Blood. 105:1162–1169.
doi:10.1182/blood-2004-03-1211
Liu, Y.J., V. Soumelis, N. Watanabe, T. Ito, Y.H. Wang, Rde.W. Malefyt, M.
Omori, B. Zhou, and S.F. Ziegler. 2007a. TSLP: an epithelial cell cyto-
kine that regulates T cell dierentiation by conditioning dendritic cell
maturation. Annu. Rev. Immunol. 25:193–219. doi:10.1146/annurev
.immunol.25.022106.141718
Luft, T., M. Jeord, P. Luetjens, T. Toy, H. Hochrein, K.A. Master man,
C. Maliszewski, K. Shortman, J. Cebon, and E. Maraskovsky. 2002.
Functionally distinct dendritic cell (DC) populations induced by physio-
logic stimuli: prostaglandin E(2) regulates the migratory capacity of specic
DC subsets. Blood. 100:1362–1372. doi:10.1182/blood-2001-12-0360
Maldonado-López, R., T. De Smedt, P. Michel, J. Godfroid, B. Pajak, C.
Heirman, K. Thielemans, O. Leo, J. Urbain, and M. Moser. 1999.
CD8alpha+ and CD8alpha- subclasses of dendritic cells direct the de-
velopment of distinct T helper cells in vivo. J. Exp. Med. 189:587–592.
doi:10.1084/jem.189.3.587
Mantovani, A., and A. Sica. 2010. Macrophages, innate immunity and can-
cer: balance, tolerance, and diversity. Curr. Opin. Immunol. 22:231–237.
doi:10.1016/j.coi.2010.01.009
and Ms. Dan Su at the Department of Pathology at Baylor University Medical
Center. We thank Dr. Michael Ramsay for continuous support.
This work was supported by the Baylor Health Care Systems Foundation, and the
National Institutes of Health (grants R0-1 CA89440 and R21 AI056001 to A. Karolina
Palucka; grants U19 AI057234, R0-1 CA78846, and CA85540 to J. Banchereau).
J. Banchereau holds the Caruth Chair for Transplantation Immunology Research.
A. Karolina Palucka holds the Ramsay Chair for Cancer Immunology Research.
The authors have no conicting nancial interests.
Submitted: 7 October 2010
Accepted: 27 January 2011
REFERENCES
Ali, S.A., M. Ahmad, J. Lynam, C.S. McLean, C. Entwisle, P. Loudon, E.
Choolun, S.E. McArdle, G. Li, S. Mian, and R.C. Rees. 2004. Anti-
tumour therapeutic ecacy of OX40L in murine tumour model. Vaccine.
22:3585–3594. doi:10.1016/j.vaccine.2004.03.041
Aspord, C., A. Pedroza-Gonzalez, M. Gallegos, S. Tindle, E.C. Burton, D.
Su, F. Marches, J. Banchereau, and A.K. Palucka. 2007. Breast cancer in-
structs dendritic cells to pr ime interleukin 13–secreting CD4+ T cells
that facilitate tumor development. J. Exp. Med. 204:1037–1047. doi:10
.1084/jem.20061120
Bell, D., P. Chomarat, D. Broyles, G. Netto, G.M. Harb, S. Lebecque, J.
Valladeau, J. Davoust, K.A. Palucka, and J. Banchereau. 1999. In breast
carcinoma tissue, immature dendritic cells reside within the tumor,
whereas mature dendritic cells are located in peritumoral areas. J. Exp.
Med. 190:1417–1426. doi:10.1084/jem.190.10.1417
Berzofsky, J.A., and M. Terabe. 2008. A novel immunoregulatory axis
of NKT cell subsets regulating tumor immunity. Cancer Immunol.
Immunother. 57:1679–1683. doi:10.1007/s00262-008-0495-4
Brimnes, M.K., L. Bonifaz, R.M. Steinman, and T.M. Moran. 2003.
Inuenza virus–induced dendritic cell maturation is associated with
the induction of strong T cell immunity to a coadministered, normally
nonimmunogenic protein. J. Exp. Med. 198:133–144. doi:10.1084/
jem.20030266
Camp, J.T., F. Elloumi, E. Roman-Perez, J. Rein, D.A. Stewart, J.C. Har rell,
C.M. Perou, and M.A. Troester. 2011. Interactions with broblasts are
distinct in Basal-like and luminal breast cancers. Mol. Cancer Res. 9:3–13.
doi:10.1158/1541-7786.MCR-10-0372
Caux, C., C. Massacrier, B. Vanbervliet, B. Dubois, I. Durand, M. Cella, A.
Lanzavecchia, and J. Banchereau. 1997. CD34+ hematopoietic pro-
genitors from human cord blood dierentiate along two independent
dendritic cell pathways in response to granulocyte-macrophage colony-
stimulating factor plus tumor necrosis factor alpha: II. Functional analy-
sis. Blood. 90:1458–1470.
Condeelis, J., and J.W. Pollard. 2006. Macrophages: obligate partners for
tumor cell migration, invasion, and metastasis. Cell. 124:263–266. doi:10
.1016/j.cell.2006.01.007
Coughlin, S.S., and D.U. Ekwueme. 2009. Breast cancer as a global
health concern. Cancer Epidemiol. 33:315–318. doi:10.1016/j.canep
.2009.10.003
Coussens, L.M., and Z. Werb. 2002. Inammation and cancer. Nature.
420:860–867. doi:10.1038/nature01322
Croft, M., T. So, W. Duan, and P. Soroosh. 2009. The signicance of OX40 and
OX40L to T-cell biology and immune disease. Immunol. Rev. 229:173–
191. doi:10.1111/j.1600-065X.2009.00766.x
DeNardo, D.G., J.B. Barreto, P. Andreu, L. Vasquez, D. Tawk, N. Kolhatkar,
and L.M. Coussens. 2009. CD4(+) T cells regulate pulmonary metastasis
of mammary carcinomas by enhancing protumor properties of macro-
phages. Cancer Cell. 16:91–102. doi:10.1016/j.ccr.2009.06.018
Dudziak, D., A.O. Kamphorst, G.F. Heidkamp, V.R. Buchholz, C.
Trumpfheller, S. Yamazaki, C. Cheong, K. Liu, H.W. Lee, C.G. Park,
et al. 2007. Dierential antigen processing by dendritic cell subsets
in vivo. Science. 315:107–111. doi:10.1126/science.1136080
Finkelman, F.D., A. Lees, R. Birnbaum, W.C. Gause, and S.C. Morris. 1996.
Dendritic cells can present antigen in vivo in a tolerogenic or immuno-
genic fashion. J. Immunol. 157:1406–1414.
Gilliet, M., V. Soumelis, N. Watanabe, S. Hanabuchi, S. Antonenko, R. de
Waal-Malefyt, and Y.J. Liu. 2003. Human dendritic cells activated by
490 TSLP–OX40L–IL-13 axis in human breast cancer | Pedroza-Gonzalez et al.
Mantovani, A., P. Romero, A.K. Palucka, and F.M. Marincola. 2008. Tumour
immunity: eector response to tumour and role of the microenviron-
ment. Lancet. 371:771–783. doi:10.1016/S0140-6736(08)60241-X
Morris, A., J.T. Vetto, T. Ramstad, C.J. Funatake, E. Choolun, C. Entwisle, and
A.D. Weinberg. 2001. Induction of anti-mammary cancer immunity by
engaging the OX-40 receptor in vivo. Breast Cancer Res. Treat. 67:71–80.
doi:10.1023/A:1010649303056
Neve, R.M., K. Chin, J. Fridlyand, J. Yeh, F.L. Baehner, T. Fevr, L. Clark, N.
Bayani, J.P. Coppe, F. Tong, et al. 2006. A collection of breast cancer cell
lines for the study of functionally distinct cancer subtypes. Cancer Cell.
10:515–527. doi:10.1016/j.ccr.2006.10.008
Park, J.M., M. Terabe, L.T. van den Broeke, D.D. Donaldson, and J.A.
Berzofsky. 2005. Unmasking immunosurveillance against a syngeneic
colon cancer by elimination of CD4+ NKT regulatory cells and IL-13.
Int. J. Cancer. 114:80–87. doi:10.1002/ijc.20669
Park, J.M., M. Terabe, D.D. Donaldson, G. Forni, and J.A. Berzofsky. 2008.
Natural immunosurveillance against spontaneous, autochthonous breast
cancers revealed and enhanced by blockade of IL-13-mediated nega-
tive regulation. Cancer Immunol. Immunother. 57:907–912. doi:10.1007/
s00262-007-0414-0
Piconese, S., B. Valzasina, and M.P. Colombo. 2008. OX40 triggering blocks
suppression by regulatory T cells and facilitates tumor rejection. J. Exp.
Med. 205:825–839. doi:10.1084/jem.20071341
Pulendran, B., J.L. Smith, G. Caspary, K. Brasel, D. Pettit, E. Maraskovsky,
and C.R. Maliszewski. 1999. Distinct dendritic cell subsets dierentially
regulate the class of immune response in vivo. Proc. Natl. Acad. Sci. USA.
96:1036–1041. doi:10.1073/pnas.96.3.1036
Rako-Nahoum, S., and R. Medzhitov. 2009. Toll-like receptors and cancer.
Nat. Rev. Cancer. 9:57–63. doi:10.1038/nrc2541
Sarin, A., M.S. Williams, M.A. Alexander-Miller, J.A. Berzofsky, C.M.
Zacharchuk, and P.A. Henkart. 1997. Target cell lysis by CTL granule
exocytosis is independent of ICE/Ced-3 family proteases. Immunity.
6:209–215. doi:10.1016/S1074-7613(00)80427-6
Shimamura, T., T. Fujisawa, S.R. Husain, B. Joshi, and R.K. Puri. 2010.
Interleukin 13 mediates signal transduction through interleukin 13
receptor alpha2 in pancreatic ductal adenocarcinoma: role of IL-13
Pseudomonas exotoxin in pancreatic cancer therapy. Clin. Cancer Res.
16:577–586. doi:10.1158/1078-0432.CCR-09-2015
Skinnider, B.F., A.J. Elia, R.D. Gascoyne, L.H. Trümper, F. von Bonin, U.
Kapp, B. Patterson, B.E. Snow, and T.W. Mak. 2001. Interleukin 13
and interleukin 13 receptor are frequently expressed by Hodgkin
and Reed-Sternberg cells of Hodgkin lymphoma. Blood. 97:250–255.
doi:10.1182/blood.V97.1.250
Skinnider, B.F., A.J. Elia, R.D. Gascoyne, B. Patterson, L. Trumper, U. Kapp,
and T.W. Mak. 2002. Signal transducer and activator of transcription 6 is
frequently activated in Hodgkin and Reed-Ster nberg cells of Hodgkin
lymphoma. Blood. 99:618–626. doi:10.1182/blood.V99.2.618
Smirnov, D.A., and V.G. Cheung. 2008. ATM gene mutations result in
both recessive and dominant expression phenotypes of genes and
microRNAs. Am. J. Hum. Genet. 83:243–253. doi:10.1016/j.ajhg
.2008.07.003
Soule, H.D., J. Vazguez, A. Long, S. Albert, and M. Brennan. 1973. A human
cell line from a pleural eusion derived from a breast carcinoma. J. Natl.
Cancer Inst. 51:1409–1416.
Soumelis, V., P.A. Reche, H. Kanzler, W. Yuan, G. Edward, B. Homey, M.
Gilliet, S. Ho, S. Antonenko, A. Lauerma, et al. 2002. Human epithelial
cells trigger dendr itic cell mediated allergic inammation by producing
TSLP. Nat. Immunol. 3:673–680.
Steinbrink, K., H. Jonuleit, G. Müller, G. Schuler, J. Knop, and A.H.
Enk. 1999. Interleukin-10-treated human dendritic cells induce a
melanoma-antigen-specic anergy in CD8(+) T cells resulting in a fail-
ure to lyse tumor cells. Blood. 93:1634–1642.
Steinman, R.M., and J. Banchereau. 2007. Taking dendritic cells into medi-
cine. Nature. 449:419–426. doi:10.1038/nature06175
Steinman, R.M., D. Hawiger, and M.C. Nussenzweig. 2003. Tolerogenic
dendritic cells. Annu. Rev. Immunol. 21:685–711. doi:10.1146/annurev
.immunol.21.120601.141040
Swift, M., P.J. Reitnauer, D. Morrell, and C.L. Chase. 1987. Breast and other
cancers in families with ataxia-telangiectasia. N. Engl. J. Med. 316:1289–
1294. doi:10.1056/NEJM198705213162101
Terabe, M., S. Matsui, N. Noben-Trauth, H. Chen, C. Watson, D.D. Donaldson,
D.P. Carbone, W.E. Paul, and J.A. Berzofsky. 2000. NKT cell-mediated re-
pression of tumor immunosurveillance by IL-13 and the IL-4R-STAT6
pathway. Nat. Immunol. 1:515–520. doi:10.1038/82771
Terabe, M., S. Matsui, J.M. Park, M. Mamura, N. Noben-Trauth, D.D.
Donaldson, W. Chen, S.M. Wahl, S. Ledbetter, B. Pratt, et al. 2003.
Transforming growth factor-beta production and myeloid cells are an
eector mechanism through which CD1d-restricted T cells block cyto-
toxic T lymphocyte-mediated tumor immunosurveillance: abrogation
prevents tumor recurrence. J. Exp. Med. 198:1741–1752. doi:10.1084/
jem.20022227
Todaro, M., Y. Lombardo, M.G. Francipane, M.P. Alea, P. Cammareri, F.
Iovino, A.B. Di Stefano, C. Di Bernardo, A. Agrusa, G. Condorelli,
et al. 2008. Apoptosis resistance in epithelial tumors is mediated by tumor-
cell-derived interleukin-4. Cell Death Dier. 15:762–772. doi:10.1038/
sj.cdd.4402305
Trieu, Y., X.Y. Wen, B.F. Skinnider, M.R. Bray, Z. Li, J.O. Claudio, E. Masih-
Khan, Y.X. Zhu, S. Trudel, J.A. McCart, et al. 2004. Soluble interleukin-
13Ralpha2 decoy receptor inhibits Hodgkin’s lymphoma growth
in vitro and in vivo. Cancer Res. 64:3271–3275. doi:10.1158/0008-5472
.CAN-03-3764
Villablanca, E.J., L. Raccosta, D. Zhou, R. Fontana, D. Maggioni, A. Negro, F.
Sanvito, M. Ponzoni, B. Valentinis, M. Bregni, et al. 2010. Tumor-mediated
liver X receptor-alpha activation inhibits CC chemokine receptor-7 ex-
pression on dendritic cells and dampens antitumor responses. Nat. Med.
16:98–105. doi:10.1038/nm.2074
Watanabe, N., S. Hanabuchi, M.A. Marloie-Provost, S. Antonenko, Y.J.
Liu, and V. Soumelis. 2005a. Human TSLP promotes CD40 ligand-
induced IL-12 production by myeloid dendritic cells but maintains
their Th2 priming potential. Blood. 105:4749–4751. doi:10.1182/
blood-2004-09-3622
Watanabe, N., Y.H. Wang, H.K. Lee, T. Ito, Y.H. Wang, W. Cao, and Y.J. Liu.
2005b. Hassall’s corpuscles instruct dendritic cells to induce CD4+CD25+
regulatory T cells in human thymus. Nature. 436:1181–1185. doi:
10.1038/nature03886
Weinberg, A.D., M.M. Rivera, R. Prell, A. Morris, T. Ramstad, J.T. Vetto,
W.J. Urba, G. Alvord, C. Bunce, and J. Shields. 2000. Engagement of
the OX-40 receptor in vivo enhances antitumor immunity. J. Immunol.
164:2160–2169.
Xie, F., Q. Wang, Y. Chen, Y. Gu, H. Mao, W. Zeng, and X. Zhang. 2010.
Costimulatory molecule OX40/OX40L expression in ductal carcinoma
in situ and invasive ductal carcinoma of breast: an immunohistochemistry-
based pilot study. Pathol. Res. Pract. 206:735–739. doi:10.1016/j.prp.2010
.05.016
Zhang, W.J., B.H. Li, X.Z. Yang, P.D. Li, Q. Yuan, X.H. Liu, S.B. Xu, Y. Zhang,
J. Yuan, G.S. Gerhard, et al. 2008. IL-4-induced Stat6 activities aect
apoptosis and gene expression in breast cancer cells. Cytokine. 42:39–47.
doi:10.1016/j.cyto.2008.01.016
Ziegler, S.F., and D. Artis. 2010. Sensing the outside world: TSLP regulates
barrier immunity. Nat. Immunol. 11:289–293. doi:10.1038/ni.1852
... In addition, Th2 cells are well recognized as tumor promoters. Th2 cells are "driven" by OX40 ligand (L)-expressing dendritic cells in response to cancer-derived thymic stromal lymphopoietin (TSLP) [66]. Th2 CD4 + T lymphocytes secrete IL4 and IL13. ...
Article
Full-text available
Simple Summary Although Warthin’s tumor is a well-known and frequent tumor in the salivary gland, its pathogenesis is not fully understood. Warthin’s tumor is composed of oncocytic epithelial cells lining papillary and cystic structures in a lymphoid stroma. Previously several hypotheses have been postulated. The risk factors for its development are known and they include aging, smoking, and radiation exposure. Recent findings have suggested that chronic inflammation and aging cells promote the growth of Warthin’s tumor. In this short review, we propose that DNA dame, metabolic dysfunction of mitochondria, senescence-associated secretory phenotype, human papillomavirus, and IgG-4 may be involved in the development of Warthin’s tumor. Abstract Warthin’s tumor is the second most frequent neoplasm next to pleomorphic adenoma in the salivary gland, mostly in the parotid gland. The epithelial cells constituting a tumor are characterized by the presence of mitochondria that undergo structural and functional changes, resulting in the development of oncocytes. In addition to containing epithelial cells, Warthin’s tumors contain abundant lymphocytes with lymph follicles (germinal centers) that are surrounded by epithelial cells. The pathogenesis of Warthin’s tumor is not fully understood, and several hypotheses have been proposed. The risk factors for the development of Warthin’s tumor, which predominantly occurs in males, include aging, smoking, and radiation exposure. Recently, it has been reported that chronic inflammation and aging cells promote the growth of Warthin’s tumor. Several reports regarding the origin of the tumor have suggested that (1) Warthin’s tumor is an IgG4-related disease, (2) epithelial cells that compose Warthin’s tumor accumulate mitochondria, and (3) Warthin’s tumor is a metaplastic lesion in the lymph nodes. It is possible that the pathogenesis of Warthin’s tumor includes mitochondrial metabolic abnormalities, accumulation of aged cells, chronic inflammation, and senescence-associated secretory phenotype (SASP). In this short review, we propose that DNA damage, metabolic dysfunction of mitochondria, senescent cells, SASP, human papillomavirus, and IgG4 may be involved in the development of Warthin’s tumor.
... In particular, it was demonstrated that Tfh cells are a major source of IL-4 within the TMR [65]. This is of special importance since increased levels of IL-4 are commonly found in various types of primary and metastatic cancers, including CRC, and since IL-4 is known to act as a tumor-promoting cytokine [65][66][67][68][69][70][71]. ...
Article
Full-text available
Simple Summary MicroRNA (miR) 494-5p has been associated with cancer progression, but molecules mediating this are not well understood. Here, we show that the 3’UTR of JAK1 is physically targeted, and JAK1 is downregulated in expression by miR-494-5p in colorectal cancer (CRC). Additionally, CRC cell proliferation, spontaneous and IL-4-induced invasion, and migration were significantly reduced by this miR, as well as IL-4-induced phosphorylation of JAK1, STAT6, and AKT. High JAK1 expression significantly correlated with reduced patient survival. Together, our findings suggest JAK1 as a novel target of miR-494-5p, with its translational repression contributing to CRC progression and the initial steps of metastasis. Abstract MiR-494-5p expression has been suggested to be associated with colorectal cancer (CRC) and its metastases in our previous studies. However, functional investigations on the molecule-mediating actions of this miR in CRC are lacking. In silico analysis in the present study revealed a putative binding sequence within the 3′UTR of JAK1. Overexpression of miR-494-5p in cultured CRC significantly reduced the luciferase activity of a reporter plasmid containing the wild-type JAK1-3′UTR, which was abolished by seed sequence mutation. Furthermore, the overexpression of miR-494-5p in CRC cell lines led to a significant reduction in JAK1 expression, proliferation, in vitro migration, and invasion. These effects were abolished by co-transfection with a specific double-stranded RNA that inhibits endogenous miR-494-5p. Moreover, IL-4-induced migration, invasion, and phosphorylation of JAK1, STAT6, and AKT proteins were reduced after an overexpression of this miR, suggesting that this miR affects one of the most essential pathways in CRC. A Kaplan–Meier plotter analysis revealed that patients with high JAK1 expression show reduced survival. Together, these data suggest that miR-494-5p physically inhibits the expression of JAK1 at the translational level as well as in migration and invasion, supporting the hypothesis of miR-494-5p as an early tumor suppressor and inhibitor of early steps of metastasis in CRC.
Article
Breast cancer (BC) is one of the most common malignancies in women worldwide. Numerous studies in immuno‐oncology and successful trials of immunotherapy have demonstrated the causal role of the immune system in cancer pathogenesis. The interaction between the tumor and the immune system is known to have a dual nature. Despite cytotoxic lymphocyte activity against transformed cells, a tumor can escape immune surveillance and leverage chronic inflammation to maintain its own development. Research on antitumor immunity primarily focuses on the role of the tumor microenvironment, whereas the systemic immune response beyond the tumor site is described less thoroughly. Here, a comprehensive review of the formation of the immune profile in breast cancer patients is offered. The interplay between systemic and local immune reactions as self‐sustaining mechanism of tumor progression is described and the functional activity of the main cell populations related to innate and adaptive immunity is discussed. Additionally, the interaction between different functional levels of the immune system and their contribution to the development of the pro‐ or anti‐tumor immune response in BC is highlighted. The presented data can potentially inform the development of new immunotherapy strategies in the treatment of patients with BC.
Article
Thymic stromal lymphopoietin (TSLP) is a member of the IL-2 cytokine family and has been widely recognized as a master regulator of type 2 inflammatory responses at barrier surfaces. Recent studies found dysregulation of the TSLP–TSLP receptor (TSLPR) pathway is associated with the pathogenesis of not only allergic diseases but also a wide variety of cancers including both solid tumors and hematological tumors. Thus, the blockade of TSLP represents an attractive therapeutic strategy for allergic diseases and cancer. In this study, we report the development of a novel humanized anti-TSLP monoclonal antibody (mAb) HZ-1127. Binding affinity, specificity, and ability of HZ-1127 in inhibiting TSLP were tested. HZ-1127 selectively binds to the TSLP cytokine with high affinity and specificity. Furthermore, HZ-1127 dramatically inhibits TSLP-dependent STAT5 activation and is more potent than Tezepelumab, which is an FDA-approved humanized mAb against TSLP for severe asthma treatment in inhibiting TSLP-induced CCL17 and CCL22 chemokines secretion in human peripheral blood mononuclear cells. Our pre-clinical study demonstrates that HZ-1127 may serve as a potential therapeutic agent for allergic diseases and cancer.
Article
Checkpoint inhibitor therapy has become increasingly important and has been endorsed as a treatment regimen in breast cancer. But benefits were limited to a small proportion of patients. We aimed to develop an improved signature on the basis of immune genes for detection of potential benefit from immunotherapy. Gene expression data of patients with breast cancer initially extracted from The Cancer Genome Atlas were analyzed. Ten genes were selected from the interaction of differentially expressed genes as well as immune-related genes to develop a survival signature. We compared the high-risk and low-risk groups by gene set enrichment analysis, immune infiltration, checkpoint molecule expression and immunophenoscore. Ten genes were extracted from interactions of differentially expressed and immune-related genes. The immune risk score was determined on the basis of the Cox regression coefficient of hub genes and validated with the GSE96058 dataset. Immune cell infiltrates, including CD8 + T cells, plasma cells, follicular helper T cells, CD4 + memory T cells, M1 macrophages, regulatory T cells and resting NK cells, were more highly infiltrated in the high-risk group as compared to the low-risk group. Checkpoint molecules, including CTLA-4, PD-L1, TIM-3, VISTA, ICOS, PD-1, and PD-L2, were expressed at markedly lower levels in the high-risk group as compared to the low-risk group. Immunophenoscores, as a surrogate of response to immune checkpoint therapy, was observed significant lower in the high-risk group. The 10-gene prognostic signature could identify patients’ survival and was correlated with the biomarkers of immune checkpoint inhibitor therapy, which may guide precise therapeutic decisions in clinical practice.
Article
Background: The enriched proteins within in vitro fertilisation (IVF)-generated human embryonic microenvironment could reverse progestin resistance in endometrial cancer (EC). Methods: The expression of thymic stromal lymphopoietin (TSLP) in EC was evaluated by immunoblot and IHC analysis. Transcriptome sequencing screened out the downstream pathway regulated by TSLP. The role of TSLP, androgen receptor (AR) and KANK1 in regulating the sensitivity of EC to progestin was verified through a series of in vitro and in vivo experiments. Results: TSLP facilitates the formation of a BMP4/BMP7 heterodimer, resulting in activation of Smad5, augmenting AR signalling. AR in turn sensitises EC cells to progestin via KANK1. Downregulation of TSLP, loss of AR and KANK1 in EC patients are associated with tumour malignant progress. Moreover, exogenous TSLP could rescue the anti-tumour effect of progestin on mouse in vivo xenograft tumour. Conclusions: Our findings suggest that TSLP enhances the sensitivity of EC to progestin through the BMP4/Smad5/AR/KANK1 axis, and provide a link between embryo development and cancer progress, paving the way for the establishment of novel strategy overcoming progestin resistance using embryo original factors.
Preprint
Full-text available
Cutaneous T-cell lymphoma (CTCL) is a potentially fatal clonal malignancy of T cells primarily affecting the skin. The most common form of CTCL, mycosis fungoides (MF), can be difficult to diagnose resulting in treatment delay. The pathogenesis of CTCL is not fully understood due to limited data from patient studies. We performed single-cell RNA sequencing and spatial transcriptomics profiling of skin from patients with MF-type CTCL, and an integrated comparative analysis with human skin cell atlas datasets from healthy skin, atopic dermatitis and psoriasis. We reveal the co-optation of Th2-immune gene programmes by malignant CTCL cells and modelling of the tumour microenvironment to support their survival. We identify MHC-II+ fibroblast subsets reminiscent of lymph node T-zone reticular cells and monocyte-derived dendritic cells that can maintain Th2-like tumour cells. CTCL Th2-like tumour cells are spatially associated with B cells, forming aggregates reminiscent of tertiary lymphoid structures which are more prominent with progressive disease. Finally, we validated the enrichment of B cells in CTCL skin infiltrates and its association with disease progression across three independent patient cohorts. Our findings provide diagnostic aids, potential biomarkers for disease staging and therapeutic strategies for CTCL.
Article
Background. Thymic stromal lymphopoietin (TSLP) and its receptor (TSLPR) are expressed in various cancer cells. However, their role in cancer development is not well defined. Aim. To investigate the effects of anti-TSLPR antibody on the viability, proapoptotic genes expression, and production of pro-inflammatory cytokines in MCF-7 and A549 cancer cells. Materials and Methods. MCF-7 and A549 cells were exposed to anti-TSLPR monoclonal antibody for 24, 48, and 72 h. The effect on cell viability was examined by MTT assay. The expression levels of TP53, BAX, and CASP3 genes were evaluated by the quantitative reverse transcription polymerase chain reaction (qRT-PCR). Levels of interleukin (IL)-6, tumor necrosis factor-alpha (TNF-α), and transforming growth factor (TGF-β1) were measured by the enzyme-linked immunosorbent assay (ELISA). Results. The treatment of MCF-7 cells with anti- TSLPR antibody slightly stimulates cell proliferation after 48 h and 72 h following initial cytotoxicity in 24 h with a significant reduction in IL-6 and TNF-α production. A significant increase in the BAX expression in anti-TSLPR treated cells at a concentration of 2.5 μg/ml at 24-h point was evident. In anti-TSLPR-treated A549 cells, no decrease in cell count was observed, and slight dose-dependent stimulation of cell proliferation was evident in 48 h and 72 h of culture. A significant increase in TP53, BAX, and CASP3 expression upon treatment with 2.5 μg/ml of anti-TSLPR was evident in A549 cells. Conclusion. The effects of anti-TSLPR on cell viability, proapoptotic gene expression, and production of pro-inflammatory cytokines (IL-6 and TNF-α) vary in MCF-7 and A549 cells.
Article
Full-text available
Simple Summary New insights into the foundation of cellular and molecular cancer immunology have revealed that immune cells play crucial roles in the development and growth of breast cancer (BC). T-cells are one of the most important cells in the tumor microenvironment and are divided into several subtypes including helper, cytotoxic, and regulatory subsets according to their transcription factors, markers, and functions. This article provides a comprehensive review of contradictory functions of various T-cell subsets in the prognosis and treatment of patients with BC, and crosstalk between tumor cells and T-cells. The literature shows that the role of T-cells in BC immunity depends on a variety of factors, including the tumor type or subtype, the stage of the disease, the localization of the cells in the tumor tissue and the presence of different cells or cytokines. Abstract Breast cancer (BC) is the most common cancer type in women and the second leading cause of death. Despite recent advances, the mortality rate of BC is still high, highlighting a need to develop new treatment strategies including the modulation of the immune system and immunotherapies. In this regard, understanding the complex function of the involved immune cells and their crosstalk with tumor cells is of great importance. T-cells are recognized as the most important cells in the tumor microenvironment and are divided into several subtypes including helper, cytotoxic, and regulatory T-cells according to their transcription factors, markers, and functions. This article attempts to provide a comprehensive review of the role of T-cell subsets in the prognosis and treatment of patients with BC, and crosstalk between tumor cells and T-cells. The literature overwhelmingly contains controversial findings mainly due to the plasticity of T-cell subsets within the inflammatory conditions and the use of different panels for their phenotyping. However, investigating the role of T-cells in BC immunity depends on a variety of factors including tumor types or subtypes, the stage of the disease, the localization of the cells in the tumor tissue and the presence of different cells or cytokines.
Article
Full-text available
We have analyzed the presence of immature and mature dendritic cells (DCs) within adenocarcinoma of the breast using immunohistochemistry. Immature DCs were defined by expression of CD1a-, Langerin-, and intracellular major histocompatibility complex class II–rich vesicles. Mature DCs were defined by expression of CD83 and DC-Lamp. Breast carcinoma cells were defined by morphology and/or cytokeratin expression. We demonstrate two levels of heterogeneity of DCs infiltrating breast carcinoma tissue: (a) immature CD1a+ DCs, mostly of the Langerhans cell type (Langerin+), were retained within the tumor bed in 32/32 samples and (b) mature DCs, CD83+DC-Lamp+, present in 20/32 samples, are confined to peritumoral areas. The high numbers of immature DCs found in the tumor may be best explained by high levels of macrophage inflammatory protein 3α expression by virtually all tumor cells. Confirming the immature/mature DC compartmentalization pattern, in vitro–generated immature DCs adhere to the tumor cells, whereas mature DCs adhere selectively to peritumoral areas. In some cases, T cells are clustering around the mature DCs in peritumoral areas, thus resembling the DC–T cell clusters of secondary lymphoid organs, which are characteristic of ongoing immune reactions.
Article
Full-text available
Article
Full-text available
Cells of the dendritic family display some unique properties that confer to them the capacity to sensitize naive T cells in vitro and in vivo. In the mouse, two subclasses of dendritic cells (DCs) have been described that differ by their CD8α expression and their localization in lymphoid organs. The physiologic function of both cell populations remains obscure. Studies conducted in vitro have suggested that CD8α+ DCs could play a role in the regulation of immune responses, whereas conventional CD8α− DCs would be more stimulatory. We report here that both subclasses of DCs efficiently prime antigen-specific T cells in vivo, and direct the development of distinct T helper (Th) populations. Antigen-pulsed CD8α+ and CD8α− DCs are separated after overnight culture in recombinant granulocyte/macrophage colony-stimulating factor and injected into the footpads of syngeneic mice. Administration of CD8α− DCs induces a Th2-type response, whereas injection of CD8α+ DCs leads to Th1 differentiation. We further show that interleukin 12 plays a critical role in Th1 development by CD8α+ DCs. These findings suggest that the nature of the DC that presents the antigen to naive T cells may dictate the class selection of the adaptative immune response.
Article
Full-text available
Using a mouse model in which tumors show a growth-regression-recurrence pattern, we investigated the mechanisms for down-regulation of cytotoxic T lymphocyte–mediated tumor immuno-surveillance. We found that interleukin 4 receptor (IL-4R) knockout and downstream signal transducer and activator of transcription 6 (STAT6) knockout, but not IL-4 knockout, mice resisted tumor recurrence, which implicated IL-13, the only other cytokine that uses the IL-4R–STAT6 pathway. We confirmed this by IL-13 inhibitor (sIL-13Rα2–Fc) treatment. Loss of natural killer T cells (NKT cells) in CD1 knockout mice resulted in decreased IL-13 production and resistance to recurrence. Thus, NKT cells and IL-13, possibly produced by NKT cells and signaling through the IL-4R–STAT6 pathway, are necessary for down-regulation of tumor immunosurveillance. IL-13 inhibitors may prove to be a useful tool in cancer immunotherapy.
Article
Full-text available
Basal-like breast cancers have several well-characterized distinguishing molecular features, but most of these are features of the cancer cells themselves. The unique stromal-epithelial interactions, and more generally, microenvironmental features of basal-like breast cancers have not been well characterized. To identify characteristic microenvironment features of basal-like breast cancer, we performed cocultures of several basal-like breast cancer cell lines with fibroblasts and compared these with cocultures of luminal breast cancer cell lines with fibroblasts. Interactions between basal-like cancer cells and fibroblasts induced expression of numerous interleukins and chemokines, including IL-6, IL-8, CXCL1, CXCL3, and TGFβ. Under the influence of fibroblasts, basal-like breast cancer cell lines also showed increased migration in vitro. Migration was less pronounced for luminal lines; but, these lines were more likely to have altered proliferation. These differences were relevant to tumor biology in vivo, as the gene set that distinguished luminal and basal-like stromal interactions in coculture also distinguishes basal-like from luminal tumors with 98% accuracy in 10-fold cross-validation and 100% accuracy in an independent test set. However, comparisons between cocultures where cells were in direct contact and cocultures where interaction was solely through soluble factors suggest that there is an important impact of direct cell-to-cell contact. The phenotypes and gene expression changes invoked by cancer cell interactions with fibroblasts support the microenvironment and cell-cell interactions as intrinsic features of breast cancer subtypes.
Article
Dendritic cells (DC) are critically involved in the initiation of primary immune processes, including tumor rejection. In our study, we investigated the effect of interleukin-10 (IL-10)–treated human DC on the properties of CD8+ T cells that are known to be essential for the destruction of tumor cells. We show that IL-10–pretreatment of DC not only reduces their allostimulatory capacity, but also induces a state of alloantigen-specific anergy in both primed and naive (CD45RA+) CD8+ T cells. To investigate the influence of IL-10–treated DC on melanoma-associated antigen-specific T cells, we generated a tyrosinase-specific CD8+ T-cell line by several rounds of stimulation with the specific antigen. After coculture with IL-10–treated DC, restimulation of the T-cell line with untreated, antigen-pulsed DC demonstrated peptide-specific anergy in the tyrosinase-specific T cells. Addition of IL-2 to the anergic T cells reversed the state of both alloantigen- or peptide-specific anergy. In contrast to optimally stimulated CD8+ T cells, anergic tyrosinase-specific CD8+ T cells, after coculture with peptide-pulsed IL-10–treated DC, failed to lyse an HLA-A2–positive and tyrosinase-expressing melanoma cell line. Thus, our data demonstrate that IL-10–treated DC induce an antigen-specific anergy in cytotoxic CD8+ T cells, a process that might be a mechanism of tumors to inhibit immune surveillance by converting DC into tolerogenic antigen-presenting cells.
Article
In response to granulocyte-macrophage colony-stimulating factor plus tumor necrosis factor α, cord blood CD34+ hematopoietic progenitor cells differentiate along two unrelated dendritic cell (DC) pathways: (1) the Langerhans cells (LCs), which are characterized by the expression of CD1a, Birbeck granules, the Lag antigen, and E cadherin; and (2) CD14+ cell-derived DCs, characterized by the expression of CD1a, CD9, CD68, CD2, and factor XIIIa (Caux et al, J Exp Med 184:695, 1996). The present study investigates the functions of each population. Although the two populations are equally potent in stimulating naive CD45RA cord blood T cells through apparently identical mechanisms, each also displays specific activities. In particular CD14-derived DCs show a potent and long-lasting (from day 8 to day 13) antigen uptake activity (fluorescein isothiocyanate dextran or peroxidase) that is about 10-fold higher than that of CD1a+ cells, which is restricted to the immature stage (day 6). The antigen capture is exclusively mediated by receptors for mannose polymers. The high efficiency of antigen capture of CD14-derived cells is coregulated with the expression of nonspecific esterase activity, a tracer of lysosomial compartment. In contrast, the CD1a+ population never expresses nonspecific esterase activity. The most striking difference is the unique capacity of CD14-derived DCs to induce naive B cells to differentiate into IgM-secreting cells, in response to CD40 triggering and interleukin-2. Thus, although the two populations can allow T-cell priming, initiation of humoral responses might be preferentially regulated by the CD14-derived DCs. Altogether, those results show that different pathways of DC development might exist in vivo: (1) the LC type, which might be mainly involved in cellular immune responses, and (2) the CD14-derived DC related to dermal DCs or circulating blood DCs, which could be involved in humoral immune responses.
Article
Recent studies have demonstrated that the malignant Reed-Sternberg cells of Hodgkin’s lymphoma (HL) secrete and are responsive to interleukin (IL)-13. We hypothesized that overexpression of a soluble IL-13 decoy receptor (sIL-13Rα2) via adenoviral-mediated gene transfer would inhibit IL-13-induced Reed-Sternberg cell proliferation. Western blot and ELISA analysis verified expression of sIL-13Rα2 in cell lysates and supernatants of AdsIL-13Rα2-transduced COS-7 cells. Treatment of two IL-13-responsive HL-derived cell lines, HDLM-2 and l-1236, with AdsIL-13Rα2-conditioned medium, resulted in the inhibition of cell proliferation, and down-regulated the phosphorylation of signal transducer and activator of transcription 6 (STAT6), an important mediator of IL-13 signaling. i.v. delivery of AdsIL-13Rα2 in NOD/SCID mice with s.c. implanted HDLM-2 cells delayed tumor onset and growth while enhancing survival compared with control mice. Intratumoral administration of AdsIL-13Rα2 led to the regression or stabilization of established tumors and was associated with diminished STAT6 phosphorylation. Our data demonstrate that AdsIL-13Rα2 can suppress HL growth in vitro and .
Article
OX40, a membrane-bound member of the tumor-necrosis-factor-receptor (TNFR) superfamily, plays an important role in proliferation, survival and infiltration of activated T cells via binding to OX40L. Recent studies indicate that OX40/OX40L system mediates the adhesion and infiltration of adult T cell leukemia (ATL). Previously, we detected OX40 expression in breast carcinoma cell lines and tissues. The correlation of expression of OX40 and OX40L and clinical features in breast carcinogenesis, however, has not been well characterized. The expression of OX40 and OX40L in 107 invasive ductal carcinomas (IDCa), 9 ductal carcinomas in situ (DCIS), and 31 fibroadenomas from breast tissues and its relationship with the clinical features were determined using immunohistochemistry (peroxidase-conjugated polymer method, ChemMate™ Envision™ Detection kit). The positive immunostaining rates for OX40 in IDCa, DCIS and fibroadenomas from breast tissues were 85.0%, 66.7% and 38.7% respectively, showing a significant difference in OX40 expression among IDCa, DCIS and fibroadenoma of breast (z=5.206, P=0.001). Increased staining intensity of OX40 was associated with TNM stages (z=2.112, P=0.017). Meanwhile, a relation of OX40 expression with lymph node metastatic status in IDCa was found (P=0.041). The expression of OX40L did not show any obvious difference among IDCa, DCIS and fibroadenomas from breast tissues. OX40L expression was also not related to histopathological parameters in IDCa except for progesterone receptor (PR) being positive (P=0.005). However, a high coincidental positive rate for OX40 and OX40L was observed in biopsy samples with IDCa (P=0.017, Kappa=0.231). The present results suggest that high OX40 expression may be associated with malignant transformation, progression, invasion and metastasis in breast cancer biology.