Skip to main content
Erschienen in: Breast Cancer Research 4/2014

Open Access 01.08.2014 | Research article

Cancer/stroma interplay via cyclooxygenase-2 and indoleamine 2,3-dioxygenase promotes breast cancer progression

verfasst von: Jing-Yi Chen, Chien-Feng Li, Cheng-Chin Kuo, Kelvin K Tsai, Ming-Feng Hou, Wen-Chun Hung

Erschienen in: Breast Cancer Research | Ausgabe 4/2014

Abstract

Introduction

Expression of indoleamine 2,3-dioxygenase (IDO) in primary breast cancer increases tumor growth and metastasis. However, the clinical significance of stromal IDO and the regulation of stromal IDO are unclear.

Methods

Metabolomics and enzyme-linked immunosorbent assay (ELISA) were used to study the effect of cyclooxygenase-2 (COX-2)-overexpressing breast cancer cells on IDO expression in co-cultured human breast fibroblasts. Biochemical inhibitors and short-hairpin RNA (shRNA) were used to clarify how prostaglandin E2 (PGE2) upregulates IDO expression. Associations of stromal IDO with clinicopathologic parameters were tested in tumor specimens. An orthotopic animal model was used to examine the effect of COX-2 and IDO inhibitors on tumor growth.

Results

Kynurenine, the metabolite generated by IDO, increases in the supernatant of fibroblasts co-cultured with COX-2-overexpressing breast cancer cells. PGE2 released by cancer cells upregulates IDO expression in fibroblasts through an EP4/signal transducer and activator of transcription 3 (STAT3)-dependent pathway. Conversely, fibroblast-secreted kynurenine promotes the formation of the E-cadherin/Aryl hydrocarbon receptor (AhR)/S-phase kinase-associated protein 2 (Skp2) complex, resulting in degradation of E-cadherin to increase breast cancer invasiveness. The enhancement of motility of breast cancer cells induced by co-culture with fibroblasts is suppressed by the IDO inhibitor 1-methyl-tryptophan. Pathological analysis demonstrates that upregulation of stromal IDO is a poor prognosis factor and is associated with of COX-2 overexpression. Co-expression of cancer COX-2 and stromal IDO predicts a worse disease-free and metastasis-free survival. Finally, COX-2 and IDO inhibitors inhibit tumor growth in vivo.

Conclusion

Integration of metabolomics and molecular and pathological approaches reveals the interplay between cancer and stroma via COX-2, and IDO promotes tumor progression and predicts poor patient survival.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13058-014-0410-1) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

JY conducted cell culture, biochemical, and molecular biology assays and prepared the draft of the manuscript. CF did IHC study and pathological analysis and helped to draft the manuscript. CC performed metabolomics study and data analysis and helped to draft the manuscript. KK participated in the design of the study and helped to draft the manuscript. MF conceived of the study and participated in the preparation of primary cancer-associated fibroblasts. WC conceived of the study and participated in the design of the study and wrote the manuscript. All authors read and approved the final manuscript.
Abkürzungen
2-HG
2-hydroxyglutarate
3′4′-DMF
3′4′-dimethoxyflavone
3-MC
3′-methylcholanthrene
AhR
aryl hydrocarbon receptor
CAF
cancer-associated fibroblast
CXCL12
chemokine (C-X-C motif) ligand 12
CXCL14
chemokine (C-X-C motif) ligand 14
DAPI
4′,6-diamidino-2-phenylindole
DTF
desmoid-type fibromatosis
ELISA
enzyme-linked immunosorbent assay
EMT
epithelial-mesenchymal transition
EP4
prostaglandin E receptor 4
IDH
isocitrate dehydrogenase
IDO
indoleamine 2,3-dioxygenase
MMP
matrix metalloproteinase
NF-κB
nuclear factor kappa-light-chain-enhancer of activated B cells
PGE2
prostaglandin E2
SDF-1
stromal cell-derived factor 1
SFT
solitary fibrous tumor
shRNA
short-hairpin RNA
Skp2
S-phase kinase-associated protein 2
STAT3
signal transducer and activator of transcription 3
TET2
Tet methylcytosine dioxygenase 2
UPLC
ultraperformance liquid chromatography

Background

Chronic inflammation is strongly associated with the development of cancer [1]-–[3]. One of the crucial mediators of inflammatory reaction is cyclooxygenase (COX). The COX family of enzymes comprises two members (COX-1 and COX-2) and is the main controller of eicosanoid biosynthesis. Studies of human breast tumor tissues demonstrate that upregulation of COX-2 has been detected in approximately 40% of human breast tumor tissues, as well as preinvasive ductal carcinoma in situ lesions [4]. Elevated expression of COX-2 is associated with large tumor size, advanced histologic grade, axillary node metastasis, and unfavorable disease-free survival [4],[5]. In addition, COX-2 expression also links with increased tumor angiogenesis [6]. Epidemiologic investigations suggest that use of nonsteroidal antiinflammatory drugs or selective COX-2 inhibitors reduces breast cancer risk [7],[8].
Results of animal study also support an oncogenic role of COX-2. Transgenic COX-2 overexpression induces mammary tumor formation in mice [9]. This tumorigenic transformation is highly dependent on PGE2 production and angiogenic switch. In addition, HER-2/Neu oncogene-induced mammary tumorigenesis and angiogenesis are dramatically attenuated in COX-2 knockout mice, suggesting a key role of COX-2 in breast cancer [10]. Recent studies also show that COX-2 inhibitors exhibit antitumor and antiangiogenic activities in vivo and exhibit chemopreventive effects against mammary carcinogenesis induced by 7,12-dimethyl-benz(a)anthracene in rats [11]. All of the results suggest that COX-2 is involved in multiple steps of breast carcinogenesis and is a potential target for cancer prevention and therapy.
Interplay between breast cancer cells and cancer-associated fibroblasts (CAFs), the most abundant and active stromal cells, is crucial for tumor growth, progression, angiogenesis, and therapeutic resistance [12]. Cancer cells release a number of factors to enhance the production of cytokines, chemokines, and matrix metalloproteinases (MMPs) from CAFs, which in turn facilitate cancer cell proliferation, migration, and invasion. Previous study demonstrated that stromal fibroblasts present in invasive breast carcinomas can secrete large amounts of stromal cell-derived factor 1 (SDF-1) to enhance tumor growth and angiogenesis [13]. Co-injection of breast cancer cells and fibroblasts also promotes the progression of ductal carcinoma in situ to invasive breast carcinoma by stimulating chemokine (C-X-C motif) ligand 14 (CXCL14) and chemokine (C-X-C motif) ligand 12 (CXCL12) production [14]. However, most studies addressing the crosstalk between cancer and stromal cells focus on protein factors like cytokines and chemokines. Whether other small molecules such as lipids or metabolites participate in cancer-stromal cell interaction is largely unknown.
The tumor-promoting role of CAFs via upregulation of COX-2 in ductal carcinoma in situ of the breast was first demonstrated by Hu et al. [15]. The authors showed that co-culture with fibroblasts increases COX-2 expression in breast cancer cells and subsequently induces MMP-9 and MMP-14 in these cells to promote invasion. They also elucidated the underlying mechanism by demonstrating that inhibition of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and COX-2 activity reduces the invasion-promoting effect of fibroblasts. These data suggest that fibroblasts secrete some factors to activate NF-κB-mediated transcription of COX-2 in breast cancer cells to enhance tumor progression.
However, several issues remain elusive. First, does PGE2 generated by COX-2-expressing cancer cells also affect gene expression or behavior of stromal fibroblasts? Second, do CAFs secrete small molecules (other than proteins or peptides) to regulate cancer cell invasion? Finally, can the importance of cancer-stroma interaction in cancer progression be validated in clinical samples?
In this study, we address these questions and try to clarify the underlying mechanism.

Methods

Cell culture

Human breast cancer cell lines MCF-7 and MDA-MB-231 were purchased from the Bioresource Collection and Research Center (BCRC) and ATCC. Immortalized human breast fibroblasts, RMF-EG [16], were kindly provided by Dr. Charlotte Kuperwasser (Tufts University, Boston, MA, USA). These cells were cultured in DMEM/F12 containing 10% fetal bovine serum (FBS). Other experimental materials and procedures are provided in Additional file 1.

Establishment of inducible COX-2-expression MCF-7 cell line

To establish an inducible COX-2-expression cell line, MCF-7 cells (1 × 106) were resuspended in buffer R containing 2 μg pCMV-Tet3G plasmid. Transfection was performed by using Neon microporation transfection system at room temperature with 1,250 V, 20 milliseconds, and two pulses. After 48 hours, the cells were selected with 1 mg/ml G418 for 2 weeks.
For the delivery of the second plasmid, pCMV-Tet3G stably transfected cells (1 × 106) were resuspended in buffer R containing 2 μg of pTRE-mCherry-COX-2 plasmid. Transfection was performed by using Neon microporation transfection system at room temperature with 1,250 V, 20 milliseconds, and two pulses. After 48 hours, the cells were subjected to selection with 100 μg/ml hygromycin B. The stable cell line harbors both pCMV-Tet3G and pTRE-mCherry-COX-2 plasmid was used for induction experiment. The cells were maintained at 37°C in a 5% CO2-humidified atmosphere and were incubated with doxycycline to induce COX-2 expression before co-culture assay.

Co-culture assay

In the co-culture system, 1 × 105 RMF-EG cells were grown in the bottom of a six-well plate in DMEM/F12 with 10% FBS, and 1 × 106 breast cancer cells were seeded on the 0.4-μm polyester membrane of a transwell insert in the same medium. MCF-7 cells were treated with or without doxycycline (1 μg/ml) for 72 hours. The conditioned medium, breast cancer cells, and RMF-EG cells were harvested for metabolomics and Western blotting analysis.

Metabolite profiling

The proteins in the conditioned medium were removed by using 3-kDa ultracentrifugation filter devices. The metabolites in the filtered medium were extracted by using iced 50% methanol and were subsequently dried by a speedvac. Metabolite profiles were analyzed with the Metabolomics Core of National Health Research Institutes by using a high-resolution ultraperformance liquid chromatography (UPLC) coupled online to a triple-quadrupole time-of-flight mass spectrometry system, as described previously [17]. Metabolite identity was predicted with Human Metabolome Database [18].

RNA extraction and quantitative reverse transcription-PCR analysis

Total RNA was isolated from cells by using an RNA extraction kit (Qiagen, Valencia, CA, USA) and 1 μg of RNA was reverse-transcribed to cDNA. Target mRNAs were quantified by using real-time PCR reactions with SYBR green fluorescein, and actin served as an internal control. cDNA synthesis was performed at 95°C for 3 minutes, and the conditions for PCR were 28 cycles of denaturation (95°C/1 minute), annealing (60°C/1 minute) extension (72°C/1 minute), and 1 cycle of final extension (72°C/10 minutes). The primers used are tryptophan 2,3-dioxygenase (TDO)-forward: 5′-GGGAACTACCTGCATTTGGA-3′; TDO-reverse: 5′-GTGCATCCGAGAAACAACCT-3′; IDO-forward: 5′-GCGCTGTTGGAAATAGCTTC-3′; IDO-reverse: 5′-CAGGACGTCAAAGCACTGAA-3′; E-cadherin-forward: 5′-CCTGGGACTCCACCTACAGA-3′; E-cadherin-reverse: 5′-GGATGAACACAGCGTGAGAGA-3′; actin-forward: 5′-TGTTACCAACTGGGACGACA-3′; actin-reverse: 5′-GGGGTGTTGAAGGTCTCAAA-3′.

Immunoprecipitation and Western blot analysis

MCF-7or COX-2-overexpressing MCF-7 cells were treated with or without 100 μM kynurenine for 24 hours; the cells were harvested with an RIPA buffer (50 mM Tris–HCl, pH 7.4, 150 mM NaCl, 1% NP-40, 0.1% SDS, 0.5% sodium deoxycholate, 2 mM EDTA, and 50 mM NaF), and cellular lysates were incubated with anti-AhR antibody overnight at 4°C with rotation. Immunocomplexes were pulled down by Protein-G agarose bead, washed with RIPA buffer 3 times, and eluted with a sample buffer in boiled water for 10 minutes. The eluted samples were subjected to SDS-PAGE separation, and proteins were transferred to nitrocellulose membranes. Finally, the blots were probed with anti-E-cadherin or anti-Skp2 antibody and developed with enhanced chemiluminescence reagent.

Migration assay

Migration assays were conducted in transwells with 8-μm-pore filter inserts. Then 1 × 104 MCF-7 or COX-2-overexpressing MCF-7 cells were seeded in the upper chamber. The lower chambers were filled with DMEM medium containing 1% FBS and 100 μM kynurenine. After 24 hours, the cells on the upper surface were removed by wiping with a cotton swab, and the cells that migrated to the lower surface were fixed. The cells were stained with 4′,6-diamidino-2-phenylindole (DAPI), and the cell number in 15 randomly selected fields was counted under a microscope (100×). Experiments were performed independently at least 3 times.

Protein ubiquitination assay

MCF-7 cells treated with or without kynurenine were incubated with the proteasome inhibitor MG132 or the lysosome inhibitor chloroquine. The cells were harvested with a lysis buffer (20 mM Tris–HCl at pH 7.5, 150 mM sodium chloride, 1 mM calcium chloride, and 1% Triton X-100 and protease inhibitors), and cellular lysates were incubated with an E-cadherin antibody overnight at 4°C with rotation. Protein-G beads were added to the samples and incubated for another 1 hour at 4°C. Immunocomplexes were eluted and were subjected to SDS-PAGE separation, and proteins were transferred to nitrocellulose membranes. Finally, the blots were probed by using an anti-ubiquitin antibody to detect the ubiquitination status of E-cadherin.

Immunofluorescent staining and confocal microscopy

MCF-7 cells were treated with or without 100 μM kynurenine for 6 hours and fixed with 3.7% formaldehyde for 15 minutes at room temperature. Cells were washed twice with PBS and permeabilized by 0.1% Triton X-100 solution for 10 minutes. After permeabilization, cells were incubated with 0.05% BSA solution to block nonspecific binding. Anti-AhR mouse monoclonal antibody (1:80) or anti-E-cadherin goat polyclonal antibody (1:250) was added and incubated at room temperature for 1 hour. After extensive washing, Alexas Fluro 594 anti-mouse IgG or Alexas Fluro 488 anti-goat IgG was added and incubated for another 1 hour. Cell nuclei were stained with DAPI solution. Finally, coverslips were washed twice with PBS and subsequently placed in mounting solution. The fluorescent image was observed under a confocal microscope.

In vivoorthotopic animal study

MCF-7 or MCF-7-COX2 (8 × 106) cells were mixed with RMF-EG (6 × 106) cells in Hanks balanced salt solution and Matrigel (BD Transduction Laboratories, San Jose, CA, USA). Cells were inoculated into the fourth mammary fat pads of 6-week-old female BALB/cAnN.Cg-Foxn1nu/CrlNarl mice. Before the inoculation of the cancer cell/fibroblast mixture, all mice were primed with 6 mg/kg of 17β-estradiol twice a week for 3 weeks.
After inoculation, 17β-estradiol was continuously given to mice throughout the experiments. Measurement of tumor growth was begun at 4 weeks after injection, and tumor volume was calculated by using the equation: tumor volume = (length × width2)/2. After 10 weeks, mice injected with COX-2-overexpressing MCF-7 and RMF-EG produced tumors with volumes approximately 200 mm3 and were randomly divided into four groups that received vehicle (DMSO), NS-398 (10 mg/kg), L-1-methy-tryptophan (10 mg/kg), or both inhibitors 5 times per week.
Two weeks later, animals were killed, and tumors were isolated from mice. The statistical difference between experimental groups was evaluated with repeated-measures two-way ANOVA analysis. The animal-use protocol was approved by the Institutional Animal Care and Use Committee of National Health Research Institutes.

Patients and statistical analysis

Paraffin-embedded human breast tumor tissues were obtained from Chi-Mei Medical Center (Tainan, Taiwan) between 1998 and 2004. The slides were stained with anti-COX-2 or anti-IDO antibodies. The COX-2 and IDO stainings were interpreted by using the H-score, defined by the following equation: H-score = ΣPi (i + 1), as previously described [19], where i is the intensity of the stained tumor cells (0 to 3+), and Pi is the percentage of stained tumor cells with various intensities. We classified tumors with cancer cells and stromal cells showing H-scores no less than the median of all scored cases as having high COX-2 and IDO expression, respectively.
The follow-up duration ranged from 5.4 to 143.6 months, with a mean of 87.1 months. Survival analyses for disease-specific and metastasis-free survival were performed by using Kaplan-Meier plots and compared by using the log-rank test. The correlation between COX-2 and IDO expression with clinicopathologic parameters was examined with χ2 test. P value < 0.05 was considered statistically significant. This study was approved by the Research Ethics Committee of National Health Research Institutes. Written informed consent was obtained from all patients participating in this study.

Results

COX-2-overexpressing breast cancer cells upregulated IDO expression in co-cultured fibroblasts

We analyzed the metabolite profile of the supernatant of RMF-EG human breast fibroblasts co-cultured with MCF-76 or COX-2-overexpressing MCF-7 cells and found that several metabolites were increased in the supernatant of COX-2-overexpressing MCF-7/RMF-EG co-culture. A peak with the m/z ratio of 209 was increased about twofold (Figure 1A). By using Human Metabolome Database search, a candidate metabolite was predicted to be kynurenine. UPLC/MS/MS analysis demonstrated that fragmentation of kynurenine standard yielded three peaks with m/z ratio of 209, 192, and 146, which is consistent with the reported data (accession: K0009019, MassBank, [20]) (Figure 1B). Significant increase of kynurenine was confirmed with an ELISA assay (Figure 1C).
The rate-limiting enzymes in the generation of kynurenine are indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO). We found a 2.5-fold of increase of IDO mRNA in RMF-EG cells co-cultured with COX-2-overexpressing MCF-7 cells, whereas the expression of TDO was not changed (Figure 1Di). A similar increase of IDO protein level was also found (Figure 1Dii). IDO was very low or undetectable in MCF-7- and COX-2-overexpressing MCF-7 cells, indicating that the kynurenine in the co-cultured medium was produced mainly by RMF-EG cells (Figure 1E). Co-culture of the COX-2-overexpressing MDA-MB-231 cells also upregulated IDO expression in RMF-EG cells (Figure 1F). These data suggest that COX-2-overexpressing breast cancer cells stimulate IDO expression and increase kynurenine secretion in co-cultured fibroblasts.

PGE2transcriptionally elevated IDO expression in RMF-EG fibroblasts through the EP4/STAT3 signaling pathway

We found that PGE2 increased IDO mRNA and protein levels in RMF-EG cells (Figure 2Ai and 2Aii). In addition, our data showed that only PGE2-alcohol (an EP4 agonist) significantly upregulated IDO expression (Figure 2B). Knockdown of EP4 abolished PGE2-induced increase of IDO in these cells (Figure 2Ci and 2Cii). By using different IDO deletion promoters, we demonstrated that PGE2 stimulated IDO transcription via the −1,140/-844 region from the transcription start site (see Additional file 2: Figure S1). This region contained two potential γ-interferon-activated sites (GASs) that could be activated by different signal transducer and activator of transcription (STAT) proteins [21],[22]. Both STAT1 and STAT3 have been implicated in the regulation of IDO expression [23],[24].
We performed a ChIP assay and found that the binding of STAT3 to IDO promoter was increased in RMF-EG cells co-cultured with COX-2-overexpressing MCF-7 cells, whereas the binding of STAT1 was decreased (see Additional file 3: Figure S2). Knockdown of STAT3 abolished the increase of IDO induced by the EP4 agonist in RMF-EG cells (Figure 2D). Ectopic expression of STAT3 upregulated IDO (2.8-fold) in these cells (Figure 2E). Thus, COX-2-overexpressing breast cancer cells upregulate IDO expression in fibroblasts through the PGE2/EP4/STAT3 pathway.

IDO-expressing fibroblasts enhanced the migration of breast cancer cells through downregulation of E-cadherin

We next studied the effect of kynurenine on breast cancer cells. Kynurenine did not affect the proliferation of MCF-7 cells (Figure 3A). However, kynurenine significantly increased the motility of MCF-7 and COX-2-overexpressing MCF-7 cells (Figure 3B). The conditioned medium of RMF-EG fibroblasts preincubated with COX-2-overexpressing MCF-7 cells also increased the motility of MCF-7 cells (Figure 3C). We used 1-methyl-L-tryptophan to inhibit IDO activity in RMF-EG fibroblasts induced by co-culture with COX2-overexpressing MCF-7 cells and found that the stimulatory effect on cell motility was blocked (Figure 3C). These data suggested that kynurenine released by IDO-expressing fibroblasts enhanced the migration of breast cancer cells.
We investigated the expression of epithelial-to-mesenchymal markers in kynurenine-treated breast cancer cells and found that E-cadherin was reduced in a time-dependent manner (Figure 3Di). E-cadherin began to decrease around 8 hours after addition of kynurenine, and a 70% of reduction was found at 24 hours. However, its mRNA did not decrease substantially (Figure 3Dii). We found that kynurenine induced degradation of E-cadherin protein via a proteasome-dependent pathway, which could be rescued by MG132 (proteasome inhibitor) but not chloroquine (lysosome inhibitor) (Figure 3E). In addition, ubiquitination of E-cadherin protein was increased in kynurenine-treated MCF-7 cells (Figure 3F). These data suggest that kynurenine induces ubiquitination and degradation of E-cadherin to promote breast cancer cell motility.

Kynurenine increased the degradation of E-cadherin in an AhR- and Skp2-dependent manner

Kynurenine has been shown to be an endogenous tumor-promoting ligand of the human AhR [25]. In addition, AhR is involved in the degradation of sex steroid receptors via a cullin 4B-dependent ubiquitination pathway [26]. We tested whether kynurenine reduced protein stability of E-cadherin through activation of AhR and found that the binding between AhR and E-cadherin was increased in kynurenine-treated MCF-7 cells (Figure 4A). Interestingly, Skp2, an F-box protein of the SCF E3 ligase, was co-immunoprecipitated with AhR, and the interaction was also increased by kynurenine. We did not detect the cullin 4B protein in the complex (data not shown). This is not a cell line-specific effect, because the interaction between AhR and E-cadherin protein was also elevated in kynurenine-treated A549 cells (see Additional file 4: Figure S3). The 3′-methylcholanthrene (3-MC), another AhR ligand, also induced co-localization of AhR and E-cadherin at the cell membrane (Figure 4B). Knockdown of Skp2 reversed kynurenine-induced reduction of E-cadherin protein without affecting AhR expression (Figure 4C). The AhR antagonist, 3′4′-dimethoxyflavone (3′4′-DMF), also inhibited the decrease of E-cadherin induced by kynurenine (Figure 4D). Additionally, kynurenine-increased migration of MCF-7 cells was blocked by 3′4′-DMF (Figure 4E). These data suggest that kynurenine induces the formation of E-cadherin/AhR/Skp2 complex and causes E-cadherin degradation.

COX-2 expression in breast cancer and IDO expression in stromal fibroblasts predicted poor disease-specific and metastasis-free survival

The correlation between COX-2 expression in tumor tissues and IDO expression in CAFs was confirmed by two approaches. First, we isolated CAFs from two breast tumor tissues without or with COX-2 overexpression and found that IDO expression in CAFs was upregulated in COX-2-overexpressing tumor (see Additional file 5: Figure S4). Second, we used immunohistochemical analysis to detect COX-2 and IDO expression in a cohort of breast cancer tissues (Figure 5A). COX-2 expression in tumors was positively correlated with a high IDO expression in CAFs (65 of 101, 64%; P < 0.001) (Table 1). COX-2 was highly expressed in stage III (19 of 24, 79%; P < 0.05), N1-N2 (61 of 85, 71%; P < 0.001), and T3-4 stage (20 of24, 83%; P < 0.05) tumor specimens. IDO expression in CAFs was significantly expressed in stage III (20 of 24, 83%; P < 0.05), N1-N2 (60 of 85, 71%; P < 0.001), and T3-4 stage (21 of 24, 88%; P < 0.001) tumor specimens. The disease-specific and metastasis-free survival declined significantly in patients with high COX-2 expression in breast tumors (P = 0.0043 and P < 0.0001, respectively) (Figure 5B and Table 2). Similarly, the disease-specific and metastasis-free survival declined significantly in patients with high IDO expression in CAFs (P = 0.0045 and P < 0.0001) (Figure 5C and Table 2). More important, high COX-2 in tumors and high IDO1 expression in CAFs predicted worse disease-free and metastasis-free survivals in breast cancer patients (P < 0.0001, Figure 5D).
Table 1
Correlation between Cox-2 and Ido-1 expression and various clinicopathologic factors
Parameters
Category
No. of cases
Cox-2 expression (tumor)
Pvalue
Ido-1 expression (CAF)
Pvalue
Low exp.
High exp.
Low exp.
High exp.
Age (years)
<60 years
141
73
68
0.444
69
72
0.647
 
≧60 years
61
28
33
 
32
29
 
Primary tumor (T)
T1
82
45
37
0.002
44
38
<0.001
 
T2
96
52
44
 
54
42
 
 
T3-4
24
4
20
 
3
21
 
Nodal status (N)
N0
117
77
40
<0.001
76
41
<0.001
 
N1-N2
85
24
61
 
25
60
 
Stage
I
63
37
26
0.006
37
26
0.002
 
II
115
59
56
 
60
55
 
 
III
24
5
19
 
4
20
 
Histologic grade
Grade I
18
13
5
0.122
15
3
0.003
 
Grade II
141
69
72
 
71
70
 
 
Grade III
43
19
24
 
15
28
 
CAF Ido-1 expression
Low Exp. (<medium)
101
65
35
<0.001
   
 
High Exp. (≧medium)
101
35
65
    
Bold figures, Statistically significant.
Table 2
Univariate log-rank analysis for disease-specific survival and metastasis-free survival
Parameters
Category
No. of case
DSS
MeFS
No. of events
Pvalue
No. of events
Pvalue
Age (years)
<60 years
141
13
0.9804
41
0.7650
 
≧60 years
61
4
 
16
 
Primary tumor (T)
T1
82
5
0.0283
9
<0.0001
 
T2
96
10
 
34
 
 
T3-4
24
2
 
14
 
Nodal status (N)
N0
117
7
0.0079
19
<0.0001
 
N1-2
85
10
 
38
 
Stage
I
63
3
0.0001
5
<0.0001
 
II
115
10
 
38
 
 
III
24
4
 
14
 
Histologic grade
Grade I
18
0
0.2066
1
0.0269
 
Grade II
141
15
 
41
 
 
Grade III
43
2
 
15
 
Cox-2 expression
Low Exp (<medium)
101
4
0.0043
15
<0.0001
(Tumor)
High Exp (≧medium)
101
13
 
42
 
Ido-1 expression
Low Exp (<medium)
101
4
0.0045
9
<0.0001
(CAF)
High Exp (≧medium)
101
13
 
48
 
Bold figures, Statistically significant.

COX-2 and IDO inhibitors suppressed growth of COX-2-overexpressing breast tumors in vivo

The effect of COX-2 and IDO inhibitors was evaluated in an orthotopic model. Inoculation of MCF-7/RMF-EG- or COX-2-overexpressing MCF-7/RMF-EG cell mixture induced tumors in nude mice primed with 17β-estradiol injection (Figure 6A). Tumor growth was higher in the COX-2-overexpressing MCF-7/RMF-EG group, and a 2.4-fold of increase of tumor volume was detected at 10 weeks (P < 0.01). The COX-2-overexpressing MCF-7/RMF-EG group was randomly divided into four subgroups (n = 3). Intratumoral injection of vehicle (DMSO, control), 10 mg/kg of NS398, 10 mg/kg of 1-methyl-L-tryptophan, or both inhibitors was conducted, and treatment was continuous for another 2 weeks. As shown in Figure 6B, tumor volume of the groups treated with NS398 or 1-methyl-L-tryptophan was smaller than that of the control group. Co-treatment of COX-2 and IDO inhibitor induced a more obvious reduction in tumor size, although it did not show an additive effect.

Discussion

Previous studies demonstrated that IDO overexpression increases the secretion of kynurenine to inhibit effect T cells to promote immune escape and tumor progression in various human cancers [27]-–[29]. The expression of IDO in cancer stroma has not been clarified. In addition, the clinical significance of stromal IDO is unclear.
In this study, we provide evidence that COX-2-overexpressing breasts cancer cells may secrete PGE2 to induce IDO expression and kynurenine production in stromal fibroblasts. In addition, we show that kynurenine in the coculture-conditioned medium is produced mainly by CAFs because IDO is not induced by COX-2 overexpression in MCF-7 cells. An important upstream regulator of IDO is interferon-γ. Yoshida et al. [30] first reported that the pulmonary IDO was induced in the mouse after intraperitoneal administration of bacterial endotoxin or during in vivo virus infection, and this induction was triggered by interferon-γ [30]. Because interferon-γ exhibits antitumor activity on various cancers in vitro and in vivo, it is unlikely that COX-2-overexpressing cancer cells produce interferon-γ to stimulate stromal IDO. For the first time, we show that cancer cell-produced PGE2 transcriptionally upregulates IDO expression through the EP4/STAT3 signaling pathway. In vivo binding of STAT3 to IDO gene promoter is confirmed by ChIP assay. Additionally, knockdown of STAT3 totally abolishes EP4 agonist-induced IDO expression. These data suggest that IDO is a direct transcriptional target of STAT3.
An unresolved question is why PGE2 stimulates IDO expression in stromal fibroblasts but not in breast cancer cells, because both cell types express EP4 receptor [31] and data not shown]. We are aware that the binding of STAT1 to IDO promoter is reduced by PGE2 (Additional file 3: Figure S2); therefore, it is possible that the expression level of STAT1 and STAT3 and the competition between these two STATs may determine the response of cells to PGE2 stimulation.
The concept of oncometabolite was established by the studies that mutations of isocitrate dehydrogenase 1 (IDH1) and IDH2 generate a novel metabolite 2-hydroxyglutarate (2-HG) that exhibits oncogenic activity in acute myeloid leukemia and glioma [32],[33]. Subsequently, 2-HG was shown to be a competitive inhibitor of α-ketoglutarate-dependent dioxygenases and inhibits histone demethylases like Tet methylcytosine dioxygenase 2 (TET2) to change promoter methylation and gene transcription [34],[35]. Kynurenine represents another oncometabolite, which acts as an immunosuppressor to create a favorable microenvironment for tumor formation and metastasis [36]. A recent study demonstrated that the tryptophan catabolism enzyme TDO is overexpressed in human brain tumors, and elevated secretion of kynurenine promotes cell migration via an AhR-dependent pathway [25].
However, the underlying mechanism by which kynurenine increases cell motility is still unclear. After screening of the EMT markers, we found that E-cadherin is decreased in kynurenine-treated breast cancer cells, and AhR is involved in this process. AhR has been shown to integrate as a component of a novel Cul4B ubiquitin E3 ligase complex and participated in the degradation of sex steroid receptors [26]. We demonstrated that kynurenine increases the interaction between AhR and E-cadherin, and the AhR/E-cadherin complex also contains Skp2, an F-box protein of SCF E3 ligase. The formation of the E-cadherin/AhR/Skp2 complex and ubiquitination of E-cadherin induced by kynurenine is also detectable in A549 cells, indicating a general mechanism of kynurenine-induced proteolysis of E-cadherin in different cancer cells. Our results provide a novel oncometabolite function of kynurenine to enhance cancer cell migration by degrading E-cadherin.
The clinical validation of tumor COX-2 and stromal IDO in this study is important to verify the cancer-stroma interplay in cancer progression. Many histopathologic studies investigated the expression of two specific genes in the epithelial components of tumor tissues to show their association and to demonstrate the vertical regulation of these two genes. The correlation and clinical significance of genes separately expressed in tumor and stroma have received little attention.
However, the gene signatures in CAFs may provide more information than originally thought. West et al. [37] first classified two stromal gene signature from tumors with solitary fibrous tumor (SFT) and desmoids-type fibromatosis (DTF) features and showed that patients with the expression of DTF had a favorable clinical outcome. Their subsequent study by using public databases and immunohistochemical approaches suggested that DTF fibroblast signature is a common tumor stroma signature in different types of cancers [38]. Mercier et al. [39] identified a hyperproliferative gene signature in CAFs and found that breast cancer patients with this signature had a poor prognosis with tamoxifen monotherapy and a great reduction in recurrence-free survival [39]. By using a mouse model of squamous skin carcinogenesis, Erez [40] demonstrated that carcinoma cells could educate CAFs to express proinflammatory genes to promote macrophage recruitment, neovascularization, and tumor growth. Additionally, this gene signature was also evident in mammary and pancreatic tumors in mice and in human cancers. By using metabolomics, molecular, and pathological approaches, we revealed that induction of stromal IDO by COX-2-overexpressing breast cancer cells promotes tumor progression and predicts poor patient survival.
Results of our animal study also clearly demonstrate the anticancer effect of COX-2 and IDO inhibitor on COX-2-overexpressing breast cancer in vivo. A combination of IDO and COX-2 inhibitor exhibits a more obvious effect on the inhibition of tumor growth. However, we did not find an additive effect. This can be because (a) the number of animals in each group is small, and (b) inhibition of COX-2 in cancer cells will attenuate stromal IDO expression, which reduces the anticancer activity of IDO inhibitor. Additional experiments are needed to clarify this issue.

Conclusion

By using a metabolomics approach, we identified potential oncometabolites involved in the crosstalk between COX-2-overexpressing breast cancer cells and fibroblasts. Molecular study elucidates the underlying mechanism by which this cancer/stroma interplay via COX-2 and IDO promotes tumor progression. In addition, pathological investigation validates the importance of cancer COX-2 and stromal IDO in the prediction of the patient’s survival. Simultaneous targeting of COX-2 and IDO may be a new strategy for breast cancer treatment.

Additional files

Acknowledgements

We deeply thank Dr. Wu KK for his continuous support and encouragement. We also thank Ms. Pei-Yung Nien for the isolation of primary CAFs and Dr. Delphine Allorge and Dr. Marion Soichot for providing human IDO promoter. This study was supported by grants from National Science Council (NSC 101-2321-B-400-003 and NSC 102-2321-B-400-003) and MOHW103-TD-B-111-05 from Excellence for Cancer Research Center Grant, the Ministry of Health and Welfare.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

JY conducted cell culture, biochemical, and molecular biology assays and prepared the draft of the manuscript. CF did IHC study and pathological analysis and helped to draft the manuscript. CC performed metabolomics study and data analysis and helped to draft the manuscript. KK participated in the design of the study and helped to draft the manuscript. MF conceived of the study and participated in the preparation of primary cancer-associated fibroblasts. WC conceived of the study and participated in the design of the study and wrote the manuscript. All authors read and approved the final manuscript.
Anhänge
Literatur
1.
Zurück zum Zitat Williams CS, Mann M, DuBois RN: The role of cyclooxygenases in inflammation, cancer, and development. Oncogene. 1999, 18: 7908-7916.CrossRefPubMed Williams CS, Mann M, DuBois RN: The role of cyclooxygenases in inflammation, cancer, and development. Oncogene. 1999, 18: 7908-7916.CrossRefPubMed
2.
Zurück zum Zitat Terzic J, Grivennikov S, Karin E, Karin M: Inflammation and colon cancer. Gastroenterology. 2010, 138: 2101-2114. e2105CrossRefPubMed Terzic J, Grivennikov S, Karin E, Karin M: Inflammation and colon cancer. Gastroenterology. 2010, 138: 2101-2114. e2105CrossRefPubMed
4.
Zurück zum Zitat Ristimaki A, Sivula A, Lundin J, Lundin M, Salminen T, Haglund C, Joensuu H, Isola J: Prognostic significance of elevated cyclooxygenase-2 expression in breast cancer. Cancer Res. 2002, 62: 632-635.PubMed Ristimaki A, Sivula A, Lundin J, Lundin M, Salminen T, Haglund C, Joensuu H, Isola J: Prognostic significance of elevated cyclooxygenase-2 expression in breast cancer. Cancer Res. 2002, 62: 632-635.PubMed
5.
Zurück zum Zitat Wulfing P, Diallo R, Muller C, Wulfing C, Poremba C, Heinecke A, Rody A, Greb RR, Bocker W, Kiesel L: Analysis of cyclooxygenase-2 expression in human breast cancer: high throughput tissue microarray analysis. J Cancer Res Clin Oncol. 2003, 129: 375-382.CrossRefPubMed Wulfing P, Diallo R, Muller C, Wulfing C, Poremba C, Heinecke A, Rody A, Greb RR, Bocker W, Kiesel L: Analysis of cyclooxygenase-2 expression in human breast cancer: high throughput tissue microarray analysis. J Cancer Res Clin Oncol. 2003, 129: 375-382.CrossRefPubMed
6.
Zurück zum Zitat Davies G, Salter J, Hills M, Martin LA, Sacks N, Dowsett M: Correlation between cyclooxygenase-2 expression and angiogenesis in human breast cancer. Clin Cancer Res. 2003, 9: 2651-2656.PubMed Davies G, Salter J, Hills M, Martin LA, Sacks N, Dowsett M: Correlation between cyclooxygenase-2 expression and angiogenesis in human breast cancer. Clin Cancer Res. 2003, 9: 2651-2656.PubMed
7.
Zurück zum Zitat Egan KM, Stampfer MJ, Giovannucci E, Rosner BA, Colditz GA: Prospective study of regular aspirin use and the risk of breast cancer. J Natl Cancer Inst. 1996, 88: 988-993.CrossRefPubMed Egan KM, Stampfer MJ, Giovannucci E, Rosner BA, Colditz GA: Prospective study of regular aspirin use and the risk of breast cancer. J Natl Cancer Inst. 1996, 88: 988-993.CrossRefPubMed
8.
Zurück zum Zitat Arun B, Goss P: The role of COX-2 inhibition in breast cancer treatment and prevention. Semin Oncol. 2004, 31: 22-29.CrossRefPubMed Arun B, Goss P: The role of COX-2 inhibition in breast cancer treatment and prevention. Semin Oncol. 2004, 31: 22-29.CrossRefPubMed
9.
Zurück zum Zitat Chang SH, Liu CH, Conway R, Han DK, Nithipatikom K, Trifan OC, Lane TF, Hla T: Role of prostaglandin E2-dependent angiogenic switch in cyclooxygenase 2-induced breast cancer progression. Proc Natl Acad Sci U S A. 2004, 101: 591-596.CrossRefPubMed Chang SH, Liu CH, Conway R, Han DK, Nithipatikom K, Trifan OC, Lane TF, Hla T: Role of prostaglandin E2-dependent angiogenic switch in cyclooxygenase 2-induced breast cancer progression. Proc Natl Acad Sci U S A. 2004, 101: 591-596.CrossRefPubMed
10.
Zurück zum Zitat Howe LR, Subbaramaiah K, Patel J, Masferrer JL, Deora A, Hudis C, Thaler HT, Muller WJ, Du B, Brown AM, Dannenberg AJ: Celecoxib, a selective cyclooxygenase 2 inhibitor, protects against human epidermal growth factor receptor 2 (HER-2)/neu-induced breast cancer. Cancer Res. 2002, 62: 5405-5407.PubMed Howe LR, Subbaramaiah K, Patel J, Masferrer JL, Deora A, Hudis C, Thaler HT, Muller WJ, Du B, Brown AM, Dannenberg AJ: Celecoxib, a selective cyclooxygenase 2 inhibitor, protects against human epidermal growth factor receptor 2 (HER-2)/neu-induced breast cancer. Cancer Res. 2002, 62: 5405-5407.PubMed
11.
Zurück zum Zitat Harris RE, Alshafie GA, Abou-Issa H, Seibert K: Chemoprevention of breast cancer in rats by celecoxib, a cyclooxygenase 2 inhibitor. Cancer Res. 2000, 60: 2101-2103.PubMed Harris RE, Alshafie GA, Abou-Issa H, Seibert K: Chemoprevention of breast cancer in rats by celecoxib, a cyclooxygenase 2 inhibitor. Cancer Res. 2000, 60: 2101-2103.PubMed
12.
Zurück zum Zitat Aboussekhra A: Role of cancer-associated fibroblasts in breast cancer development and prognosis. Int J Dev Biol. 2011, 55: 841-849.CrossRefPubMed Aboussekhra A: Role of cancer-associated fibroblasts in breast cancer development and prognosis. Int J Dev Biol. 2011, 55: 841-849.CrossRefPubMed
13.
Zurück zum Zitat Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA: Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005, 121: 335-348.CrossRefPubMed Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA: Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005, 121: 335-348.CrossRefPubMed
14.
Zurück zum Zitat Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H, Porter D, Hu M, Chin L, Richardson A, Schnitt S, Sellers WR, Polyak K: Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell. 2004, 6: 17-32.CrossRefPubMed Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H, Porter D, Hu M, Chin L, Richardson A, Schnitt S, Sellers WR, Polyak K: Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell. 2004, 6: 17-32.CrossRefPubMed
15.
Zurück zum Zitat Hu M, Peluffo G, Chen H, Gelman R, Schnitt S, Polyak K: Role of COX-2 in epithelial-stromal cell interactions and progression of ductal carcinoma in situ of the breast. Proc Natl Acad Sci U S A. 2009, 106: 3372-3377.CrossRefPubMedPubMedCentral Hu M, Peluffo G, Chen H, Gelman R, Schnitt S, Polyak K: Role of COX-2 in epithelial-stromal cell interactions and progression of ductal carcinoma in situ of the breast. Proc Natl Acad Sci U S A. 2009, 106: 3372-3377.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Kuperwasser C, Chavarria T, Wu M, Magrane G, Gray JW, Carey L, Richardson A, Weinberg RA: Reconstruction of functionally normal and malignant human breast tissues in mice. Proc Natl Acad Sci U S A. 2004, 101: 4966-4971.CrossRefPubMedPubMedCentral Kuperwasser C, Chavarria T, Wu M, Magrane G, Gray JW, Carey L, Richardson A, Weinberg RA: Reconstruction of functionally normal and malignant human breast tissues in mice. Proc Natl Acad Sci U S A. 2004, 101: 4966-4971.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Cheng HH, Kuo CC, Yan JL, Chen HL, Lin WC, Wang KH, Tsai KK, Guvén H, Flaberg E, Szekely L, Klein G, Wu KK: Control of cyclooxygenase-2 expression and tumorigenesis by endogenous 5-methoxytryptophan. Proc Natl Acad Sci U S A. 2012, 109: 13231-13236.CrossRefPubMedPubMedCentral Cheng HH, Kuo CC, Yan JL, Chen HL, Lin WC, Wang KH, Tsai KK, Guvén H, Flaberg E, Szekely L, Klein G, Wu KK: Control of cyclooxygenase-2 expression and tumorigenesis by endogenous 5-methoxytryptophan. Proc Natl Acad Sci U S A. 2012, 109: 13231-13236.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Wishart DS, Knox C, Guo AC, Eisner R, Young N, Gautam B, Hau DD, Psychogios N, Dong E, Bouatra S, Mandal R, Sinelnikov I, Xia J, Jia L, Cruz JA, Lim E, Sobsey CA, Shrivastava S, Huang P, Liu P, Fang L, Peng J, Fradette R, Cheng D, Tzur D, Clements M, Lewis A, De Souza A, Zuniga A, Dawe M, et al: HMDB: a knowledge base for the human metabolome. Nucleic Acid Res. 2009, 37: D603-610.CrossRefPubMed Wishart DS, Knox C, Guo AC, Eisner R, Young N, Gautam B, Hau DD, Psychogios N, Dong E, Bouatra S, Mandal R, Sinelnikov I, Xia J, Jia L, Cruz JA, Lim E, Sobsey CA, Shrivastava S, Huang P, Liu P, Fang L, Peng J, Fradette R, Cheng D, Tzur D, Clements M, Lewis A, De Souza A, Zuniga A, Dawe M, et al: HMDB: a knowledge base for the human metabolome. Nucleic Acid Res. 2009, 37: D603-610.CrossRefPubMed
19.
Zurück zum Zitat Chan CH, Li CF, Yang WL, Gao Y, Lee SW, Feng Z, Huang HY, Tsai KK, Flores LG, Shao Y, Hazle JD, Yu D, Wei W, Sarbassov D, Hung MC, Nakayama KI, Lin HK: The Skp2-SCF E3 ligase regulates Akt ubiquitination, glycolysis, herceptin sensitivity, and tumorigenesis. Cell. 2012, 149: 1098-1111.CrossRefPubMedPubMedCentral Chan CH, Li CF, Yang WL, Gao Y, Lee SW, Feng Z, Huang HY, Tsai KK, Flores LG, Shao Y, Hazle JD, Yu D, Wei W, Sarbassov D, Hung MC, Nakayama KI, Lin HK: The Skp2-SCF E3 ligase regulates Akt ubiquitination, glycolysis, herceptin sensitivity, and tumorigenesis. Cell. 2012, 149: 1098-1111.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Horai H, Arita M, Kanaya S, Nihei Y, Ikeda T, Suwa K, Ojima Y, Tanaka K, Tanaka S, Aoshima K, Oda Y, Kakazu Y, Kusano M, Tohge T, Matsuda F, Sawada Y, Hirai MY, Nakanishi H, Ikeda K, Akimoto N, Maoka T, Takahashi H, Ara T, Sakurai N, Suzuki H, Shibata D, Neumann S, Iida T, Tanaka K, Funatsu K, et al: MassBank: a public repository for sharing mass spectral data for life sciences. J Mass Spectrom. 2010, 45: 703-714.CrossRefPubMed Horai H, Arita M, Kanaya S, Nihei Y, Ikeda T, Suwa K, Ojima Y, Tanaka K, Tanaka S, Aoshima K, Oda Y, Kakazu Y, Kusano M, Tohge T, Matsuda F, Sawada Y, Hirai MY, Nakanishi H, Ikeda K, Akimoto N, Maoka T, Takahashi H, Ara T, Sakurai N, Suzuki H, Shibata D, Neumann S, Iida T, Tanaka K, Funatsu K, et al: MassBank: a public repository for sharing mass spectral data for life sciences. J Mass Spectrom. 2010, 45: 703-714.CrossRefPubMed
21.
Zurück zum Zitat Soichot M, Hennart B, Al Saabi A, Leloire A, Froguel P, Levy-Marchal C, Poulain-Godefroy O, Allorge D: Identification of a variable number of tandem repeats polymorphism and characterization of LEF-1 response elements in the promoter of the IDO1 gene. PLoS One. 2011, 6: e25470-CrossRefPubMedPubMedCentral Soichot M, Hennart B, Al Saabi A, Leloire A, Froguel P, Levy-Marchal C, Poulain-Godefroy O, Allorge D: Identification of a variable number of tandem repeats polymorphism and characterization of LEF-1 response elements in the promoter of the IDO1 gene. PLoS One. 2011, 6: e25470-CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Decker T, Kovarik P, Meinke A: GAS elements: a few nucleotides with a major impact on cytokine-induced gene expression. J Interferon Cytokine Res. 1997, 17: 121-134.CrossRefPubMed Decker T, Kovarik P, Meinke A: GAS elements: a few nucleotides with a major impact on cytokine-induced gene expression. J Interferon Cytokine Res. 1997, 17: 121-134.CrossRefPubMed
23.
Zurück zum Zitat Du MX, Sotero-Esteva WD, Taylor MW: Analysis of transcription factors regulating induction of indoleamine 2,3-dioxygenase by IFN-gamma. J Interferon Cytokine Res. 2000, 20: 133-142.CrossRefPubMed Du MX, Sotero-Esteva WD, Taylor MW: Analysis of transcription factors regulating induction of indoleamine 2,3-dioxygenase by IFN-gamma. J Interferon Cytokine Res. 2000, 20: 133-142.CrossRefPubMed
24.
Zurück zum Zitat Sumpter TL, Dangi A, Matta BM, Huang C, Stolz DB, Vodovotz Y, Thomson AW, Gandhi CR: Hepatic stellate cells undermine the allostimulatory function of liver myeloid dendritic cells via STAT3-dependent induction of IDO. J Immunol. 2012, 189: 3848-3858.CrossRefPubMedPubMedCentral Sumpter TL, Dangi A, Matta BM, Huang C, Stolz DB, Vodovotz Y, Thomson AW, Gandhi CR: Hepatic stellate cells undermine the allostimulatory function of liver myeloid dendritic cells via STAT3-dependent induction of IDO. J Immunol. 2012, 189: 3848-3858.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, Schumacher T, Jestaedt L, Schrenk D, Weller M, Jugold M, Guillemin GJ, Miller CL, Lutz C, Radlwimmer B, Lehmann I, von Deimling A, Wick W, Platten M: An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature. 2011, 478: 197-203.CrossRefPubMed Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, Schumacher T, Jestaedt L, Schrenk D, Weller M, Jugold M, Guillemin GJ, Miller CL, Lutz C, Radlwimmer B, Lehmann I, von Deimling A, Wick W, Platten M: An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature. 2011, 478: 197-203.CrossRefPubMed
26.
Zurück zum Zitat Ohtake F, Baba A, Takada I, Okada M, Iwasaki K, Miki H, Takahashi S, Kouzmenko A, Nohara K, Chiba T, Fujii-Kuriyama Y, Kato S: Dioxin receptor is a ligand-dependent E3 ubiquitin ligase. Nature. 2007, 446: 562-566.CrossRefPubMed Ohtake F, Baba A, Takada I, Okada M, Iwasaki K, Miki H, Takahashi S, Kouzmenko A, Nohara K, Chiba T, Fujii-Kuriyama Y, Kato S: Dioxin receptor is a ligand-dependent E3 ubiquitin ligase. Nature. 2007, 446: 562-566.CrossRefPubMed
27.
Zurück zum Zitat Mansfield AS, Heikkila PS, Vaara AT, von Smitten KA, Vakkila JM, Leidenius MH: Simultaneous Foxp3 and IDO expression is associated with sentinel lymph node metastases in breast cancer. BMC Cancer. 2009, 9: 231-CrossRefPubMedPubMedCentral Mansfield AS, Heikkila PS, Vaara AT, von Smitten KA, Vakkila JM, Leidenius MH: Simultaneous Foxp3 and IDO expression is associated with sentinel lymph node metastases in breast cancer. BMC Cancer. 2009, 9: 231-CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB: Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2, 3-dioxygenase. J Exp Med. 2002, 196: 459-468.CrossRefPubMedPubMedCentral Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB: Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2, 3-dioxygenase. J Exp Med. 2002, 196: 459-468.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Muller AJ, DuHadaway JB, Donover PS, Sutanto-Ward E, Prendergast GC: Inhibition of indoleamine 2, 3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat Med. 2005, 11: 312-319.CrossRefPubMed Muller AJ, DuHadaway JB, Donover PS, Sutanto-Ward E, Prendergast GC: Inhibition of indoleamine 2, 3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat Med. 2005, 11: 312-319.CrossRefPubMed
30.
Zurück zum Zitat Yasui H, Takai K, Yoshida R, Hayaishi O: Interferon enhances tryptophan metabolism by inducing pulmonary indoleamine 2,3-dioxygenase: its possible occurrence in cancer patients. Proc Natl Acad Sci U S A. 1986, 83: 6622-6626.CrossRefPubMedPubMedCentral Yasui H, Takai K, Yoshida R, Hayaishi O: Interferon enhances tryptophan metabolism by inducing pulmonary indoleamine 2,3-dioxygenase: its possible occurrence in cancer patients. Proc Natl Acad Sci U S A. 1986, 83: 6622-6626.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Pan MR, Hou MF, Chang HC, Hung WC: Cyclooxygenase-2 up-regulates CCR7 via EP2/EP4 receptor signaling pathways to enhance lymphatic invasion of breast cancer cells. J Biol Chem. 2008, 283: 11155-11163.CrossRefPubMed Pan MR, Hou MF, Chang HC, Hung WC: Cyclooxygenase-2 up-regulates CCR7 via EP2/EP4 receptor signaling pathways to enhance lymphatic invasion of breast cancer cells. J Biol Chem. 2008, 283: 11155-11163.CrossRefPubMed
32.
Zurück zum Zitat Gross S, Cairns RA, Minden MD, Driggers EM, Bittinger MA, Jang HG, Sasaki M, Jin S, Schenkein DP, Su SM, Dang L, Fantin VR, Mak TW: Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med. 2010, 207: 339-344.CrossRefPubMedPubMedCentral Gross S, Cairns RA, Minden MD, Driggers EM, Bittinger MA, Jang HG, Sasaki M, Jin S, Schenkein DP, Su SM, Dang L, Fantin VR, Mak TW: Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med. 2010, 207: 339-344.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Ward PS, Patel J, Wise DR, Abdel-Wahab O, Bennett BD, Coller HA, Cross JR, Fantin VR, Hedvat CV, Perl AE, Rabinowitz JD, Carroll M, Su SM, Sharp KA, Levine RL, Thompson CB: The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. 2010, 17: 225-234.CrossRefPubMedPubMedCentral Ward PS, Patel J, Wise DR, Abdel-Wahab O, Bennett BD, Coller HA, Cross JR, Fantin VR, Hedvat CV, Perl AE, Rabinowitz JD, Carroll M, Su SM, Sharp KA, Levine RL, Thompson CB: The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. 2010, 17: 225-234.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF, Tallman MS, Sun Z, Wolniak K, Peeters JK, Liu W, Choe SE, Fantin VR, Paietta E, Lowenberg B, Licht JD, Godley LA, Delwel R, Valk PJ, Thompson CB, Levine RL, Melnick A: Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010, 18: 553-567.CrossRefPubMedPubMedCentral Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF, Tallman MS, Sun Z, Wolniak K, Peeters JK, Liu W, Choe SE, Fantin VR, Paietta E, Lowenberg B, Licht JD, Godley LA, Delwel R, Valk PJ, Thompson CB, Levine RL, Melnick A: Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010, 18: 553-567.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Wang P, Xiao MT, Liu LX, Jiang WQ, Liu J, Zhang JY, Wang B, Frye S, Zhang Y, Xu YH, Lei QY, Guan KL, Zhao SM, Xiong Y: Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell. 2011, 19: 17-30.CrossRefPubMedPubMedCentral Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Wang P, Xiao MT, Liu LX, Jiang WQ, Liu J, Zhang JY, Wang B, Frye S, Zhang Y, Xu YH, Lei QY, Guan KL, Zhao SM, Xiong Y: Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell. 2011, 19: 17-30.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Platten M, Wick W, Van den Eynde BJ: Tryptophan catabolism in cancer: beyond IDO and tryptophan depletion. Cancer Res. 2012, 72: 5435-5440.CrossRefPubMed Platten M, Wick W, Van den Eynde BJ: Tryptophan catabolism in cancer: beyond IDO and tryptophan depletion. Cancer Res. 2012, 72: 5435-5440.CrossRefPubMed
37.
Zurück zum Zitat West RB, Nuyten DS, Subramanian S, Nielsen TO, Corless CL, Rubin BP, Montgomery K, Zhu S, Patel R, Hernandez-Boussard T, Goldblum JR, Brown PO, van de Vijver M, van de Rijn M: Determination of stromal signatures in breast carcinoma. PLoS Biol. 2005, 3: e187-CrossRefPubMedPubMedCentral West RB, Nuyten DS, Subramanian S, Nielsen TO, Corless CL, Rubin BP, Montgomery K, Zhu S, Patel R, Hernandez-Boussard T, Goldblum JR, Brown PO, van de Vijver M, van de Rijn M: Determination of stromal signatures in breast carcinoma. PLoS Biol. 2005, 3: e187-CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Chen JL, Espinosa I, Lin AY, Liao OY, van de Rijn M, West RB: Stromal responses among common carcinomas correlated with clinicopathologic features. Clin Cancer Res. 2013, 19: 5127-5135.CrossRefPubMedPubMedCentral Chen JL, Espinosa I, Lin AY, Liao OY, van de Rijn M, West RB: Stromal responses among common carcinomas correlated with clinicopathologic features. Clin Cancer Res. 2013, 19: 5127-5135.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Mercier I, Casimiro MC, Wang C, Rosenberg AL, Quong J, Minkeu A, Allen KG, Danilo C, Sotgia F, Bonuccelli G, Jasmin JF, Xu H, Bosco E, Aronow B, Witkiewicz A, Pestell RG, Knudsen ES, Lisanti MP: Human breast cancer-associated fibroblasts (CAFs) show caveolin-1 downregulation and RB tumor suppressor functional inactivation: implications for the response to hormonal therapy. Cancer Biol Ther. 2008, 7: 1212-1225.CrossRefPubMed Mercier I, Casimiro MC, Wang C, Rosenberg AL, Quong J, Minkeu A, Allen KG, Danilo C, Sotgia F, Bonuccelli G, Jasmin JF, Xu H, Bosco E, Aronow B, Witkiewicz A, Pestell RG, Knudsen ES, Lisanti MP: Human breast cancer-associated fibroblasts (CAFs) show caveolin-1 downregulation and RB tumor suppressor functional inactivation: implications for the response to hormonal therapy. Cancer Biol Ther. 2008, 7: 1212-1225.CrossRefPubMed
40.
Zurück zum Zitat Erez N, Truitt M, Olson P, Arron ST, Hanahan D: Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 2010, 17: 135-147.CrossRefPubMed Erez N, Truitt M, Olson P, Arron ST, Hanahan D: Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 2010, 17: 135-147.CrossRefPubMed
Metadaten
Titel
Cancer/stroma interplay via cyclooxygenase-2 and indoleamine 2,3-dioxygenase promotes breast cancer progression
verfasst von
Jing-Yi Chen
Chien-Feng Li
Cheng-Chin Kuo
Kelvin K Tsai
Ming-Feng Hou
Wen-Chun Hung
Publikationsdatum
01.08.2014
Verlag
BioMed Central
Erschienen in
Breast Cancer Research / Ausgabe 4/2014
Elektronische ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-014-0410-1

Weitere Artikel der Ausgabe 4/2014

Breast Cancer Research 4/2014 Zur Ausgabe

Mammakarzinom: Brustdichte beeinflusst rezidivfreies Überleben

26.05.2024 Mammakarzinom Nachrichten

Frauen, die zum Zeitpunkt der Brustkrebsdiagnose eine hohe mammografische Brustdichte aufweisen, haben ein erhöhtes Risiko für ein baldiges Rezidiv, legen neue Daten nahe.

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Das sind die führenden Symptome junger Darmkrebspatienten

Darmkrebserkrankungen in jüngeren Jahren sind ein zunehmendes Problem, das häufig längere Zeit übersehen wird, gerade weil die Patienten noch nicht alt sind. Welche Anzeichen Ärzte stutzig machen sollten, hat eine Metaanalyse herausgearbeitet.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.