Androgens enhance the ability of intratumoral macrophages to promote breast cancer progression

  • Authors:
    • Mio Yamaguchi
    • Kiyoshi Takagi
    • Masayasu Sato
    • Ai Sato
    • Yasuhiro Miki
    • Yoshiaki Onodera
    • Minoru Miyashita
    • Hironobu Sasano
    • Takashi Suzuki
  • View Affiliations

  • Published online on: July 12, 2021     https://doi.org/10.3892/or.2021.8139
  • Article Number: 188
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Androgens are produced locally in breast carcinoma tissues by androgen‑producing enzymes such as 5α‑reductase type 1 (5αRed1) and affect not only breast cancer cells but the tumor microenvironment as well. Tumor‑associated macrophages (TAMs) are primary components of the tumor microenvironment and contribute to tumor progression. Although previous studies suggest that androgen/androgen receptor (AR) signaling in macrophages has important roles in human diseases, androgen action on TAMs has remained largely unknown. We immunolocalized macrophage marker CD163 as well as AR and 5αRed1 in 116 breast carcinomas and correlated them with clinicopathological parameters and clinical outcomes. Moreover, we examined the roles of androgens on macrophages in breast cancer progression using cell lines 4T1 (mouse breast cancer) and RAW264.7 (macrophage) in a tumor‑bearing female BALB/c mouse model. Double immunohistochemistry revealed that AR was sporadically expressed in the macrophages in breast carcinoma tissues. Macrophage infiltration was significantly correlated with an aggressive phenotype of breast carcinomas and worse prognosis, especially in the 5αRed1‑positive group. In a sphere‑forming assay using 4T1 and RAW‑AR cells, which stably express AR, the sphere size was significantly increased due to androgens when 4T1 cells were cocultured with RAW‑AR cells. Furthermore, in vivo experiments revealed that tumor growth and Ki67, a cell proliferation marker, were increased when androgens were stably produced in breast cancer cells and AR was expressed in macrophages. In conclusion, AR is expressed in intratumoral macrophages and is associated with an aggressive phenotype of breast carcinomas, especially when breast cancer cells actively produce androgens. Thus, androgens may enhance the ability of macrophages to promote breast cancer progression.

Introduction

Biologically active steroids such as estrogens and androgens are locally produced in breast cancer tissues by sex-steroid producing enzymes. Among these enzymes, aromatase is an important therapeutic target in estrogen receptor (ER)-positive breast cancers. However, de novo or acquired resistance to aromatase inhibitors (AIs) are often experienced and this is a key problem to be solved (1). On the other hand, androgen-producing enzymes such as 17β-hydroxysteroid dehydrogenase type 5 [17βHSD5, also known as aldo-keto reductase family 1 member C3 (AKR1C3); androstenedione to testosterone] and 5α-reductase type 1 [5αRed1; testosterone to dihydrotestosterone (DHT), biologically active androgens] are also frequently expressed in breast carcinoma tissues. These enzymes contribute to intratumoral androgen synthesis (2) and 5αRed1 is known as a potent regulator of in situ DHT production (3). Because the androgen receptor (AR) is also frequently expressed in breast cancer cells (4,5), it has been considered that intratumoral androgens have pivotal roles in the progression of breast cancers. However, the significance of androgen in breast cancer has not been fully understood and opposite findings have often been reported. For instance, numerous studies have pointed out that AR expression is associated with better clinical outcomes (6), while androgen action was recently highlighted in a distinct subset of ER-negative breast cancers [namely molecular apocrine subtype or luminal AR subtype (7,8)] or ER-positive breast cancers which have become resistant to AIs (9). It is therefore important to study the action of androgens in breast cancer.

The tumor microenvironment is composed not only of tumor cells but also contains various stromal cells including macrophages, leukocytes and fibroblasts, which, together with cross-talks between tumor cells and other cellular components, is strongly associated with tumor progression (10). Tumor-associated macrophages (TAMs) are well known as the primary components of the tumor microenvironment and promote cancer progression (1116). Monocytes in the blood are attracted to tumor tissues via soluble factors released from tumor tissues, where they differentiate into TAMs (17). Based on their function, macrophages are classified into M1 or M2 phenotypes. M1 macrophages are driven by cytokines such as tumor necrosis factor α (TNFα) and interferon γ (IFNγ), while M2 macrophages are driven by interleukin (IL)-4 and IL-13 (18). Each macrophage type displays different functions and expression profiles of cytokines and cell-surface markers. While M1 macrophages express CD80 and CD86 and have tumor-suppressive and pro-inflammatory effects, M2 macrophages express CD163, are the predominant phenotype of TAMs in human solid tumors (18,19) and contribute to tumor malignancy by promoting tumor growth, metastasis, angiogenesis, epithelial-mesenchymal transition (EMT) and drug resistance in breast cancers (1116). Previous studies have reported that a high infiltration of M2 macrophages is correlated with lymph node metastasis, higher histological grade and cell proliferation, and contributes to worse prognosis in breast cancers (13,19).

It has been reported that AR in monocytes/macrophages plays key roles in the greater prevalence and severity of atherosclerosis in men (20,21). Moreover, it has also been reported that some macrophages express AR and that androgen/AR signaling in macrophages inhibits cutaneous wound healing (22). These findings suggest that androgen acts upon macrophages affecting various human diseases, although its role in human tumors, including breast cancers, remains unclear. We therefore focused on the relationship between androgen action in intratumoral macrophages and progression of breast cancers, with the purpose of a better understanding of the biological and/or clinical significance of androgens in breast cancers.

Materials and methods

Patients and tissues

A total of 116 specimens of invasive breast carcinomas were obtained from Japanese female patients who had undergone surgical treatment from 2007 to 2008 at Tohoku University Hospital, Sendai, Japan. The clinical outcome was evaluated by disease-free and breast cancer-specific survival. Disease-free survival was defined as the time from the date of surgery to that of the first locoregional recurrence or distant metastasis within the follow-up time and the median was 59 (range 3–84) months. Breast cancer-specific survival was defined as the time from surgery to death from breast cancer and follow-up time was 61 (from 3–84) months. All specimens had been fixed with 10% formalin neutral buffer solution and embedded in paraffin wax. Experiments and analyses were performed in accordance with the Helsinki declaration and the research protocol of this study was approved by the Ethics Committee at the Tohoku University Graduate School of Medicine (approval no. 2016-1-697, 2019-1-219). This is a retrospective study and, therefore, informed consents had not been obtained.

Immunohistochemistry

The information regarding the antibodies is listed in Table SI. Immunohistochemistry for CD163, 5αRed1 and Ki67 was carried out using a Histofine kit (Nichirei Bio, Inc., Tokyo, Japan) as described previously (23,24). The antigen-antibody complex was visualized with 3,3′-diaminobenzidine (DAB) and hematoxylin was used for counterstaining. Immunohistochemistry for ER and progesterone receptor (PR) was carried out with Ventana Benchmark XT system (Roche Diagnostics Japan). For human epidermal growth factor receptor 2 (HER2), the HercepTest (Dako) was used.

Double immunohistochemistry

Immunoreactivity of AR was firstly visualized with DAB as described above, and then antigen retrieval was carried out again and immunoreactivity of CD163 was visualized with nitro blue tetrazolium (NBT)/5-bromo-4-chloro-3-indolyl phosphate (BCIP).

Scoring of immunoreactivity

5αRed1 immunoreactivity was detected in the cytoplasm of breast carcinoma cells and considered positive when the cases had more than 10% of positive carcinoma cells (3). Infiltration of CD163-positive macrophages was scored into 4 categories (0, no focal areas; 1, small scattered focal areas; 2, numerous larger focal areas; 3, very numerous large focal areas) according to a previous study (25), and finally dichotomized for statistical analyses (0, 1: Low infiltration and 2, 3: High infiltration). ER, PR and Ki67 immunoreactivity were detected in the nucleus of carcinoma cells and the percentage of positive cells (labeling index; LI) was calculated by evaluating in more than 1,000 cells. According to a previous report (26), cases with LI of more than 1% were considered positive for ER and PR. HER2 immunoreactivity was determined according to the grading system proposed in HercepTest (Dako).

Cell lines and chemicals

Mouse breast cancer cell line 4T1 and mouse macrophage cell line RAW264.7 were obtained from American Type Culture Collection (ATCC) and RIKEN Bioresource Center (Japan), respectively. These cells were cultured in RPMI-1640 medium (Sigma-Aldrich; Merck KGaA) with 10% fetal bovine serum (FBS) (Biosera) and incubated at 37°C under 5% CO2. In experiments using androgen, these cells were cultured in phenol red-free RPMI-1640 with 10% dextran-coated charcoal-stripped FBS.

Coculture experiment

The 4T1 cells were cocultured with RAW264.7 cells using ThinCerts™ (pore size 0.4 µm, Greiner bio-one). RAW264.7 cells were seeded in the bottom plate (3×105 cells/well) and allowed to attach for 24 h. Then, the 4T1 cells were placed onto an upper Transwell (2×105 cells/well) and cocultured with RAW264.7 cells for 3 days.

Quantitative real-time PCR

Total RNA (500 ng) was extracted using TRI Reagent (Molecular Research Center, Inc.) and cDNA was synthesized using a ReverTra Ace qPCR RT Master Mix with gDNA Remover (Toyobo Co. Ltd.). Real-time PCR was performed using the THUNDERBIRD™ SYBR® qPCR Mix (Toyobo Co. Ltd.) and LightCycler nano system (Roche Diagnostics Japan). Thermocycling conditions were as follows: 95°C for 60 sec (initial denaturation), followed by 45 cycles at 95°C for 15 sec and 60°C for 30 sec. The sequences for the PCR primer sets are listed in Table SII. Relative mRNA levels of Arg-1, Ar, Srd5a1 and Akr1c6 were summarized as the ratio to Rpl13a.

Plasmid construction

Open reading frame of Ar and FLAG-tagged aldo-keto reductase family 1 member C6 (Akr1c6, mouse testosterone-producing enzyme) were amplified via PCR using KOD FX Neo (Toyobo Co. Ltd.) and cloned into pIRES2-AcGFP1 vector (Clontech Laboratories) and pcDNA3.1 (−) vector (Invitrogen; Thermo Fisher Scientific, Inc.), respectively, using DNA Ligation Kit <Mighty Mix> (Takara Bio) (pIRES-mAR and pAKR1C6-FLAG). Thermocycling conditions were as follows: 94°C for 2 min, followed by 35 cycles at 98°C for 10 sec, 62°C for 30 sec and 68°C for 90 sec. The sequences for the PCR primer sets are listed in Table SII.

Plasmid transfection and establishment of stable clones

Since 4T1 and RAW264,7 cells do not express Akr1c6 and Ar, respectively, we tried to establish their sublines which stably express these genes in order to investigate the roles of androgens in intratumoral macrophages. According to the manufacturer's instructions for transfection, RAW264.7 cells were transfected with pIRES-mAR or pIRES2-AcGFP1 using GenomONE™-Neo (Ishihara Sangyo Kaisya, Ltd.) and 4T1 cells were transfected with pAKR1C6-FLAG (4T1-AKR1C6) or pcDNA3.1(−) (4T1-CT) using Avalanche®-Everyday Transfection Reagent (APRO Science). These cells were cultured in media containing 400 µg/ml G418 (Wako), and stable clones expressing AR (RAW-AR), AKR1C6 (4T1-AKR1C6) and corresponding negative control clones (RAW-CT and 4T1-CT) were established.

Western blotting

Western blotting was performed according to previous reports (27,28) and the information regarding the primary antibodies is listed in Table SI. The cells were lysed using M-PER Mammalian Protein Extraction Reagent (Pierce Biotechnology) containing Halt Protease Inhibitor Cocktail (Sigma-Aldrich; Merck KGaA). The protein extracts (10 µg) were separated by SDS-PAGE (10% acrylamide gel) and then transferred to Hybond P polyvinylidene difluoride membrane (GE Healthcare). The membrane was blocked in 5% non-fat milk in TBS-T for 1 h at room temperature and then incubated with primary antibodies at 4°C overnight. The blots were incubated with the appropriate HRP-conjugated secondary antibody (anti-mouse; 1:10,000, cat. no. NA931 or anti-rabbit; 1:10,000, cat. no. NA934, GE Healthcare) for 1 h at room temperature. Detection of antibody-protein complexes on the membrane was visualized using ECL-Prime Western blotting detection reagents (GE Healthcare) and LAS-4000 image analyzer (Fuji Photo Film Co.).

Synthetic androgen R1881 treatment

RAW-CT and RAW-AR cells (1×105 cells/well) were cultured in a 12-well plate with or without synthetic androgen R1881 (10 nM) for 48 h.

Sphere-forming assay

A sphere-forming assay was performed using EZ-BindShut®II micro plate (IWAKI). The 4T1 cells (1×105 cells/well) were cocultured in a well with RAW-CT or RAW-AR cells (3.33×104 cells/well) with or without R1881 (10 nM). After 3 days, spheres were photographed under a microscope and the size of 100 spheres was measured using ImageJ 1.53e (https://imagej.nih.gov/ij/).

Cell proliferation assay

The 4T1-AKR1C6 or 4T1-CT cells were seeded in a 96-well plate (3,000 cells/well) and the cell proliferation was measured by the WST-8 method using a Cell Counting Kit-8 (Dojindo Molecular Technologies) for 4 days. Absorbance was determined using a Bio-Rad iMark plate reader (Bio-Rad Laboratories Inc.).

Tumor-bearing mouse model

The protocols of all animal experiments were submitted, reviewed and approved in advance by the Institutional Animal Care and Use Committee at Tohoku University (2019MdA-172). The experiments were carried out according to ARRIVE guidelines and Guidelines for Animal Experiments at Tohoku University. A total of 11 4-week-old wild-type female BALB/c mice were obtained from CLEA Japan (Tokyo, Japan) and randomly assigned to two types of tumor-bearing models as described below with no inclusion criteria. They were housed in a specific pathogen-free facility with free access to water and MR stock food and environmental enrichment. In the present study, 4T1 and RAW264.7 sublines were subcutaneously injected in both the left and right side of mice in different combinations in order to avoid individual differences. In the first experiment (n=6), 4T1-CT or 4T1-AKR1C6 cells (5×105 cells) were subcutaneously injected together with RAW-AR cells (1.25×105 cells) into the left or right side of the back of mice, respectively, using 150 µl Matrigel (Corning, Inc.). In the mirror experiment (n=5), 4T1-AKR1C6 cells (5×105 cells) were injected together with either RAW-CT or RAW-AR cells (1.25×105 cells) into the left or right side of the back of mice, respectively. Mice were monitored for body weight and tumor volume changes twice a week. Tumor volume was measured using a digital caliper at least twice a week and calculated by modified ellipsoid formula 1/2 (length × width2). Tumors were allowed to grow in mice until they reached the volume of 2,000 mm3 or 10 weeks from injection had elapsed, whichever came first. No adverse events were observed and mice were euthanized 4 weeks after injection by cervical dislocation under anesthesia using isoflurane (4%) and thereafter tumors were resected, weighted, and then fixed in 10% formalin for immunohistochemistry.

Statistical analyses

Statistical analyses were performed using JMP pro 14.0.0 (SAS Institute). χ2 test or Mann-Whitney U test were applied for correlation between the infiltration of CD163-positive macrophages and clinicopathological factors. Breast cancer-specific and disease-free survival curves were generated by Kaplan-Meier method and statistical significance was evaluated by a log-rank test. In the in vitro and in vivo experiments, the statistical analyses were performed using paired or unpaired t-test. The data are presented as the mean ± SD. In this study, P<0.05 was considered as indicative of statistical significance.

Results

Expression of AR in macrophages within breast carcinoma tissues

We first examined whether macrophages in breast carcinoma tissues expressed AR or not. By double immunohistochemistry for AR and CD163, a marker of M2 macrophages, the macrophages with immunoreactivity for both CD163 (blue) in the cell membrane and AR (brown) in the nuclei were sporadically observed in the stroma (Fig. 1A).

We next immunolocalized CD163 as well as 5αRed1 in 116 breast carcinoma tissues in order to address the clinical significance of androgen action on macrophages in breast cancer. CD163 immunoreactivity was observed in the macrophages (Fig. 1B and C) and 34% (39 out of 116 cases) were categorized as high infiltration. 5αRed1 immunoreactivity was observed in the cytoplasm of breast carcinoma cells (Fig. 1D and E) and 56% (65 out of 116 cases) were considered positive for 5αRed1. Correlation between macrophage infiltration and clinicopathological parameters is presented in Table I. Macrophage infiltration was positively correlated with lymph node metastasis (P=0.0057), histological grade (P<0.0001) and Ki67 labeling index (LI) (P<0.0001), while it was negatively correlated with ER (P=0.0020) and PR (P=0.0014). No significant correlation was detected between macrophage infiltration and 5αRed1 immunoreactivity (P=0.74). When we compared the correlation between macrophage infiltration and clinicopathological parameters in the 5αRed1-negative group (n=51) and the 5αRed1-positive group (n=65) (Table II), macrophage infiltration was correlated with lymph node metastasis (P=0.011), PR (P=0.0061) and HER2 (P=0.015) only in the 5αRed1-positive group. On the other hand, it was negatively correlated with ER only in the 5αRed1-negative group (P=0.001), while it was correlated with histological grade and Ki67 LI in both the 5αRed1-positive and -negative group (histological grade; P=0.0013 in the 5αRed1-negative group and P=0.0069 in the 5αRed1-positive group, Ki67 LI; P=0.0017 in the 5αRed1-negative group and P=0.0020 in the 5αRed1-positive group). Furthermore, it is of interest that a significant correlation between macrophage infiltration and these parameters was detected only in the 5αRed1-positive group when the cases were limited to the ER-positive group.

Table I.

Association between CD163-positive macrophages and clinicopathological parameters in 116 breast carcinomas.

Table I.

Association between CD163-positive macrophages and clinicopathological parameters in 116 breast carcinomas.

CD163 positive macrophages

Low (n=77)High (n=39)P-value
Agea (years)55 (27–87)58 (37–76)0.3700
Menopause
  Premenopause30110.2500
  Postmenopause4728
Stage
  I49180.1000
  II1911
  III910
Pathological T factor
  pT156220.0770
  pT2-42117
Lymph node metastasis
  Negative59200.0057
  Positive1819
Histological grade
  1 (well)396 <0.0001
  2 (intermediate)3016
  3 (poor)817
ER
  Negative9140.0020
  Positive6825
PR
  Negative17200.0014
  Positive6019
HER2
  Negative69300.0680
  Positive89
5αRed1
  Negative33180.7400
  Positive4421
Ki67 LI a (%)8 (1–44)21 (1–60) <0.0001

a Data are presented as median (minimum-maximum). All other values are presented as the number of cases. P<0.05 is considered indicative of statistical significance and are presented in bold print. 5αRed1, 5α-reductase 1; ER, estrogen receptor; HER2, human epidermal growth factor receptor 2; LI, labeling index; PR, progesterone receptor.

Table II.

Association between CD163-positive macrophages and clinicopathological parameters according to 5αRed1 and ER status in 116 breast carcinomas.

Table II.

Association between CD163-positive macrophages and clinicopathological parameters according to 5αRed1 and ER status in 116 breast carcinomas.

All cases (n=116)ER-positive cases (n=93)


Parameters5αRed1-negative (n=51)5αRed1-positive (n=65)5αRed1-negative (n=41)5αRed1-positive (n=52)
Age0.420.730.530.61
Menopause0.140.810.400.69
Stage0.200.150.670.26
Pathological T factor0.140.290.280.36
Lymph node metastasis0.180.0110.630.043
(Positive) (Positive)
Histological grade0.00130.00690.140.0011
(Positive)(Positive) (Positive)
ER0.00100.23
(Negative)(Negative)
PR0.0820.00610.180.0059
(Negative) (Negative)
HER20.920.0150.230.0079
(Positive) (Positive)
Ki67 LI (%)0.00170.00200.0910.0009
(Positive)(Positive) (Positive)

[i] P-values of statistical analyses for the correlation between macrophage infiltration and clinicopathological parameters are provided. P<0.05 is considered significant and is represented in bold print. 5αRed1, 5α-reductase 1; ER, estrogen receptor; HER2, human epidermal growth factor receptor 2; LI, labeling index; PR, progesterone receptor.

We next examined the correlation between macrophage infiltration and clinical outcomes of breast cancer patients. As shown in Fig. 2A and B, macrophage infiltration was significantly correlated with an increased risk of recurrence (P=0.0049) and worse prognosis (P=0.016). In contrast, when we analyzed according to 5αRed1 status, macrophage infiltration was significantly correlated with increased risk of recurrence (P=0.01, Fig. 2C) and worse prognosis (P=0.045, Fig. 2E) only in the 5αRed1-positive group and no significant correlation was detected in the 5αRed1-negative group (Fig. 2D and F). This tendency was still observed when the cases were limited to the ER-positive group (Fig. 2G-I), although breast cancer-specific survival curve could not be generated because no patients had died in the ER-positive/5αRed1-negative group.

Androgen is necessary for the pro-proliferative effects of TAMs

We suggested that androgens may have important roles in macrophage-induced breast cancer progression by immunohistochemical analyses for CD163 and 5αRed1 as described above. We then tested this hypothesis in in vitro experiments using mouse breast cancer and macrophage cell lines, 4T1 and RAW264.7.

When RAW264.7 cells interacted with 4T1 cells in a coculture chamber, mRNA and protein expression of Arg-1, an M2 macrophage marker was significantly increased when compared to the control (monoculture group), indicating that RAW264.7 cells were successfully polarized into an M2 phenotype (Fig. 3A). We next investigated mRNA expression, in 4T1 and RAW264.7 cells, of mouse AR (Ar) as well as 5αRed1 (Srd5a1) and Akr1c6, the ortholog of human AKR1C3 and involved in androgen synthesis in the mouse (29). Real-time PCR analysis showed that Ar (Fig. 3B) and Akr1c6 (Fig. 3C) mRNA was almost negligible in both cell lines, while Srd5a1 mRNA was detected in both (Fig. 3D). In addition, the mRNA level of these genes was almost comparable in coculture condition (Fig. 3E and F). To investigate the effects of androgens on macrophages, we generated RAW264.7 sublines expressing AR (RAW-AR) and the corresponding negative control (RAW-CT) (Fig. 3G). When we treated RAW-CT and RAW-AR cells with synthetic androgen R1881, mRNA expression of M2 macrophage marker Arg-1 was not affected by R1881 (Fig. 3H). We confirmed that AR protein was not detected in 4T1 cells (Fig. 3G), and 4T1 cells were cocultured with RAW-CT or RAW-AR cells and cell proliferation was investigated by sphere-forming assay. As shown in Fig. 3I, sphere size was significantly increased by R1881 when the cells were cocultured with RAW-AR (P<0.01), while R1881 had no effect on monocultured 4T1 cells or those cocultured with RAW-CT.

Figure 3.

Effects of androgen action in macrophages on breast cancer cell proliferation. (A) RAW264.7 cells were cocultured with 4T1 cells for 3 days and Arginase-1 (Arg-1) mRNA expression (M2 macrophage marker) was evaluated by real-time PCR in RAW264.7 cells. Arg-1 mRNA was evaluated as the ratio to ribosomal protein L13A (Rpl13a) mRNA level and relative Arg-1 mRNA level was presented as the fold change compared to the control (monoculture) group. The statistical analysis was carried out using unpaired t-test and the data are presented as the mean ± SD (n=3). ***P<0.001 vs. the control group. Expression of ARG-1 protein in RAW264.7 cells was evaluated by immunoblotting. β-actin was used as a loading control. (B-D) Androgen receptor (Ar), (B) aldo-keto reductase family 1 member C6 (Akr1c6) (C) and 5α-reductase 1 (Srd5a1) (D) mRNA expression in 4T1 and RAW264.7 cells was evaluated by real-time PCR as the ratio to Rpl13a. The data are presented as the mean ± SD (n=3). (E and F) Ar, Akr1c6 and Srd5a1 mRNA expression was evaluated by real-time PCR in 4T1 (E) and RAW264.7 cells (F) in coculture for 3 days. These mRNAs were evaluated as the ratio to Rpl13a mRNA level. The statistical analysis was carried out using unpaired t-test and the data are presented as the mean ± SD (n=3). (G) Expression of AR in 4T1, RAW-CT and RAW-AR cells was assessed by immunoblotting. β-actin was used as a loading control. (H) RAW-CT and RAW-AR cells were treated with synthetic androgen R1881 (10 nM) for 48 h and Arg-1 mRNA expression was evaluated by real-time PCR in RAW264.7. Arg-1 mRNA was evaluated as the ratio to Rpl13a mRNA level and relative Arg-1 mRNA level was presented as the fold change compared to control (without R1881). The statistical analysis was carried out using unpaired t-test and the data are presented as the mean ± SD (n=3). (I) 4T1 cells were cocultured with either RAW-CT or RAW-AR cells, and then exposed to R1881 (10 nM) in the low attachment surface plate for 3 days. Sphere size was evaluated by measuring 100 spheres in each well using ImageJ and averages were calculated. The statistical analyses were carried out using unpaired t-test and the data are presented as the mean ± SD (n=3). **P<0.01 vs. the control group (without R1881). ND, not detected; NS, not significant.

Androgen acts on macrophages to enhance breast cancer progression in a tumor-bearing mouse model

In order to further address whether androgen acts on macrophages to promote breast cancer progression, we performed in vivo experiments using tumor-bearing mice. We generated 4T1 cells stably expressing AKR1C6 (4T1-AKR1C6) using pAKR1C6-FLAG vector or control vector (Fig. 4A). When we compared in vitro cell proliferation of these cells, it was comparable in 4T1-AKR1C6 and 4T1-CT cells (Fig. 4A). 4T1-CT or 4T1-AKR1C6 cells were subcutaneously injected together with RAW-AR cells on the left or right side of the back of mice, respectively (Fig. 4B; left image). The tumors on the right side (4T1-AKR1C6+RAW-AR) grew more rapidly compared with those on the left side (4T1-CT+RAW-AR) (Fig. 4C and D). In addition, immunohistochemistry demonstrated that Ki67 LI was higher in the tumor on the right side (70%) compared to those on the left side (30%) (Fig. 4E). We then performed the mirror experiment, where 4T1-AKR1C6 cells were subcutaneously injected together with RAW-CT and RAW-AR cells on the left or right side of the back of mice, respectively (Fig. 4B; right image). The tumors on the right side (4T1-AKR1C6+RAW-AR) grew more rapidly compared with those on the left side (4T1-AKR1C6+RAW-CT) (Fig. 4F and G). Ki67 LI was higher in the tumors on the right side (80%) compared to those on the left side (30%) (Fig. 4H).

Discussion

This is the first report demonstrating expression of the androgen receptor (AR) in macrophages in human solid tumors, including breast carcinomas. To date, the focus of study for the action of androgen in breast cancers has been mainly its mediation by AR in breast carcinoma cells, while AR in stromal cells has not been explored. On the other hand, AR signaling in cancer stromal cells has been implicated in prostate carcinomas and associated with higher proliferation and invasion of prostate carcinoma cells (30,31). Moreover, expression of AR in macrophages has been so far reported in several human diseases. For instance, macrophages with AR expression contribute to the greater prevalence and severity of atherosclerosis in men, by upregulating cholesteryl ester content (20). In addition, testosterone has been reported to induce TNFα, NO and IL-6 in a polycystic ovary syndrome rat model and contribute to the pathogenesis of polycystic ovary syndrome (32). Therefore, it is reasonably speculated that androgens affect breast cancer progression through AR, not only in breast cancer cells but also intratumoral macrophages.

Breast carcinoma cells express androgen-producing enzymes such as 5α-reductase type 1 (5αRed1) and locally produce dihydrotestosterone (DHT), a bioactive androgen (2). We therefore hypothesized that intratumoral androgens possibly affect macrophages which express AR in a paracrine-manner. Then, we correlated macrophage infiltration with characteristics of breast carcinomas in all pathological samples. Macrophage infiltration was positively correlated with lymph node metastasis, higher histological grade and Ki67 LI while negatively correlated with the estrogen receptor (ER) and progesterone receptor (PR) in all 116 cases, showing agreement with previous reports (13,19). When we further analyzed them according to 5αRed1 status, correlation between macrophage infiltration and clinicopathological parameters showed differences between 5αRed1-positive and 5αRed1-negative groups, and this tendency was more prominent when the cases were limited to ER-positive breast cancers. Macrophage infiltration was in particular correlated with a more aggressive phenotype of breast carcinomas with lymph node metastasis, higher histological grade, PR negativity, HER2 positivity and higher Ki67 LI in the 5αRed1-positive group but not in the 5αRed1-negative group. These findings indicate that the action of androgen in macrophages is important for the pro-tumorigenic roles of macrophages in breast cancers, especially in ER-positive breast cancers. This may be partly explained by the high diversity of cytokine receptor expression in breast carcinomas. For instance, it has been reported that the expression profile of cytokine receptor genes is quite different between the luminal type and basal-type breast cancer cell lines. Rearranged during transfection (RET), a receptor for glial cell line-derived neurotrophic factor (GDNF), is highly expressed in luminal-type breast cancer cell lines (33) and while production of GDNF is accelerated by androgens in testicular peritubular cells (34). Therefore, a future challenge will be to identify the soluble factors regulated by androgens in macrophages and their receptors in breast carcinoma tissues.

In the present study, macrophage infiltration was significantly correlated with increased risk of recurrence and worse prognosis, similarly as previous reports (35,36). Interestingly, this correlation was observed in the 5αRed1-positive group but not in the 5αRed1-negative group, suggesting that androgen action in intratumoral macrophages contributes to breast cancer progression and worse prognosis. Similar results were obtained even when the cases were limited to the ER-positive group, who had received adjuvant endocrine therapy. Although endocrine therapy targeting estrogen synthesis or ER has successfully improved clinical outcome of ER-positive breast cancer patients, de novo or acquired resistance is still frequently experienced in the clinical setting. We previously reported that intratumoral androgen production is increased following aromatase inhibitor (AI) treatment (37), and increased androgen signaling in breast cancer cells possibly causes resistance to AI (9). In addition, macrophages infiltrate more in metastatic breast carcinoma tissues than in primary breast carcinoma tissues (15). Considering our present findings and these previous reports, androgen action in macrophages as well as breast carcinoma cells seems to play pivotal roles for acquisition of endocrine resistance. Our recent findings may thus provide important clues to overcome endocrine resistance.

We next constructed in vitro and in vivo experiments using mouse breast cancer and macrophage cell lines, 4T1 and RAW264.7. Real-time PCR analysis showed that Ar and Akr1c6 mRNA was negligible in both 4T1 and RAW264.7 cells. However, cell lines could not necessarily mimic the microenvironment of human breast carcinoma tissues. We demonstrated the expression of AR in breast cancer-associated macrophages by double-immunohistochemistry, and androgen-producing enzymes such as 17βHSD5 and 5αRed1 are frequently expressed in human breast carcinomas and contribute to intratumoral androgen synthesis (2,3). We therefore tried to mimic this situation using mouse cell lines by establishing AR- and AKR1C6-overexpressing sublines. In the sphere-forming assay using 4T1 and RAW-AR cell lines, which stably express AR, sphere size was significantly increased by androgens when 4T1 cells were cocultured with RAW-AR cells. Because 4T1 cells did not express AR, increased sphere size was due to androgen action in RAW-AR cells. Furthermore, in vivo experiments using a tumor-bearing mouse model showed that tumor volume as well as Ki67 LI were increased significantly when androgens were stability produced in breast cancer cells and AR was expressed in macrophages. Intratumoral macrophages produce numerous kinds of soluble factors and they are strongly linked to pro-tumorigenic roles for macrophages (38), although little is known about androgen-regulated soluble factors in intratumoral macrophages. However, some cytokines such as TNFα and IL-6 are induced by androgens in macrophages in both pathological and physiological conditions (17,22,39) and finasteride, a potent 5α-reductase inhibitor suppresses TNFα secretion from prostatic macrophages (40). In breast cancer, TNFα enhances invasiveness of breast cancer cells through matrix metalloproteinase (MMP) induction in macrophages (41). Moreover, IL-6 expression in macrophages isolated from hepatocellular carcinoma (HCC) is markedly upregulated by interaction with HepG2 HCC cell line and IL-6 promotes stemness of HCC via the signal transducer and activator of transcription (STAT)3 pathway (42). Recently, AR signaling in macrophages has been shown to promote migration and invasion of prostate cancer cell lines by increased triggering receptor expressed on myeloid cells-1 (TREM-1) signaling, regulating the expression of its downstream cytokines such as CCL2, CCL7, CXCL8 and CCL13 (31). On the other hand, there was no significant correlation between macrophage infiltration and 5αRed1 in human breast carcinoma tissues and R1881 did not alter Arg-1 mRNA expression in RAW-AR cells. Taken together, these results show that androgen action may not necessarily correlate with macrophage polarization but androgen activates macrophages by inducing soluble factors which have pro-tumorigenic roles in human malignancies. Further examination is needed to identify androgen-induced soluble factors in intratumoral macrophages, in order to explore the relationships between androgens and the breast cancer microenvironment.

One of the limitations of the present study is the lack of quantitative data on AR-positive macrophages due to the relatively weak immunoreactivity of AR in macrophages, making it impossible for us to directly evaluate the significance of androgen action upon them. Alternative quantitative methods such as flow cytometry will be required in the future. Moreover, we did not compare the tumor formation of 4T1-CT and 4T1-AKR1C6 cells without RAW264.7 cells, although we concluded that androgen action may not correlate with macrophage polarization as described above. In addition, the effect of finasteride, 5α-reductase inhibitor on intratumoral macrophages was not investigated in the present study. Further in vivo examination is needed to reveal in more detail the action of androgen in macrophages. Finally, in vitro and in vivo experiments were conducted using 4T1 mouse breast cancer cells, which do not express ER and we could not experimentally verify the role of androgen action of macrophages in ER-positive breast cancers. The reason why we used 4T1 is that the mouse models of ER-positive breast cancer cells are lacking (43) and 4T1 cells are widely used for mouse breast cancer models in many previous studies (4446). In addition, we could not differentiate human monocytic THP-1 cells into the M2 phenotype by coculture with human breast cancer cell lines including MCF-7 cells, which express high levels of ER. Therefore, the present study is the starting point of further investigation into novel androgen action focusing on intratumoral macrophages in breast carcinoma tissues, which may help understanding the complex breast cancer microenvironment.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

This work was partly supported by JSPS KAKENHI Grant Number 19K09065 and 19K07410.

Availability of data and materials

All data and material presented in this article are available from the corresponding author upon reasonable request.

Author's contributions

MY performed cell and animal experiments and prepared the manuscript. KT contributed to the study conception and design and performed statistical analysis. MS performed the immunohistochemistry. AS assisted with the animal experiments. YM and YO gave skillful suggestions in regards to the immunohistochemistry. MM and HS collected the clinical samples and clinicopathological information. TS performed the pathological analysis. All authors read and approved the final manuscript and agreed to be accountable for all aspects of the work in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The study using clinical samples was approved by the Ethics Committee at Tohoku University Graduate School of Medicine (approval no. 2016-1-697, 2019-1-219). Experiments and analyses were performed in accordance to the Helsinki declaration. This is a retrospective study and therefore informed consent had not been obtained. All animal experiments were performed in accordance with ARRIVE guidelines with the approval of the Institutional Animal Care and Use Committee at Tohoku University (2019MdA-172).

Patient consent for publication

Not applicable.

Competing interests

The authors have no competing interests.

References

1 

Luqmani YA and Alam-Eldin N: Overcoming resistance to endocrine therapy in breast cancer: New approaches to a nagging problem. Med Princ Pract. 25 (Suppl 2):S28–S40. 2016. View Article : Google Scholar

2 

Suzuki T, Darnel AD, Akahira JI, Ariga N, Ogawa S, Kaneko C, Takeyama J, Moriya T and Sasano H: 5alpha-reductases in human breast carcinoma: Possible modulator of in situ androgenic actions. J Clin Endocrinol Metab. 86:2250–2257. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Suzuki T, Miki Y, Moriya T, Akahira J, Ishida T, Hirakawa H, Yamaguchi Y, Hayashi S and Sasano H: 5Alpha-reductase type 1 and aromatase in breast carcinoma as regulators of in situ androgen production. Int J Cancer. 120:285–291. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Hall RE, Aspinall JO, Horsfall DJ, Birrell SN, Bentel JM, Sutherland RL and Tilley WD: Expression of the androgen receptor and an androgen-responsive protein, apolipoprotein D, in human breast cancer. Br J Cancer. 74:1175–1180. 1996. View Article : Google Scholar : PubMed/NCBI

5 

Kuenen-Boumeester V, Van der Kwast TH, Claassen CC, Look MP, Liem GS, Klijn JG and Henzen-Logmans SC: The clinical significance of androgen receptors in breast cancer and their relation to histological and cell biological parameters. Eur J Cancer. 32A:1560–1565. 1996. View Article : Google Scholar : PubMed/NCBI

6 

Vera-Badillo FE, Templeton AJ, de Gouveia P, Diaz-Padilla I, Bedard PL, Al-Mubarak M, Seruga B, Tannock IF, Ocana A and Amir E: Androgen receptor expression and outcomes in early breast cancer: A systematic review and meta-analysis. J Natl Cancer Inst. 106:djt3192014. View Article : Google Scholar : PubMed/NCBI

7 

Farmer P, Bonnefoi H, Becette V, Tubiana-Hulin M, Fumoleau P, Larsimont D, Macgrogan G, Bergh J, Cameron D, Goldstein D, et al: Identification of molecular apocrine breast tumours by microarray analysis. Oncogene. 24:4660–4671. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y and Pietenpol JA: Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 121:2750–2767. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Fujii R, Hanamura T, Suzuki T, Gohno T, Shibahara Y, Niwa T, Yamaguchi Y, Ohnuki K, Kakugawa Y, Hirakawa H, et al: Increased androgen receptor activity and cell proliferation in aromatase inhibitor-resistant breast carcinoma. J Steroid Biochem Mol Biol. 144:513–522. 2014. View Article : Google Scholar : PubMed/NCBI

10 

De Palma M, Biziato D and Petrova TV: Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer. 17:457–474. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Vasiliadou I and Holen I: The role of macrophages in bone metastasis. J Bone Oncol. 2:158–166. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Gan L, Qiu Z, Huang J, Li Y, Huang H, Xiang T, Wan J, Hui T, Lin Y, Li H and Ren G: Cyclooxygenase-2 in tumor-associated macrophages promotes metastatic potential of breast cancer cells through Akt pathway. Int J Biol Sci. 12:1533–1543. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Yang J, Li X, Liu X and Liu Y: The role of tumor-associated macrophages in breast carcinoma invasion and metastasis. Int J Clin Exp Pathol. 8:6656–6664. 2015.PubMed/NCBI

14 

Li J, He K, Liu P and Xu LX: Iron participated in breast cancer chemoresistance by reinforcing IL-6 paracrine loop. Biochem Biophys Res Commun. 475:154–160. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Liu H, Wang J, Zhang M, Xuan Q, Wang Z, Lian X and Zhang Q: Jagged1 promotes aromatase inhibitor resistance by modulating tumor-associated macrophage differentiation in breast cancer patients. Breast Cancer Res Treat. 166:95–107. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Gelsomino L, Giordano C, Camera GL, Sisci D, Marsico S, Campana A, Tarallo R, Rinaldi A, Fuqua S, Leggio A, et al: Leptin signaling contributes to aromatase inhibitor resistant breast cancer cell growth and activation of macrophages. Biomolecules. 10:5432020. View Article : Google Scholar : PubMed/NCBI

17 

Chanmee T, Ontong P, Konno K and Itano N: Tumor-associated macrophages as major players in the tumor microenvironment. Cancers (Basel). 6:1670–1690. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, et al: Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 41:14–20. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Sousa S, Brion R, Lintunen M, Kronqvist P, Sandholm J, Mönkkönen J, Kellokumpu-Lehtinen PL, Lauttia S, Tynninen O, Joensuu H, et al: Human breast cancer cells educate macrophages toward the M2 activation status. Breast Cancer Res. 17:1012015. View Article : Google Scholar : PubMed/NCBI

20 

McCrohon JA, Death AK, Nakhla S, Jessup W, Handelsman DJ, Stanley KK and Celermajer DS: Androgen receptor expression is greater in macrophages from male than from female donors. A sex difference with implications for atherogenesis. Circulation. 101:224–226. 2000. View Article : Google Scholar : PubMed/NCBI

21 

Huang CK, Pang H, Wang L, Niu Y, Luo J, Chang E, Sparks JD, Lee SO and Chang C: New therapy via targeting androgen receptor in monocytes/macrophages to battle atherosclerosis. Hypertension. 63:1345–1353. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Lai JJ, Lai KP, Chuang KH, Chang P, Yu IC, Lin WJ and Chang C: Monocyte/macrophage androgen receptor suppresses cutaneous wound healing in mice by enhancing local TNF-alpha expression. J Clin Invest. 119:3739–3751. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Yamaguchi M, Takagi K, Sato A, Miki Y, Miyashita M, Sasano H and Suzuki T: Rac1 activation in human breast carcinoma as a prognostic factor associated with therapeutic resistance. Breast Cancer. 27:919–928. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Yamaguchi M, Takagi K, Narita K, Miki Y, Onodera Y, Miyashita M, Sasano H and Suzuki T: Stromal CCL5 promotes breast cancer progression by interacting with ccr3 in tumor cells. Int J Mol Sci. 22:19182021. View Article : Google Scholar : PubMed/NCBI

25 

Hamlin IM: Possible host resistance in carcinoma of the breast: A histological study. Br J Cancer. 22:383–401. 1968. View Article : Google Scholar : PubMed/NCBI

26 

Hammond ME, Hayes DF, Wolff AC, Mangu PB and Temin S: American society of clinical oncology/college of american pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Oncol Pract. 6:195–197. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Takagi K, Miki Y, Onodera Y, Ishida T, Watanabe M, Sasano H and Suzuki T: ARHGAP15 in human breast carcinoma: A potent tumor suppressor regulated by androgens. Int J Mol Sci. 19:E8042018. View Article : Google Scholar

28 

Sato A, Takagi K, Miki Y, Yoshimura A, Hara M, Ishida T, Sasano H and Suzuki T: Cytochrome c1 as a favorable prognostic marker in estrogen receptor-positive breast carcinoma. Histol Histopathol. 34:1365–1375. 2019.PubMed/NCBI

29 

Vergnes L, Phan J, Stolz A and Reue K: A cluster of eight hydroxysteroid dehydrogenase genes belonging to the aldo-keto reductase supergene family on mouse chromosome 13. J Lipid Res. 44:503–511. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Niu Y, Altuwaijri S, Yeh S, Lai KP, Yu S, Chuang KH, Huang SP, Lardy H and Chang C: Targeting the stromal androgen receptor in primary prostate tumors at earlier stages. Proc Natl Acad Sci USA. 105:12188–12193. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Cioni B, Zaalberg A, van Beijnum JR, Melis MHM, van Burgsteden J, Muraro MJ, Hooijberg E, Peters D, Hofland I, Lubeck Y, et al: Androgen receptor signalling in macrophages promotes TREM-1-mediated prostate cancer cell line migration and invasion. Nat Commun. 11:44982020. View Article : Google Scholar : PubMed/NCBI

32 

Figueroa F, Davicino R, Micalizzi B, Oliveros L and Forneris M: Macrophage secretions modulate the steroidogenesis of polycystic ovary in rats: Effect of testosterone on macrophage pro-inflammatory cytokines. Life Sci. 90:733–739. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Levano KS, Jung EH and Kenny PA: Breast cancer subtypes express distinct receptor repertoires for tumor-associated macrophage derived cytokines. Biochem Biophys Res Commun. 411:107–110. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Chen LY, Brown PR, Willis WB and Eddy EM: Peritubular myoid cells participate in male mouse spermatogonial stem cell maintenance. Endocrinology. 155:4964–4974. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Gwak JM, Jang MH, Kim DI, Seo AN and Park SY: Prognostic value of tumor-associated macrophages according to histologic locations and hormone receptor status in breast cancer. PLoS One. 10:e01257282015. View Article : Google Scholar : PubMed/NCBI

36 

Klingen TA, Chen Y, Aas H, Wik E and Akslen LA: Tumor-associated macrophages are strongly related to vascular invasion, non-luminal subtypes, and interval breast cancer. Hum Pathol. 69:72–80. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Takagi K, Miki Y, Nagasaki S, Hirakawa H, Onodera Y, Akahira J, Ishida T, Watanabe M, Kimijima I, Hayashi S, et al: Increased intratumoral androgens in human breast carcinoma following aromatase inhibitor exemestane treatment. Endocr Relat Cancer. 17:415–430. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Yang L and Zhang Y; Tumor-associated macrophages, : From basic research to clinical application. J Hematol Oncol. 10:582017. View Article : Google Scholar : PubMed/NCBI

39 

Schneider CP, Schwacha MG, Samy TS, Bland KI and Chaudry IH: Androgen-mediated modulation of macrophage function after trauma-hemorrhage: Central role of 5alpha-dihydrotestosterone. J Appl Physiol (1985). 95:104–112. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Zhao R, Wang X, Jiang C, Shi F, Zhu Y, Yang B, Zhuo J, Jing Y, Luo G, Xia S and Han B: Finasteride accelerates prostate wound healing after thulium laser resection through DHT and AR signalling. Cell Prolif. 51:e124152018. View Article : Google Scholar : PubMed/NCBI

41 

Hagemann T, Robinson SC, Schulz M, Trümper L, Balkwill FR and Binder C: Enhanced invasiveness of breast cancer cell lines upon co-cultivation with macrophages is due to TNF-alpha dependent up-regulation of matrix metalloproteases. Carcinogenesis. 25:1543–1549. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Wan S, Zhao E, Kryczek I, Vatan L, Sadovskaya A, Ludema G, Simeone DM, Zou W and Welling TH: Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterology. 147:1393–1404. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Mohibi S, Mirza S, Band H and Band V: Mouse models of estrogen receptor-positive breast cancer. J Carcinog. 10:352011. View Article : Google Scholar : PubMed/NCBI

44 

Pulaski BA and Ostrand-Rosenberg S: Mouse 4T1 breast tumor model. Curr Protoc Immunol. 20:Chapter 20: Unit 20.2. 2001.PubMed/NCBI

45 

Cho HJ, Jung JI, Lim DY, Kwon GT, Her S and Park JH and Park JH: Bone marrow-derived, alternatively activated macrophages enhance solid tumor growth and lung metastasis of mammary carcinoma cells in a Balb/C mouse orthotopic model. Breast Cancer Res. 14:R812012. View Article : Google Scholar : PubMed/NCBI

46 

Jang JY, Lee JK, Jeon YK and Kim CW: Exosome derived from epigallocatechin gallate treated breast cancer cells suppresses tumor growth by inhibiting tumor-associated macrophage infiltration and M2 polarization. BMC Cancer. 13:4212013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2021
Volume 46 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yamaguchi M, Takagi K, Sato M, Sato A, Miki Y, Onodera Y, Miyashita M, Sasano H and Suzuki T: Androgens enhance the ability of intratumoral macrophages to promote breast cancer progression. Oncol Rep 46: 188, 2021
APA
Yamaguchi, M., Takagi, K., Sato, M., Sato, A., Miki, Y., Onodera, Y. ... Suzuki, T. (2021). Androgens enhance the ability of intratumoral macrophages to promote breast cancer progression. Oncology Reports, 46, 188. https://doi.org/10.3892/or.2021.8139
MLA
Yamaguchi, M., Takagi, K., Sato, M., Sato, A., Miki, Y., Onodera, Y., Miyashita, M., Sasano, H., Suzuki, T."Androgens enhance the ability of intratumoral macrophages to promote breast cancer progression". Oncology Reports 46.3 (2021): 188.
Chicago
Yamaguchi, M., Takagi, K., Sato, M., Sato, A., Miki, Y., Onodera, Y., Miyashita, M., Sasano, H., Suzuki, T."Androgens enhance the ability of intratumoral macrophages to promote breast cancer progression". Oncology Reports 46, no. 3 (2021): 188. https://doi.org/10.3892/or.2021.8139