Main

Breast cancer is the most common type of cancer among women, and its incidence is increasing worldwide. According to statistics, about 62 570 cases of breast cancer in situ are expected to be newly diagnosed in the United States in 2014.1 Moreover, the incidence of breast cancer – like most cancers – is on the rise in develo** countries such as Brazil and China, as populations increasingly adopt western lifestyles.2 Therefore, breast cancer ranks the second most common cause of cancer death among women worldwide, with about 1.4 million new cases annually.1, 3, 4 Despite significant improvement in survival rates of patients with breast cancer, the disease remains a huge threat to women’s health, and particularly patients with ‘triple-negative’ breast cancer (TNBC), referring to cancers that express neither the estrogen receptor or progesterone receptor nor display amplification of human epidermal growth factor receptor 2, are insensitive to hormonal therapy or HER2-targeted drugs.5, 6, 7 Advanced TNBC confer an aggressive clinical course with a poor prognosis compared with non-TNBC.8 Furthermore, breast cancer is highly malignant with considerable metastatic potential, and metastatic breast cancer is a principle cause of female mortality.9 Unfortunately, there is currently no effective therapy to control the recurrence and metastasis of breast cancer, and therefore the development of new therapies is essential.

Signal transducer and activator of transcription 3 (Stat3) has important roles in cancer and other disease, and presents tremendous therapeutic potential.10 Stat3 is a point of convergence for multiple oncogenic signaling pathways. Meanwhile, Stat3 as a proto-oncogene could mediate cellular and biological processes.10 In a variety of human cancers, constitutively active Stat3 signaling promotes tumorigenesis and tumor progression by dysregulating the expression of key genes that control cell apoptosis (such as Bcl-2, Bcl-xl and Mcl-1), proliferation (cyclin d1, c-Myc), angiogenesis (vascular endothelial growth factor), migration, invasion or metastasis (matrix metalloproteinase 1 (MMP1), MMP7 and MMP-9).11, 12, 13, Full size image

We then performed a Matrigel invasion assay. Figure 3c shows that both 4T1 and MDA-MB-231 cells exhibit significantly decreased invasion in the presence of nifuroxazide than vehicle. Moreover, constitutive Stat3 also has an important role in controlling cell migration and invasion by regulating the expression of genes involved MMP-2, -9 and others.30 Therefore, we also investigated whether phosphorylated-Stat3 (Tyr705), MMP-2, -9, which are considered to be related with cell migration and invasion, are involved in nifuroxazide-mediated migration and invasion. As Figure 3d indicates, nifuroxazide treatment decreased the expression of phosphorylated-Stat3 (Tyr705), MMP-2 and MMP-9 without affecting the total Stat3 expression level in 4T1 cells. Altogether, our results showed that nifuroxazide possessed a strong ability on breast cancer cell migration and invasion.

Retardation of mammary tumor growth in vivo

The remarkable inhibitory effects of nifuroxazide on 4T1 cells proliferation in vitro implied that it might inhibit tumor growth in vivo. To verify this hypothesis, we examine the antitumor activity of nifuroxazide in vivo. 4T1 tumor-bearing mice were dosed daily at the dose of 10 and 50 mg/kg for 24 days. Nonsignificant changes in body weight were observed in nifuroxazide-treated animals (Supplementary Figure S3a). Importantly, there was a significant reduction in tumor outgrowth and in tumor weight in the nifuroxazide-treated groups (Figures 4a and b; Supplementary Figure S3b), showing that nifuroxazide has strong antitumor activity.

Figure 4
figure 4

Antitumor effects of nifuroxazide in vivo. (a) Tumor size were measured and calculated every 3 days and presented as mean±S.D. (n=6; *P<0.05). (b) Represented weight of tumor from mice of different groups, respectively. Data were mean ±S.D. (n=6; *P<0.05). (c) Tumor cell proliferation was evaluated on paraffin-embedded 4T1 tumor sections by Ki-67 immunohistochemical staining. The treatment with nifuroxazide resulted in markedly reduced proliferation versus vehicle group. (d) Apoptosis was measured on paraffin-embedded 4T1 tumor sections by CC-3 immunohistochemical staining. The treatment with nifuroxazide significantly increased apoptosis in a dose-dependent manner compared with vehicle group. (e) Immunohistochemistry was performed to measure the expression of MMP-9 in tumor tissues isolated from vehicle and nifuroxazide-treated mice. The treatment with nifuroxazide markedly reduced MMP-9-positive cells versus vehicle group (20 × )

To determine the mechanisms underlying nifuroxazide activity, 4T1-induced tumors collected on day 31 were examined for proliferation and apoptosis. Nifuroxazide treatment caused a significant reduction in proliferating cells stained positive for nuclear Ki-67 (Figure 4c). Moreover, as shown in Figure 4d, CC-3-positive cells were showing an increase in the nifuroxazide-treated sections versus sections of the untreated groups. Overall, these data suggest that nifuroxazide inhibits cell proliferation and induces apoptosis in breast tumor tissues through inhibition of Ki-67 and activation of caspase-3. In addition, there is increasing evidence to indicate that MMPs have important roles in tumor invasion and metastasis.31 We therefore measured the effects of nifuroxazide on the expression of MMP-9 by immunohistochemistry (IHC). As shown in Figure 4e, treatment of mice with nifuroxazide inhibited the expression of MMP-9 in 4T1 tumor tissues.

Nifuroxazide treatment decreases lung metastasis

Previous studies have showed that 4T1 murine breast tumor have a high metastatic potential and spontaneously metastasize to lung as early as 2 weeks after inoculation.32, 33, 34 To analyze the effects of nifuroxazide on metastasis, lungs of 4T1 tumor-bearing mice killed on day 31 were removed and metastatic nodules were quantified. In vehicle-treated groups, multiple large nodules were evident, whereas the extent of lung metastasis was markedly reduced in nifuroxazide-treated mice (50 mg/kg groups; Figures 5a and b). Moreover, there was a remarkable decrease in lung weight after nifuroxazide treatment compared with the untreated control (Figure 5c). Importantly, histological analyses demonstrated that the number of micrometastatic nodules per field in the nifuroxazide-treated group at 50 mg/kg was also significant fewer than the other groups (Supplementary Figure S4). Overall, these results further indicated that nifuroxazide could inhibit lung metastasis in breast cancer.

Figure 5
figure 5

Effects of nifuroxazide on metastasis. (a) Lung metastatic nodules were visualized to show the inhibitory effects of nifuroxazide on 4T1 tumor 24 days after treatment. (b) The mean lung metastasis nodules of each group, the treatment with nifuroxazide at 10 mg/kg and 50 mg/kg resulted in significant inhibition of lung metastasis versus vehicle control. Bars showed mean±S.D. (n=6; **P<0.01; ***P<0.001). (c) Weight of lungs in each group. Bars showed mean±S.D. (n=6; *P<0.05; **P<0.01)

Nifuroxazide modulates the lung metastatic environment

MDSCs, as mainly characterized by CD11b+ and Gr1+ double-positive myeloid cells in mice have been observed to accumulate in highly metastatic breast carcinoma 4T1. Moreover, it has been shown that accumulation of MDSCs into the lung has a key role in the development of metastasis, meanwhile, MDSCs are closely related to a lung metastatic in patients with breast cancer.35, 36, 37 Therefore, we further investigated lung myeloid cells infiltration in 4T1 tumor-bearing mice by FCM after 24 days of treatment. As shown in Figures 6a and b, the data showed that the percentage of MDSCs decreased in the 10 mg/kg-treated group compared with the control group. Moreover, we found that ~22% reduction of MDSCs in the lung after 50 mg/kg nifuroxazide treatment (Figure 6c). The statistical analysis demonstrated that nifuroxazide treatment reduced the number of MDSCs in lung in a dose-dependent manner (Figure 6d). These results suggested that nifuroxazide potently reduced the infiltration of MDSCs into the lung, which could inhibit tumor cell distant colonization.

Figure 6
figure 6

Nifuroxazide reduced lung Gr1+CD11b+MDSCs infiltration. Gr1+CD11b+cells were gated and analyzed by FCM for the expression of MDSCs. MDSCs isolated from the lungs of 4T1 tumor-bearing mice were treated with vehicle (a); or treated with nifuroxazide at 10 mg/kg (b); or treated with nifuroxazide at 50 mg/kg (c). (d) Statistic results of each group. Treatment of nifuroxazide significantly reduces the number of MDSCs compared with vehicle group. Values represented mean±S.D. (n=6; *P<0.05; **P<0.01)

Safety profile of nifuroxazide

As mentioned above, during the treatment of 4T1 tumor-bearing mice, we did not observed adverse effects, such as toxic death, skin ulceration and body weight loss, in the nifuroxazide-treated groups. To further investigate the safety profile of nifuroxazide, we determined here whether the nifuroxazide could cause the blood system’s abnormality, we performed blood routine analysis and blood chemistry analysis assays. Hematological and serum biochemistry analysis of the mice did not show any pathological changes (Figure 7a). Furthermore, no pathologic changes after nifuroxazide treatment were observed in the heart, liver, spleen and kidney by microscopic examination compared with the vehicle-treatment group (Figure 7b).

Figure 7
figure 7

Evaluation of side effects of nifuroxazide in mice. (a) Hematological and serum biochemistry analysis of blood were done. Units of the parameters are as follows. WBC, PLT, 109/l; RBC, 1012/l; TP, g/l; ALT, AST, U/l; CREA, UA, uM; TG, CHOL, mM. (b) Nifuroxazide did not cause obvious pathologic abnormalities in normal tissues. H&E staining of paraffin-embedded sections of the heart, liver, spleen and kidney (20 × )