Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2020

Open Access 01.12.2020 | Research

Evodiamine suppresses non-small cell lung cancer by elevating CD8+ T cells and downregulating the MUC1-C/PD-L1 axis

verfasst von: Ze-Bo Jiang, Ju-Min Huang, Ya-Jia Xie, Yi- Zhong Zhang, Chan Chang, Huan-Ling Lai, Wenjun Wang, Xiao-Jun Yao, Xing-Xing Fan, Qi-Biao Wu, Chun Xie, Mei-Fang Wang, Elaine Lai-Han Leung

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2020

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

Accumulating evidence showed that regulating tumor microenvironment plays a vital role in improving antitumor efficiency. Programmed Death Ligand 1 (PD-L1) is expressed in many cancer cell types, while its binding partner Programmed Death 1 (PD1) is expressed in activated T cells and antigen-presenting cells. Whereas, its dysregulation in the microenvironment is poorly understood. In the present study, we confirmed that evodiamine downregulates MUC1-C, resulting in modulating PD-L1 expression in non-small cell lung cancer (NSCLC).

Methods

Cell viability was measured by MTT assays. Apoptosis, cell cycle and surface PD-L1 expression on NSCLC cells were analyzed by flow cytometry. The expression of MUC1-C and PD-L1 mRNA was measured by real time RT-PCR methods. Protein expression was examined in evodiamine-treated NSCLC cells using immunoblotting or immunofluorescence assays. The effects of evodiamine treatment on NSCLC sensitivity towards T cells were investigated using human peripheral blood mononuclear cells and Jurkat, apoptosis and IL-2 secretion assays. Female H1975 xenograft nude mice were used to assess the effect of evodiamine on tumorigenesis in vivo. Lewis lung carcinoma model was used to investigate the therapeutic effects of combination evodiamine and anti-PD-1 treatment.

Results

We showed that evodiamine significantly inhibited growth, induced apoptosis and cell cycle arrest at G2 phase of NSCLC cells. Evodiamine suppressed IFN-γ-induced PD-L1 expression in H1975 and H1650. MUC1-C mRNA and protein expression were decreased by evodiamine in NSCLC cells as well. Evodiamine could downregulate the PD-L1 expression and diminish the apoptosis of T cells. It inhibited MUC1-C expression and potentiated CD8+ T cell effector function. Meanwhile, evodiamine showed good anti-tumor activity in H1975 tumor xenograft, which reduced tumor size. Evodiamine exhibited anti-tumor activity by elevation of CD8+ T cells in vivo in Lewis lung carcinoma model. Combination evodiamine and anti-PD-1 mAb treatment enhanced tumor growth control and survival of mice.

Conclusions

Evodiamine can suppress NSCLC by elevating of CD8+ T cells and downregulating of the MUC1-C/PD-L1 axis. Our findings uncover a novel mechanism of action of evodiamine and indicate that evodiamine represents a potential targeted agent suitable to be combined with immunotherapeutic approaches to treat NSCLC cancer patients. MUC1-C overexpression is common in female, non-smoker, patients with advanced-stage adenocarcinoma.
Hinweise

Supplementary Information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13046-020-01741-5.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
NSCLC
Non-small cell lung cancer
TCM
Traditional Chinese medicine
MUC1
MUCIN 1, cell surface associated
PD-L1
Programmed Death Ligand 1
PBMC
Human peripheral blood mononuclear cells
EGFE
Epidermal growth factor receptor
CTLA4
Cytotoxic T-lymphocyte associated protein 4
PBS
Public Broadcasting Service
PD-1
Programmed cell death protein 1

Background

Lung cancer in the world is the most commonly diagnosed carcinoma and becomes a paramount cause of cancer death, which is frequently associated with poor clinical outcomes [13].. Despite progress and development of new treatment and drugs, only 17.7% of all patients with lung cancer are alive ≥5 years after diagnosis [1, 3]. Immune cells are thought to be inactivated in tumor microenvironment, via the engagement of inhibitory receptors such as the famous Cytotoxic T-Lymphocyte Associated Protein 4(CTLA4) and Programmed cell death protein 1 (PD-1) [46]. Blocking immune checkpoints such as PD-L1 and PD-1 has improved the treatment of non -small cell lung cancer (NSCLC) [7]. Increasing clinical evidence shows that interaction between PD-1 and PD-L1 inhibits activation, expansion and effector functions of CD8+ T cells, and helps cancer cells evade immune destruction in the tumor microenvironment that is unfavorable to anti-cancer activities [8]. In addition, it is well known that the relation between microenvironment and immunology plays a vital role in the clinical treatment of NSCLC patients [911]. Activated CD8+ T cells have been demonstrated to have anti-cancer immunity in many different types of cancer [12, 13]. The presence of activated CD8+ T cells within the tumor and in the peritumoral stroma has been shown to have significant positive prognostic importance [14]. However, PD-1/PD-L1 inhibitor in clinical blockade has been limited by the response rate of about ∼20–30% in patients with NSCLC [15, 16]. The cancer microenvironment makes a vital role in the utility of immunotherapy [1719]. Increasing clinical evidence has also shown that tumor-infiltrating CD8+ lymphocytes (TILs) in the tumor environment is associated with survival of patients with cancer, further supporting the close relationship between immune escape and tumor microenvironment [2022].
Increasing evidence has indicated that MUC1-C plays a critical role in anti-cancer properties; for example, the transcriptional regulation of genes associated with tumor invasion [23], proliferation, metastasis [24], angiogenesis [24], apoptosis, inflammation [25], and drug resistance [26, 27] have been linked to poor outcomes in lung cancer [20]. Cancer cells usually have a high expression of MUC1-C and abnormal glycosylation of MUC1-C protein which is also overexpressed in human lung cancers and associated with a poor outcome [28]. Silencing the MUC1-C can improve the anticancer effect on lung and breast cancer [20, 28]. Accumulating evidence shows that MUC1-C regulates many genes, such as PD-L1, that promote the evasion of NSCLC cells and inhibit the effect of immune cells [29, 30]. Therefore, MUC1-C is a target for the downregulation of PD-L1 in NSCLC cells [31]. However, the effect of evodiamine on MUC1-C remains underexplored.
Evodiamine is a novel alkaloid, which was isolated from the fruit of Tetradium [32], and it has been considered an effective Chinese medicine for the treatment of gastropathy, hypertension, and eczema [33]. Several studies reported that evodiamine has various biological effects, including anti-nociceptive, anti-bacterial, anti-obesity and anti-cancer activities [3436]. However, to date, the effect of evodiamine on the PD-1/PD-L1 axis remains underexplored.
In this study, the effects of evodiamine on cell viability, cell cycle, and apoptosis in the human NSCLC cell lines were investigated and the underlying mechanisms are further explored. More importantly, the anticancer and immunomodulatory activities of evodiamine in the human NSCLC cells in vitro and in vivo models are carefully examined. Our results confirm the efficacy of combining evodiamine and anti-PD-1 mAb treatment against NSCLC cells. Our research also explored the involvement of MUC1-C/PD-L1 signaling of evodiamine in anti-lung cancer. Evodiamine can improve immunity in vivo by inhibiting PD-L1 expression in cancer. Therefore, our findings disclose the inhibition of evodiamine anti-NSCLC, which might have potential clinical implications.

Materials and method reagents

Materials

Evodiamine was supplied by Selleck Chemicals (Houston, TX, USA) and was dissolved in dimethyl sulfoxide (DMSO) and stored at − 20 °C. Primary antibodies against GAPDH (#5174), PD-L1(#13684), MUC1-C(#16564), C-MYC(#18583) were provided by Cell Signaling Technology (Danvers, MA, USA). Fluorescein secondary antibodies were provided by LI-COR Biosciences (Lincoln, NE, USA). Dead Cell Apoptosis Kit with Annexin V-FITC/PI were provided by BD Biosciences (San Jose, CA, USA).

Cell lines and cell culture

The proliferation of lung cancer cells was assessed using the MTT assay as described previously [37]. After treatment of 72 h, 20 μl MTT (5 mg/ml) solution was added to each well and incubated for 4 h. Then, 100 μl of the DMSO was added to each well. Finally, the colorimetric intensity of the plates was measured at the wavelength of 570 nm by the Tecan microplate reader (Morrisville, NC, USA).

Flow cytometric analysis

Apoptosis was analysis as described previously [38]. After treatment of 24 h, the percentage of apoptotic cells on evodiamine-treated NSCLC was analyzed using a BD FACSAria III flow cytometer. The percentages of the sub-G1, S, G1 and G2 phases cells were quantitatively determined using flow cytometer. For cell surface PD-L1 on lung cancer cell lines, cells after treatment were suspended in FACS stain buffer and incubated with APC anti-human CD274 at 4 °C for 30 min and cells were resuspended, and measured analyzed using flow cytometer.

Western blot analysis

The detailed procedure was reported previously [37]. The following antibodies were used in this experiment: GAPDH, MUC1-C (D5K9I) XP, PD-L1 and C-MYC. The protein expression was analyzed by using an LI-COR Odyssey scanner (Belfast, ME, USA).

H1975 and H1650 co-cultured with PBMC

Human peripheral blood mononuclear cells (PBMC)/H1975 and H1650 cells were seeded at a density of 3 × 104 cells. PBMCS were isolated from healthy donors by using Ficoll-Paque density centrifugation. Then, the obtained peripheral blood lymphocytes were added to the co-culture system at a ratio of 2:1. PBMC/lung cancer cell H1975 /H1650 co-cultured cells in six well plates were treated with evodiamine or vehicle. PBMC/H1975 (CshRNA), H1975 (MUC1-CshRNA) co-cultured cells and PBMC/H1650 (CshRNA), H1650(MUC1-CshRNA) co-cultured cells in 6 well plates were treated with evodiamine or vehicle. Cells were treated with evodiamine for 48 or 72 h. Afterward, lymphocyte cells were harvested from the co-culture system, and the T cells were stained for apoptosis assay.

Transient transfection assay

Cells were transfected by using lentiviral vectors with control shRNAand MUC1-CshRNA. Puromycin was used for optimal selection of the transfected cells. For flow cytometry for cell cycle and apoptosis analysis, cells were stained with antibody in stain buffer for 30 min (in the dark at 4 °C). Cells were analyzed by BD FACSAria III flow cytometer (BD Biosciences). The surface expression of PD-L1 and MUC1-C were detected with flow cytometry.

Protein extraction and western blotting

H1975 were treated with evodiamine for 24 h, and protein was extracted from cells by using NE-PER™ Nuclear and cytoplasm extraction reagents (Thermo Fisher Scientific, Waltham, MA, USA). The expression of PD-L1 and MUC1-C on H1975 was detected using Western blotting.

Using quantitative real-time RT-PCR to detect the mRNA expression

After Total RNA isolated, cDNAs were synthesized by a cDNA Reverse Transcription SuperMix Kit (Bio-RAD) Using the Power SYBR Green PCR Master Mix (Roche) to detect the PD-L1 and MUC1-C mRNA expression. Primers used for qPCR were reported previously [39].

Promoter-reporter assay

H1975 and H1650 cells were transfected with PD-L1 promoter-Luciferase reporter expression (pD-L1-Luc) or mock vehicle (Active Motif). After treated with evodiamine for 48 h, the cells were lysed and analyzed by using the Dual-Luciferase® Reporter Assay System (Promega, Madison, Wisconsin, USA).

IL-2 assays

H1975 and H1975 MUC1-C knockout were treated with IFN-γ for 24 h. Jurkat cells were activated and added to H1975 and H1650 cells at a ratio of 2:1 or 4:1 Jurkat: H1975/H1650. The cell culture media were collected, and the IL-2 expression was measured using ELISA-kit and flow cytometry.

Immunohistochemistry

Lung cancer patient and mouse lung cancer samples were processed for the staining of MUC1-C, PD-L1, CD4, CD8. Immunohistochemical images were captured and the numbers of MUC1-C, CD4, PD-L1, CD8, IFN-γ and Granzyme B positive cells in sections were counted.

Tumor xenograft studies and Lewis lung carcinoma model

Animal experiments were performed in accordance with the guidelines by the care and use of laboratory Animals. H1975 xenografts on female C57BL/C mice were treatment with 10, 20, 30 mg/kg of evodiamine and xenografts were allowed to grow for over 1 week when tumors were detectable with calipers before treatment by gavages.
For the Lewis lung carcinoma model, the Lewis lung carcinoma cells (5 × 105 cells) were intravenously injected into female C57BL/C mice at age 8–10 weeks. Single-cell suspensions of tumors and blood and spleen cells were stained, and cells were acquired using a flow cytometer and analyzed with flowJo software to detect tumor multiplicity in the lung.

Immunohistochemistry

Human tissue of patient with NSCLCwere collected from 2014 to 2017 in Taihe Hospital. Immunohistochemistry analyses were performed using an DAKO EnVision system as described previously. The following antibodies were used: MUC1-C from Cell Signaling (catalog no. #16564). This study about patients with NSCLC was approved by the ethics committee of Hubei Taihe Hospital.

Statistical analysis

All data in this article were expressed as the mean ± SD of three individual experiments. Differences between groups were determined by one-way analysis of variance (ANOVA) by Graph Pad Prism 8 followed by the Bonferroni test to compare all pairs of columns. When P < 0.05, the results were considered to be statistically significant in this study. Survival rates of mice in our experiment were generated based on the Kaplan–Meier method, statistical significance was determined by the log-rank test, P value < 0.05 is considered statistically significant.

Results

Evodiamine can inhibit growth and induce apoptosis of NSCLC cells

To detect the cytotoxicity of evodiamine on NSCLC cells, NSCLC cells were treated different concentration of evodiamine for 72 h. Figure 1a showed the chemical structure of evodiamine. The MTT data showed that evodiamine inhibited the growth of two cell lines (H1650 and H1975) in a dose-dependent manner (Fig. 1b). Figure 1b and Extended Data Fig. 2A show that among four cell lines (H1975, H1650, H2228 and HCC827), H1975 and H1650 are more sensitive to evodiamine, with an IC50 value of 5.59 ± 0.88 and 4.53 ± 1.46 μM, respectively, whereas H2228 and HCC827 were less sensitive. H1975 and H1650 are EGFR tyrosine kinase inhibitor resistant lung cancer cells. Evodiamine had much less cytotoxicity in the normal cell line CD19 and BEAS-2B (Fig. 1b and Extended Data Fig. 2A). To detect the apoptosis cells of evodiamine in H1650 and H1975 cells, the percentage of apoptosis cells was examined. As illustrated in Fig. 1c-d and Extended Data Fig. 1A-B, the proportion of apoptotic cells in H1650 and H1975 was significantly increased after treatment with evodiamine. Cleaved PARP is also detected (Fig. 1e), and the data show that cleaved PARP is significantly increased by evodiamine treatment for 24 h. The cell cycle distribution of evodiamine on H1975 and H1650shows that evodiamine inhibits H1650 and H1975 NSCLC cells, and it induces cell cycle arrest at G2 phase (Fig. 1 F and Extended Data Fig. 1B).

Evodiamine decreases PD-L1 and MUC1-C expression in NSCLC cells

The potential signaling that may be involved in the inhibitory response by evodiamine in NSCLC cells was explored. The results show that evodiamine decreased the expression of PD-L1 mRNA and MUC1-C mRNA in H1975 and H1650 at 24 h (Fig. 2a-d). Evodiamine specifically decreases cell surface expression of PD-L1 in H1975 and H1650 cells (Fig. 2e-h) Moreover, evodiamine decreases PD-L1 and MUC1-C protein in H1975 and H1650 cells in a dose-dependent manner (Fig. 2i-j).

Evodiamine increases cell apoptosis in NSCLC in a MUC1-C-dependent manner

The effects of targeting MUC1-C on the response of H1975 cells to evodiamine were investigated. The western blot assay results Fig. 3a demonstrate that MUC1-C was successfully knocked down in both H1975 and H1650 cell. The results of this work show that knockdown of MUC1-C in H1650 and H1975 cells results in downregulating the surface expression of PD-L1(Fig. 3b). Evodiamine-treatment induced apoptosis is significantly reduced in H1650 and H1975 with MUC1-C knockdown (Fig. 3c-f). In Fig. 3g-h), the MTT results also demonstrate that evodiamine increases cell apoptosis and inhibits growth in H1650 and H1975 in a MUC1-C dependent manner. H1975 and H1650 were transfected with control (pCMV6-AC) and MUC1-C plasmid DNA to demonstrate MUC1-C is a potential novel mechanism of evodiamine of tumor inhibition. Then cells were treated with evodiamine for an additional 24 or 72 h. Extended Data Fig. 6 data shows that expression of MUC1-C abrogates the effect of evodiamine on MUC1-C expression and cell growth inhibition. H1975 and H1650 cells were treated with evodiamine, and PD-L1 promoter activity were determined to investigate whether evodiamine activates the PD-L1 promoter. Extended Data Fig. 2B data demonstrates that evodiamine inactivates the PD-L1 promoter in H1975 and H1650.

Evodiamine inhibits the inhibition of IFN-γ-induced PD-L1 expression in H1650 and H1975

Interferon-γ (IFN-γ) is an important cytokine with different pleiotropic in the tumor microenvironment, has long been praised as an effector cytokine of anti-cancer immunity and can inhibit cancer development and growth through various mechanisms. However, IFN-γ has also been involved in tumor cell development and transformation. IFN-γ also helps cancer cells evade immune destruction, suggesting that IFN-γ has an existing side effect of promoting tumor. Reports showed that IFN-γcan stimulate the expression of PD-L1 in tumor cells, which bind with PD-1, inhibiting activation, expansion, and effector functions of CD8+ T cells, and helping cancer cells evade immune destruction to contribute to tumor immune evasion [40]. To effect of evodiamine on the IFN-γ-induced upregulation of PD-L1 expression in H1650 and H1975 cells was assessed. The optimal induction of PD-L1 was obtained after 24 h treatment with 50 ng/ml IFN-γ, Pretreatment of H1650 cells with 8–16 μM evodiamine inhibited IFN-γ-induced PD-L1 and MUC1-C mRNA expression (Fig. 4a). The experiment was repeated with H1975 that also showed inducible expression PD-L1 as well as H1650 cells that constitutively expressed PD-L1 (Fig. 4b) to confirm that the effect of evodiamine on PD-L1 expression is not cell line specific. PD-L1 expression on the cell membrane plays an important role in tumor escape. The interaction between PD-1 on T cells and PD-L1 on tumor cells on cell membrane inhibits activation, expansion, and effector functions of CD8+ T cells and helps cancer cells evade immune destruction. Remarkably, overexpression of PD-L1 in human cancers is associated with poor clinical outcomes. Thus, flow cytometry was used to examine the surface expression of PD-L1 on H1650 and H1975 cells after evodiamine treatment, and flow data show that evodiamine specifically decreases IFN-γ-induced surface expression of PD-L1 in H650 and H1975 (Fig. 4c-f). Moreover, pre-treatment of H1650 cell lines cells with 8–16 μM evodiamine can decrease the IFN-γ-induced PD-L1 protein expression in H1650 and H1975 cells (Fig. 4g-h).

MUC1-C inhibition could diminish the PD-L1 expression to decrease the T cell apoptosis

PBMC cells were co-cultured with H1650 or H1975 cells, both expressing high PD-L1. The results show that the apoptosis rate of T cells increased (Extended Data Fig. 3). Extended Data Fig. 3A-D show that evodiamine can elevate CD8+ T cell function by increasing IFN-γ and GzmB expression. MUC1-C expression was silenced with shRNA in H1975 and H1650 cells to understand whether MUC1-C could affect T cells proliferation and apoptosis when they were co-cultured with H1975 and H1650 cells. Extended Data Fig. 3E-F showthat the apoptosis of T cells in co-culture assay decreased. These results indicate that inhibiting MUC1-C in H1650 and H1975 could reverse the apoptosis of T cells. Evodiamine, which inhibits MUC1-C in H1650 and H1975, also reversed the apoptosis of T cells. We hypothesize that evodiamine downregulates PD-L1 expression in H1975 and H1650 by targeting the MUC1-C. In our T cell-mediated lung cancer cell killing assays, evodiamine-treated H650 and H1975 cells displayed enhanced sensitivity towards activated Jurkat T cells. In our study, evodiamine could significantly increase of IL-2 secreted detected from co-cultures (Extended Data Fig. 2C).

Evodiamine inhibits H1975 tumor xenograft tumor growth by downregulation of MUC1-C

H1975 xenograft was performed on female nude mice in this experiment to verify the effect of evodiamine in vivo. The body weight of evodiamine-treated mice did not change when compared to untreated animals (Fig. 5a). Compared with the controls, tumor growth control showed a suppressive trend starting at day 15 in the evodiamine-treated mice, with smaller tumor volume (Fig. 5b), size (Fig. 5c) and weight (Fig. 5d). The western blot analysis in Fig. 5e shows that MUC1-C downregulation after evodiamine treatment. Consistent with novel discovery in vitro ((Fig. 5f)), the levels of MUC1-C were all downregulated by evodiamine treatment. These data altogether show that evodiamine can inhibit NSCLC growth in vivo and in vitro at least partially by downregulation of MUC1-C.

Evodiamine exhibits anti-tumor activity by elevation of CD8+ T cells in vivo

The Lewis lung carcinoma model was used to study the activity of evodiamine on CD8+ T cells in controlling tumor progression and metastasis to verify the effect of cancer immunotherapies in mice with evodiamine. The body weight of evodiamine-treated mice did not change compared to untreated animals (Fig. 6a).
Evodiamine improved the survival of the mice (Fig. 6b). Compared with the controls, tumor growth control showed a suppressive trend starting at day 18 in the evodiamine-treated mice (Fig. 6c) and have small volume (Fig. 6c) and size (Fig. 6d) and weight (Fig. 6e). The tumor-infiltrating T cells in treatment group were analyzed, and the CD8+ T cells had better activity and increased cell numbers compared with the control group (Fig. 6f-g). In the lung mice model, the CD8+ T cells of the evodiamine treatment mice had higher activity than those of control mice (Fig. 6f-h, Extended Data Fig. 4). Evodiamine could enhance the effector function of mouse CD8+ T cells in vivo. The number of tumor-infiltrating CD8+ T cells in evodiamine-treated mice increased (Fig. 6f-g, Extended Data Fig. 4L), and these cells showed potentiated effector function.

Combination evodiamine and PD-1 mAb treatment enhanced tumor growth control and survival of Lewis lung carcinoma model

Next, the possibility of combination evodiamine and PD-1 mAb to improve the therapeutic effects of either drug alone was investigated. Treatment with combination evodiamine and PD-1 mAb improve the survival of the mice (Fig. 7a). The body weight of evodiamine-treated mice did not change compared to untreated animals (Fig. 7b). The results show that a single PD-1 mAb treatment failed to restrain tumor growth in the mouse model effectively (Fig. 7c-e), and mice treated with a combination of evodiamine and PD-1 mAb had small volume (Fig. 7c) and size (Fig. 7d) and weight (Fig. 7e). These data revealed that the percentage of CD4+ T cells in blood, spleen or tumor is largely unchanged by either evodiamine or anti-PD-1 treatment or the combination (Extended Data Fig. 5). By contrast, we found significantly more tumor-infiltrating CD8+ T cells measured by frequency and number in the combination PD-1 antibody and evodiamine treatment group (Fig. 7f). We also found that in the combination PD-1 antibody and evodiamine treatment group, tumor-infiltrating CD8+ T cells showed stronger activation phenotypes and more increased IFN-γ, TNFα and Granzyme B (GrzmB) production (Fig. 7f). Extended Data Fig. 5C shows the proportion of regulatory T (Treg) cells in combination evodiamine and PD-1 mAb treatment (CD4+FoxP3+T cell) in the spleen, blood and tumor is decreased (Fig. 6f and Extended Data Fig. 5).

MUC1-C expression is highly expressed in NSCLC, have high positive rates in female, non-smoker, adenocarcinoma and advanced stage

Finally, the clinical relevance of MUC1-C in tumor of patient was investigated, and MUC1-C expression in 131 of patients with NSCLC was detected using immunohistochemical method. MUC1-C was higher in patients with late-stage (III and IV) than in the patients with early-stage (I) tumors (Table. 1). A high level of MUC1-C is associated with advanced lung cancer progression. These results indicated that MUC1-C is highly expressed in NSCLC, has high positive rates in female, non-smoker, adenocarcinoma and advanced-stage patients. Compared with healthy donors, NSCLC tumor tissue has high PD-L1 and MUC1-C mRNA expression (Extended Data Fig. 2D-E).
Table 1
Clinical pathological characteristics correlation analysis with MUC1-C expression in a 131 NSCLC cohort
Characteristics
Total
MUC1-C expression
F/X2
P-value
Negative (%)
Low (%)
High (%)
Age (years)
    
0.776
0.678
 < =65
105
7 (6.7)
32 (30.5)
66 (62.9)
  
 > 65
26
1 (3.8)
10 (38.5)
15 (57.7)
  
Gender
    
16.215
< 0.001
 Male
105
8 (7.6)
41 (39.0)
56 (53.3)
  
 Female
26
0 (0.0)
1 (3.8)
25 (96.2)
  
Smoking history
    
17.501
< 0.001
 Yes
90
8 (8.9)
37 (41.1)
45 (50.0)
  
 No
41
0 (0.0)
5 (12.2)
36 (87.8)
  
Pathological pattern
    
60.153
< 0.001
 Adenocarcinoma
74
0 (0.0)
7 (9.5)
67 (90.5)
  
 Squamous cell
57
8 (14.0)
35 (61.4)
14 (24.6)
  
Cell differentiation
    
5.542
0.236
 Poorly
28
1 (3.6)
12 (42.9)
15 (53.6)
  
 Moderately
93
5 (5.4)
28 (30.1)
60 (64.5)
  
 Well
10
2 (20.0)
2 (20.0)
6 (60.0)
  
TNM stage
    
13.306
0.038
 I
62
4 (6.5)
18 (29.0)
40 (64.5)
  
 II
37
3 (8.1)
19 (51.4)
15 (40.5)
  
 III
28
1 (3.6)
5 (17.9)
22 (78.6)
  
 IV
4
0 (0.0)
0 (0.0)
4 (100.0)
  

Discussion

In the present study paper, we demonstrated that evodiamine not only significantly inhibits proliferation and increases apoptotic but also shows good anti-tumor activity, which reduces tumor size in H1975 tumor xenograft and Lewis lung carcinoma model,. We demonstrated that knock out the MUC1-C in H1650 and H1975, both expressing high MUC1-C, can increase IC50 of evodiamine effected on these two cell lines. In our experiment, we observed the inhibitory effect of PD-L1 mRNA and membrane PD-L1 following treatment with evodiamine in IFN-γ stimulation H1650 and H1975. Activation PD-L1 could, through the PD-L1/PD-1 axis could induce the T cells apoptosis in the co-culture system. Evodiamine could downregulate the PD-L1 expression to diminish the apoptosis of T cells. It inhibited MUC1-C expression and potentiated CD8+ T-cell effector function. Evodiamine exhibited anti-tumor activity by elevating of CD8+ T cells in vivo in the Lewis lung carcinoma model. We observed that combination of evodiamine and anti-PD-1 mAb treatment enhanced tumor growth control and survival.
The potential impact of Chinese herbal medicines and their ingredients in numerous types of tumor treatment has attracted widespread attention [41]. Extensive investigations demonstrated that evodiamine, an alkaloid extracted from Euodia rutaecarpa, inhibit the proliferation of various tumors and induce tumor cell apoptosis [42]. Evodiamine isthe one of the most popular, multi-purpose Chinese herbal medicine for the treatment of many indications, including headaches, menstruation disorder, amenorrhea, abdominal, pain, diarrhea, vomiting, postpartum hemorrhage, gastrointestinal disorders and others [4345]. Evodiamine, as a novel occurring indole alkaloid with attractive multi-targeting antiproliferative activity, has been investigated as a leading compound that possesses multi-targeting profiles [43, 44, 46]. Evodiamine derivatives with Novel boron-containing were designed, which have improved the anti-tumor potency of the evodiamine and showed a good antitumor activity in vitro and in vivo by reactive oxygen species (ROS) [43]. More and more evidence have reported that evodiamine has inhibitory effects on lung tumor growth and metastasis by suppressed cell viability, induced G2/M cell cycle arrest and inhibited cell migration [45]. However, the anti-cancer mechanisms of evodiamine on the immune checkpoint PD-1/PD-L1 axis and its immune effects remain underexplored until now. We confirmed that in the current study evodiamine increases cell apoptosis in NSCLC in a MUC1-C-dependent manner. Evodiamine has a good anti-cancer tumor function by inhibiting NF-κB [47, 48]. A Bouillez reported that MUC1-C can drives the transcription of CD274 in tumor cells through NF-κBp65 or C-MYC on the PD-L1 promoter, resulting in integrating PD-L1 activation on cancer cells with suppression of immune effectors on T cells and then helping cancer cells immune evasion and poor clinical outcome [31, 39]. Our data demonstrated that evodiamine could downregulate MUC1-C and decrease the PD-L1 promoter in H1975 and H1650 cells (Extended Data Fig. 2B and Extended Data Fig. 6), leading to inhibiting MUC1-C expression and potentiating CD8+ T-cell effector function.
Tumor vaccine fused with soluble PD-1 with the MUC1 gene showed good immunogenicity and anti-tumor effect by enhancing the activation of lymphocytes, and accumulates CD8+ tumor-infiltrating lymphocytes, resulting in reduced in tumor growth [49, 50]. MUC1 has been considered a possible immunotherapeutic target. TG4010, a therapeutic cancer vaccine, expressing MUC1 as well as interleukin 2, and is combine with PD-L1 immune inhibitor nivolumab, has been approved by the FDA for the first-line treatment of patients with NSCLC [51]. Chimeric antigen receptors (CARs) targeting T cells to MUC1 were developed and demonstrated the therapeutic efficacy of CAR T cells directed against Tn-MUC1 and presented aberrantly glycosylated antigens as a clinical trial to target solid cancers which express MUC1 with CAR T cells [5254]. Consistently, Xiuling Xu reported that knockdown of MUC1-C expression in A549 and H460 effectively increases the sensitivity of these cells to the apoptotic cytotoxicity of anti-cancer therapeutics, suggesting that MUC1-C may contribute to acquired chemoresistance [28]. IFN-γ, an important cytokine in tumor microenvironments, is secreted from various types of immune cells such as T cells, activate macrophages, and B cells and natural killer (NK) cells. IFN-γ could potentially increase the expression of PD-L1 in tumor cells, which biding with PD-1, in resulting inhibiting activation, expansion, and effector functions of CD8+ T cells and helps cancer cells evade immune destruction so that to contribute to tumor immune evasion. Similarly, in our T cell-mediated lung cancer cell killing assays, evodiamine-treated H650 and H1975 cells displayed enhanced sensitivity towards activated Jurkat T cells. In our study, evodiamine could block the interaction of the PD-1/PD-L1 axis, resulting in a significant increase of IL-2 secreted detected from co-cultures. In addition to its impact on cell membrane signaling, MUC1-C is imported to the nucleus, where it associates with a different transcription factor, including p53, NF-κB p65, STAT1/3, c-MYC, HIF-1 among others. Xiuling Xu reported that knockdown of MUC1-C expression in A549 and H460 effectively increased the sensitivity of these cells to the apoptotic cytotoxicity of anti-cancer therapeutics, suggesting that MUC1-C may contribute to acquired chemoresistance. In our study, we found that silencing the MUC1-C in H1975 and H1650, both expressing high MUC1-C, can attenuate the cytotoxicity effect of Evodiamine. MUC1-C is a transmembrane subunit of the MUC1 glycoprotein, which can translocate into the nucleus and trigger expression of other cancer-related oncogenes. Interestingly, we also observed an inhibitory effect of Evodiamine on the expression of PD-L1 mRNA and membrane PD-L1 following IFN-γ stimulation. Similarly, we found that evodiamine inhibits the translocation of MUC1-C to the nucleus and suppress PD-L1 expression as well in the nuclei of NSCLC cells with IFN-γ-caused PD-L1 expression [31]. Evodiamine inhibits MUC1-C expression in the nucleus, where it associates with various transcription factor [55].
MUC1-C is a transmembrane glycoprotein, which is aberrantly expressed in > 80% Stage IB NSCLC [31, 55, 56]. Our IHC results are incompatible with those mentioned above. It should be noted that stage IB lung carcinomas were involved in Dongrong Situ et al.’s study [31]. By contrasts, our study focused on population of patients with I–IV lung carcinomas diseases, including adenocarcinoma and Squamous Cell Cancer (Table.1). We found the MUC1-C expression has high positive rates in female, non-smoker patients with advanced stage patients. Here, We reported a novel finding that evodiamine inhibits PD-L1 expression in lung adenocarcinoma by targeting MUC1-C. Suppression of MUC1-C expression via MUC1-C shRNA resulted in a decrease of PD-L1 protein and mRNA expression [57]. Evodiamine can also decrease MUC1-C and PD-L1 protein and mRNA levels in NSCLC through inhibiting MUC1-C expression. Silencing MUC1 is associated with a C-MYC decrease of PD-L1 mRNA expression, suggesting that MUC1-C regulation of PD-L1 is likely mediated by transcriptional mechanism [31]. GO203, targeting MUC1-C, can suppress PD-L1 expression of NSCLC and breast cancer, and induces effectors of innate and adaptive immunity, resulting in improve anticancer effects [31] We determined that evodiamine can block PD-L1 expression on mRNA and protein levels, perhaps through inhibiting the MUC1-C. Silencing MUC1-C in H1975 decreases apoptosis following evodiamine treatment. Currently, several preclinical and clinical trials for the combination of other therapy with immune inhibitors in the treatment patients reveal biomarkers of response and resistance to anti-PD-1 monotherapy and combined anti-CTLA-4 and anti-PD-1 immunotherapy, significantly improve the anti-tumor effect [6, 5861]. Collectively, the combination of evodiamine and PD-1 mAb treatment enhance anti-cancer and survival in a Lewis lung carcinoma model.

Conclusions

Overall, our results show that evodiamine suppresses NSCLC by elevation of CD8+ T cells and downregulation of the MUC1-C/PD-L1 axis. This paper corroborates a potential novel mechanism of evodiamine that controls Lung adenocarcinoma cell growth. Our data implies that the rational targeting MUC1-C combined with immunotherapeutic approaches is a potential therapeutic strategy against lung cancer. In our clinical pathological characteristic correlation study, the result indicated that MUC1-C is highly expressed in NSCLC, has high positive rates in female, non-smoker patients with advanced-stage adenocarcinoma. Blocking the MUC1-C/PDL-1 axis is a potential new therapeutic strategy for these subgroup of advanced stage patients.

Acknowledgments

This work was supported by The Science and Technology Development Fund, Macau SAR (Project no: 0096/2018/A3), the NSFC overseas and Hong Kong and Macao Scholars Cooperative Research Fund Project (Project no: 81828013) and Taihe Hospital with internal funding and the Science and Technology Development Fund, Macau SAR (Project no: 0096/2018/A3, 130/2017/A3, and 0099/2018/A3).
All animal experiments were in accordance with institutional animal care guidelines and conducted according to the committee-approved protocols of the State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute. (Macau, China). All procedures in studies involving human participants were performed in accordance with the ethical standards and approved by the ethics committee of Hubei Taihe Hospital.
Not applicable.

Competing interests

All the authors have no financial or commercial conflict of interest to disclose.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.CrossRefPubMed Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.CrossRefPubMed
2.
Zurück zum Zitat Barta JA, Powell CA, Wisnivesky JP. Global epidemiology of lung cancer. Ann Glob Health. 2019;85:1.CrossRef Barta JA, Powell CA, Wisnivesky JP. Global epidemiology of lung cancer. Ann Glob Health. 2019;85:1.CrossRef
3.
4.
Zurück zum Zitat Horton BL, Williams JB, Cabanov A, Spranger S, Gajewski TF. Intratumoral CD8(+) T-cell apoptosis is a major component of T-cell dysfunction and impedes antitumor immunity. Cancer Immunol Res. 2018;6(1):14–24.PubMedCrossRef Horton BL, Williams JB, Cabanov A, Spranger S, Gajewski TF. Intratumoral CD8(+) T-cell apoptosis is a major component of T-cell dysfunction and impedes antitumor immunity. Cancer Immunol Res. 2018;6(1):14–24.PubMedCrossRef
5.
Zurück zum Zitat Zhao Y, Lee CK, Lin CH, Gassen RB, Xu X, Huang Z, et al. PD-L1:CD80 Cis-heterodimer triggers the co-stimulatory receptor CD28 while repressing the inhibitory PD-1 and CTLA-4 pathways. Immunity. 2019;51(6):1059–73 e1059.PubMedPubMedCentralCrossRef Zhao Y, Lee CK, Lin CH, Gassen RB, Xu X, Huang Z, et al. PD-L1:CD80 Cis-heterodimer triggers the co-stimulatory receptor CD28 while repressing the inhibitory PD-1 and CTLA-4 pathways. Immunity. 2019;51(6):1059–73 e1059.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Gide TN, Quek C, Menzies AM, Tasker AT, Shang P, Holst J, et al. Distinct immune cell populations define response to anti-PD-1 Monotherapy and anti-PD-1/anti-CTLA-4 combined therapy. Cancer Cell. 2019;35(2):238–255.e236.PubMedCrossRef Gide TN, Quek C, Menzies AM, Tasker AT, Shang P, Holst J, et al. Distinct immune cell populations define response to anti-PD-1 Monotherapy and anti-PD-1/anti-CTLA-4 combined therapy. Cancer Cell. 2019;35(2):238–255.e236.PubMedCrossRef
7.
Zurück zum Zitat Bauml JM, Mick R, Ciunci C, Aggarwal C, Davis C, Evans T, et al. Pembrolizumab after completion of locally ablative therapy for Oligometastatic non-small cell lung cancer: a phase 2 trial. JAMA Oncol. 2019;5(9):1283.PubMedCentralCrossRefPubMed Bauml JM, Mick R, Ciunci C, Aggarwal C, Davis C, Evans T, et al. Pembrolizumab after completion of locally ablative therapy for Oligometastatic non-small cell lung cancer: a phase 2 trial. JAMA Oncol. 2019;5(9):1283.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Andrews LP, Yano H, Vignali DAA. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: breakthroughs or backups. Nat Immunol. 2019;20(11):1425–34.PubMedCrossRef Andrews LP, Yano H, Vignali DAA. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: breakthroughs or backups. Nat Immunol. 2019;20(11):1425–34.PubMedCrossRef
9.
Zurück zum Zitat Dillon MT, Bergerhoff KF, Pedersen M, Whittock H, Crespo-Rodriguez E, Patin EC, et al. ATR inhibition potentiates the radiation-induced inflammatory tumor microenvironment. Clin Cancer Res. 2019;25(11):3392–403.PubMedPubMedCentralCrossRef Dillon MT, Bergerhoff KF, Pedersen M, Whittock H, Crespo-Rodriguez E, Patin EC, et al. ATR inhibition potentiates the radiation-induced inflammatory tumor microenvironment. Clin Cancer Res. 2019;25(11):3392–403.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat McLaughlin M, Patin EC, Pedersen M, Wilkins A, Dillon MT, Melcher AA, et al. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer. 2020;20(4):203–17.PubMedCrossRef McLaughlin M, Patin EC, Pedersen M, Wilkins A, Dillon MT, Melcher AA, et al. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer. 2020;20(4):203–17.PubMedCrossRef
11.
Zurück zum Zitat Jiang X, Wang J, Deng X, Xiong F, Ge J, Xiang B, et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer. 2019;18(1):10.PubMedPubMedCentralCrossRef Jiang X, Wang J, Deng X, Xiong F, Ge J, Xiang B, et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer. 2019;18(1):10.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Menares E, Galvez-Cancino F, Caceres-Morgado P, Ghorani E, Lopez E, Diaz X, et al. Tissue-resident memory CD8(+) T cells amplify anti-tumor immunity by triggering antigen spreading through dendritic cells. Nat Commun. 2019;10(1):4401.PubMedPubMedCentralCrossRef Menares E, Galvez-Cancino F, Caceres-Morgado P, Ghorani E, Lopez E, Diaz X, et al. Tissue-resident memory CD8(+) T cells amplify anti-tumor immunity by triggering antigen spreading through dendritic cells. Nat Commun. 2019;10(1):4401.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat van der Leun AM, Thommen DS, Schumacher TN. CD8(+) T cell states in human cancer: insights from single-cell analysis. Nat Rev Cancer. 2020;20(4):218–32.PubMedPubMedCentralCrossRef van der Leun AM, Thommen DS, Schumacher TN. CD8(+) T cell states in human cancer: insights from single-cell analysis. Nat Rev Cancer. 2020;20(4):218–32.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Petitprez F, de Reynies A, Keung EZ, Chen TW, Sun CM, Calderaro J, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577(7791):556–60.CrossRefPubMed Petitprez F, de Reynies A, Keung EZ, Chen TW, Sun CM, Calderaro J, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577(7791):556–60.CrossRefPubMed
15.
Zurück zum Zitat La-Beck NM, Nguyen DT, Le AD, Alzghari SK, Trinh ST. Optimizing patient outcomes with PD-1/PD-L1 immune checkpoint inhibitors for the first-line treatment of advanced non-small cell lung cancer. Pharmacotherapy. 2020;40(3):239–55.PubMedCrossRef La-Beck NM, Nguyen DT, Le AD, Alzghari SK, Trinh ST. Optimizing patient outcomes with PD-1/PD-L1 immune checkpoint inhibitors for the first-line treatment of advanced non-small cell lung cancer. Pharmacotherapy. 2020;40(3):239–55.PubMedCrossRef
16.
Zurück zum Zitat Martins F, Sofiya L, Sykiotis GP, Lamine F, Maillard M, Fraga M, et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol. 2019;16(9):563–80.PubMedCrossRef Martins F, Sofiya L, Sykiotis GP, Lamine F, Maillard M, Fraga M, et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol. 2019;16(9):563–80.PubMedCrossRef
17.
Zurück zum Zitat Nath PR, Pal-Nath D, Mandal A, Cam MC, Schwartz AL, Roberts DD. Natural killer cell recruitment and activation are regulated by CD47 expression in the tumor microenvironment. Cancer Immunol Res. 2019;7(9):1547–61.PubMedPubMedCentralCrossRef Nath PR, Pal-Nath D, Mandal A, Cam MC, Schwartz AL, Roberts DD. Natural killer cell recruitment and activation are regulated by CD47 expression in the tumor microenvironment. Cancer Immunol Res. 2019;7(9):1547–61.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Lu J, Xu Y, Wu Y, Huang XY, Xie JW, Wang JB, et al. Tumor-infiltrating CD8+ T cells combined with tumor-associated CD68+ macrophages predict postoperative prognosis and adjuvant chemotherapy benefit in resected gastric cancer. BMC Cancer. 2019;19(1):920.PubMedPubMedCentralCrossRef Lu J, Xu Y, Wu Y, Huang XY, Xie JW, Wang JB, et al. Tumor-infiltrating CD8+ T cells combined with tumor-associated CD68+ macrophages predict postoperative prognosis and adjuvant chemotherapy benefit in resected gastric cancer. BMC Cancer. 2019;19(1):920.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Bouillez A, Adeegbe D, Jin C, Hu X, Tagde A, Alam M, et al. MUC1-C promotes the suppressive immune microenvironment in non-small cell lung cancer. OncoImmunology. 2017;6:9.CrossRef Bouillez A, Adeegbe D, Jin C, Hu X, Tagde A, Alam M, et al. MUC1-C promotes the suppressive immune microenvironment in non-small cell lung cancer. OncoImmunology. 2017;6:9.CrossRef
21.
Zurück zum Zitat Shimizu S, Hiratsuka H, Koike K, Tsuchihashi K, Sonoda T, Ogi K, et al. Tumor-infiltrating CD8(+) T-cell density is an independent prognostic marker for oral squamous cell carcinoma. Cancer Med. 2019;8(1):80–93.PubMedPubMedCentralCrossRef Shimizu S, Hiratsuka H, Koike K, Tsuchihashi K, Sonoda T, Ogi K, et al. Tumor-infiltrating CD8(+) T-cell density is an independent prognostic marker for oral squamous cell carcinoma. Cancer Med. 2019;8(1):80–93.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541(7637):321–30.PubMedCrossRef Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541(7637):321–30.PubMedCrossRef
23.
Zurück zum Zitat Xin Z, Xin G, Shi M, Song L, Wang Q, Jiang B, et al. Inhibition of MUC1-C entering nuclear suppresses MYC expression and attenuates malignant growth in esophageal squamous cell carcinoma. Onco Targets Ther. 2018;11:4125–36.PubMedPubMedCentralCrossRef Xin Z, Xin G, Shi M, Song L, Wang Q, Jiang B, et al. Inhibition of MUC1-C entering nuclear suppresses MYC expression and attenuates malignant growth in esophageal squamous cell carcinoma. Onco Targets Ther. 2018;11:4125–36.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Horm TM, Schroeder JA. MUC1 and metastatic cancer: expression, function and therapeutic targeting. Cell Adhes Migr. 2013;7(2):187–98.CrossRef Horm TM, Schroeder JA. MUC1 and metastatic cancer: expression, function and therapeutic targeting. Cell Adhes Migr. 2013;7(2):187–98.CrossRef
25.
Zurück zum Zitat Kato K, Lillehoj EP, Kim KC. MUC1 regulates epithelial inflammation and apoptosis by PolyI:C through inhibition of toll/IL-1 receptor-domain-containing adapter-inducing IFN-beta (TRIF) recruitment to toll-like receptor 3. Am J Respir Cell Mol Biol. 2014;51(3):446–54.PubMedPubMedCentralCrossRef Kato K, Lillehoj EP, Kim KC. MUC1 regulates epithelial inflammation and apoptosis by PolyI:C through inhibition of toll/IL-1 receptor-domain-containing adapter-inducing IFN-beta (TRIF) recruitment to toll-like receptor 3. Am J Respir Cell Mol Biol. 2014;51(3):446–54.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Krieg C, et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 2018;24(2):144.PubMedCrossRef Krieg C, et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 2018;24(2):144.PubMedCrossRef
27.
Zurück zum Zitat Shukla SK, Purohit V, Mehla K, Gunda V, Chaika NV, Vernucci E, et al. MUC1 and HIF-1alpha signaling crosstalk induces anabolic glucose metabolism to impart gemcitabine resistance to pancreatic cancer. Cancer Cell. 2017;32(1):71–87.e77.PubMedPubMedCentralCrossRef Shukla SK, Purohit V, Mehla K, Gunda V, Chaika NV, Vernucci E, et al. MUC1 and HIF-1alpha signaling crosstalk induces anabolic glucose metabolism to impart gemcitabine resistance to pancreatic cancer. Cancer Cell. 2017;32(1):71–87.e77.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Xu X, Wells A, Padilla MT, Kato K, Kim KC, Lin Y. A signaling pathway consisting of miR-551b, catalase and MUC1 contributes to acquired apoptosis resistance and chemoresistance. Carcinogenesis. 2014;35(11):2457–66.PubMedPubMedCentralCrossRef Xu X, Wells A, Padilla MT, Kato K, Kim KC, Lin Y. A signaling pathway consisting of miR-551b, catalase and MUC1 contributes to acquired apoptosis resistance and chemoresistance. Carcinogenesis. 2014;35(11):2457–66.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Kufe DW. MUC1-C oncoprotein as a target in breast cancer: activation of signaling pathways and therapeutic approaches. Oncogene. 2013;32(9):1073–81.PubMedCrossRef Kufe DW. MUC1-C oncoprotein as a target in breast cancer: activation of signaling pathways and therapeutic approaches. Oncogene. 2013;32(9):1073–81.PubMedCrossRef
31.
Zurück zum Zitat Bouillez A, Rajabi H, Jin C, Samur M, Tagde A, Alam M, et al. MUC1-C integrates PD-L1 induction with repression of immune effectors in non-small-cell lung cancer. Oncogene. 2017;36(28):4037–46.PubMedPubMedCentralCrossRef Bouillez A, Rajabi H, Jin C, Samur M, Tagde A, Alam M, et al. MUC1-C integrates PD-L1 induction with repression of immune effectors in non-small-cell lung cancer. Oncogene. 2017;36(28):4037–46.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Mohan V, Agarwal R, Singh RP. A novel alkaloid, evodiamine causes nuclear localization of cytochrome-c and induces apoptosis independent of p53 in human lung cancer cells. Biochem Biophys Res Commun. 2016;477(4):1065–71.PubMedCrossRef Mohan V, Agarwal R, Singh RP. A novel alkaloid, evodiamine causes nuclear localization of cytochrome-c and induces apoptosis independent of p53 in human lung cancer cells. Biochem Biophys Res Commun. 2016;477(4):1065–71.PubMedCrossRef
33.
Zurück zum Zitat Meng T, Fu S, He D, Hu G, Gao X, Zhang Y, et al. Evodiamine inhibits lipopolysaccharide (LPS)-induced inflammation in BV-2 cells via regulating AKT/Nrf2-HO-1/NF-kappaB signaling Axis. Cell Mol Neurobiol. 2020. Meng T, Fu S, He D, Hu G, Gao X, Zhang Y, et al. Evodiamine inhibits lipopolysaccharide (LPS)-induced inflammation in BV-2 cells via regulating AKT/Nrf2-HO-1/NF-kappaB signaling Axis. Cell Mol Neurobiol. 2020.
34.
Zurück zum Zitat Yang S, Chen J, Tan T, Wang N, Huang Y, Wang Y, et al. Evodiamine exerts anticancer effects against 143B and MG63 cells through the Wnt/beta-catenin signaling pathway. Cancer Manag Res. 2020;12:2875–88.PubMedPubMedCentralCrossRef Yang S, Chen J, Tan T, Wang N, Huang Y, Wang Y, et al. Evodiamine exerts anticancer effects against 143B and MG63 cells through the Wnt/beta-catenin signaling pathway. Cancer Manag Res. 2020;12:2875–88.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Deng JD, Lei S, Jiang Y, Zhang HH, Hu XL, Wen HX, et al. A concise synthesis and biological study of evodiamine and its analogues. Chem Commun (Camb). 2019;55(21):3089–92.CrossRef Deng JD, Lei S, Jiang Y, Zhang HH, Hu XL, Wen HX, et al. A concise synthesis and biological study of evodiamine and its analogues. Chem Commun (Camb). 2019;55(21):3089–92.CrossRef
36.
Zurück zum Zitat Shen P, Zhang Z, Zhu K, Cao H, Liu J, Lu X, et al. Evodiamine prevents dextran sulfate sodium-induced murine experimental colitis via the regulation of NF-kappaB and NLRP3 inflammasome. Biomed Pharmacother. 2019;110:786–95.PubMedCrossRef Shen P, Zhang Z, Zhu K, Cao H, Liu J, Lu X, et al. Evodiamine prevents dextran sulfate sodium-induced murine experimental colitis via the regulation of NF-kappaB and NLRP3 inflammasome. Biomed Pharmacother. 2019;110:786–95.PubMedCrossRef
37.
Zurück zum Zitat Li X, Fan XX, Jiang ZB, Loo WT, Yao XJ, Leung EL, et al. Shikonin inhibits gefitinib-resistant non-small cell lung cancer by inhibiting TrxR and activating the EGFR proteasomal degradation pathway. Pharmacol Res. 2017;115:45–55.PubMedCrossRef Li X, Fan XX, Jiang ZB, Loo WT, Yao XJ, Leung EL, et al. Shikonin inhibits gefitinib-resistant non-small cell lung cancer by inhibiting TrxR and activating the EGFR proteasomal degradation pathway. Pharmacol Res. 2017;115:45–55.PubMedCrossRef
38.
Zurück zum Zitat Leung EL, Fan XX, Wong MP, Jiang ZH, Liu ZQ, Yao XJ, et al. Targeting tyrosine kinase inhibitor-resistant non-small cell lung cancer by inducing epidermal growth factor receptor degradation via methionine 790 oxidation. Antioxid Redox Signal. 2016;24(5):263–79.PubMedPubMedCentralCrossRef Leung EL, Fan XX, Wong MP, Jiang ZH, Liu ZQ, Yao XJ, et al. Targeting tyrosine kinase inhibitor-resistant non-small cell lung cancer by inducing epidermal growth factor receptor degradation via methionine 790 oxidation. Antioxid Redox Signal. 2016;24(5):263–79.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Maeda THM, Jin C, et al. MUC1-C induces PD-L1 and immune evasion in triple-negative breast cancer. Cancer Res. 2018;78(1):205–15.PubMedCrossRef Maeda THM, Jin C, et al. MUC1-C induces PD-L1 and immune evasion in triple-negative breast cancer. Cancer Res. 2018;78(1):205–15.PubMedCrossRef
40.
Zurück zum Zitat Burr ML, Sparbier CE, Chan YC, Williamson JC, Woods K, Beavis PA, et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature. 2017;549(7670):101–5.PubMedPubMedCentralCrossRef Burr ML, Sparbier CE, Chan YC, Williamson JC, Woods K, Beavis PA, et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature. 2017;549(7670):101–5.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Liu W, Yang B, Yang L, Kaur J, Jessop C, Fadhil R, et al. Therapeutic effects of ten commonly used Chinese herbs and their bioactive compounds on cancers. Evid Based Complement Alternat Med. 2019;2019:6057837.PubMedPubMedCentral Liu W, Yang B, Yang L, Kaur J, Jessop C, Fadhil R, et al. Therapeutic effects of ten commonly used Chinese herbs and their bioactive compounds on cancers. Evid Based Complement Alternat Med. 2019;2019:6057837.PubMedPubMedCentral
42.
Zurück zum Zitat Zhao S, Xu K, Jiang R, Li DY, Guo XX, Zhou P, et al. Evodiamine inhibits proliferation and promotes apoptosis of hepatocellular carcinoma cells via the hippo-yes-associated protein signaling pathway. Life Sci. 2020;251:117424.PubMedCrossRef Zhao S, Xu K, Jiang R, Li DY, Guo XX, Zhou P, et al. Evodiamine inhibits proliferation and promotes apoptosis of hepatocellular carcinoma cells via the hippo-yes-associated protein signaling pathway. Life Sci. 2020;251:117424.PubMedCrossRef
43.
Zurück zum Zitat Li X, Wu S, Dong G, Chen S, Ma Z, Liu D, et al. Natural product Evodiamine with borate trigger unit: discovery of potent antitumor agents against colon cancer. ACS Med Chem Lett. 2020;11(4):439–44.PubMedCrossRefPubMedCentral Li X, Wu S, Dong G, Chen S, Ma Z, Liu D, et al. Natural product Evodiamine with borate trigger unit: discovery of potent antitumor agents against colon cancer. ACS Med Chem Lett. 2020;11(4):439–44.PubMedCrossRefPubMedCentral
44.
Zurück zum Zitat Hu X, Li D, Chu C, Li X, Wang X, Jia Y, et al. Evodiamine inhibits lipopolysaccharide (LPS)-induced inflammation in BV-2 cells via regulating AKT/Nrf2-HO-1/NF-kappaB signaling Axis. Int J Mol Sci. 2018;19:11. Hu X, Li D, Chu C, Li X, Wang X, Jia Y, et al. Evodiamine inhibits lipopolysaccharide (LPS)-induced inflammation in BV-2 cells via regulating AKT/Nrf2-HO-1/NF-kappaB signaling Axis. Int J Mol Sci. 2018;19:11.
45.
Zurück zum Zitat Su T, Yang X, Deng JH, Huang QJ, Huang SC, Zhang YM, et al. Evodiamine, a novel NOTCH3 methylation stimulator, significantly suppresses lung carcinogenesis in vitro and in vivo. Front Pharmacol. 2018;9:434.PubMedPubMedCentralCrossRef Su T, Yang X, Deng JH, Huang QJ, Huang SC, Zhang YM, et al. Evodiamine, a novel NOTCH3 methylation stimulator, significantly suppresses lung carcinogenesis in vitro and in vivo. Front Pharmacol. 2018;9:434.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Guo Q, Liu Y, Zhao J, Wang J, Li Y, Pang Y, et al. Evodiamine inactivates NF-kappaB and potentiates the antitumor effects of gemcitabine on tongue cancer both in vitro and in vivo. Onco Targets Ther. 2019;12:257–67.PubMedCrossRef Guo Q, Liu Y, Zhao J, Wang J, Li Y, Pang Y, et al. Evodiamine inactivates NF-kappaB and potentiates the antitumor effects of gemcitabine on tongue cancer both in vitro and in vivo. Onco Targets Ther. 2019;12:257–67.PubMedCrossRef
48.
Zurück zum Zitat Sui H, Zhou LH, Zhang YL, Huang JP, Liu X, Ji Q, et al. Evodiamine suppresses ABCG2 mediated drug resistance by inhibiting p50/p65 NF-kappaB pathway in colorectal cancer. J Cell Biochem. 2016;117(6):1471–81.PubMedCrossRef Sui H, Zhou LH, Zhang YL, Huang JP, Liu X, Ji Q, et al. Evodiamine suppresses ABCG2 mediated drug resistance by inhibiting p50/p65 NF-kappaB pathway in colorectal cancer. J Cell Biochem. 2016;117(6):1471–81.PubMedCrossRef
49.
Zurück zum Zitat Remy-Ziller C, Thioudellet C, Hortelano J, Gantzer M, Nourtier V, Claudepierre MC, et al. Sequential administration of MVA-based vaccines and PD-1/PD-L1-blocking antibodies confers measurable benefits on tumor growth and survival: preclinical studies with MVA-betaGal and MVA-MUC1 (TG4010) in a murine tumor model. Hum Vaccin Immunother. 2018;14(1):140–5.PubMedCrossRef Remy-Ziller C, Thioudellet C, Hortelano J, Gantzer M, Nourtier V, Claudepierre MC, et al. Sequential administration of MVA-based vaccines and PD-1/PD-L1-blocking antibodies confers measurable benefits on tumor growth and survival: preclinical studies with MVA-betaGal and MVA-MUC1 (TG4010) in a murine tumor model. Hum Vaccin Immunother. 2018;14(1):140–5.PubMedCrossRef
50.
Zurück zum Zitat Lin Y, Chen S, Zhong S, An H, Yin H, McGowan E. Phase I clinical trial of PD-1 knockout anti-MUC1 CAR-T cells in the treatment of patients with non-small cell lung cancer. Ann Oncol. 2019;30:xi12. Lin Y, Chen S, Zhong S, An H, Yin H, McGowan E. Phase I clinical trial of PD-1 knockout anti-MUC1 CAR-T cells in the treatment of patients with non-small cell lung cancer. Ann Oncol. 2019;30:xi12.
51.
Zurück zum Zitat Quoix E, Lena H, Losonczy G, Forget F, Chouaid C, Papai Z, et al. TG4010 immunotherapy and first-line chemotherapy for advanced non-small-cell lung cancer (TIME): results from the phase 2b part of a randomised, double-blind, placebo-controlled, phase 2b/3 trial. Lancet Oncol. 2016;17(2):212–23.PubMedCrossRef Quoix E, Lena H, Losonczy G, Forget F, Chouaid C, Papai Z, et al. TG4010 immunotherapy and first-line chemotherapy for advanced non-small-cell lung cancer (TIME): results from the phase 2b part of a randomised, double-blind, placebo-controlled, phase 2b/3 trial. Lancet Oncol. 2016;17(2):212–23.PubMedCrossRef
52.
53.
Zurück zum Zitat Posey AD Jr, Schwab RD, Boesteanu AC, Steentoft C, Mandel U, Engels B, et al. Engineered CAR T cells targeting the cancer-associated Tn-Glycoform of the membrane Mucin MUC1 control adenocarcinoma. Immunity. 2016;44(6):1444–54.PubMedPubMedCentralCrossRef Posey AD Jr, Schwab RD, Boesteanu AC, Steentoft C, Mandel U, Engels B, et al. Engineered CAR T cells targeting the cancer-associated Tn-Glycoform of the membrane Mucin MUC1 control adenocarcinoma. Immunity. 2016;44(6):1444–54.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Liu C, Lu Z, Xie Y, Guo Q, Geng F, Sun B, et al. Soluble PD-1-based vaccine targeting MUC1 VNTR and survivin improves anti-tumor effect. Immunol Lett. 2018;200:33–42.PubMedCrossRef Liu C, Lu Z, Xie Y, Guo Q, Geng F, Sun B, et al. Soluble PD-1-based vaccine targeting MUC1 VNTR and survivin improves anti-tumor effect. Immunol Lett. 2018;200:33–42.PubMedCrossRef
55.
Zurück zum Zitat Yasumizu Y, Rajabi H, Jin C, Hata T, Pitroda S, Long MD, et al. MUC1-C regulates lineage plasticity driving progression to neuroendocrine prostate cancer. Nat Commun. 2020;11(1):338.PubMedPubMedCentralCrossRef Yasumizu Y, Rajabi H, Jin C, Hata T, Pitroda S, Long MD, et al. MUC1-C regulates lineage plasticity driving progression to neuroendocrine prostate cancer. Nat Commun. 2020;11(1):338.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Yamamoto M, Jin C, Hata T, Yasumizu Y, Zhang Y, Hong D, et al. MUC1-C integrates chromatin remodeling and PARP1 activity in the DNA damage response of triple-negative breast cancer cells. Cancer Res. 2019;79(8):2031–41.PubMedPubMedCentralCrossRef Yamamoto M, Jin C, Hata T, Yasumizu Y, Zhang Y, Hong D, et al. MUC1-C integrates chromatin remodeling and PARP1 activity in the DNA damage response of triple-negative breast cancer cells. Cancer Res. 2019;79(8):2031–41.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Pyzer AR, Stroopinsky D, Rosenblatt J, Anastasiadou E, Rajabi H, Washington A, et al. MUC1 inhibition leads to decrease in PD-L1 levels via upregulation of miRNAs. Leukemia. 2017. Pyzer AR, Stroopinsky D, Rosenblatt J, Anastasiadou E, Rajabi H, Washington A, et al. MUC1 inhibition leads to decrease in PD-L1 levels via upregulation of miRNAs. Leukemia. 2017.
58.
Zurück zum Zitat Lee JW, Zhang Y, Eoh KJ, Sharma R, Sanmamed MF, Wu J, et al. The combination of MEK inhibitor with Immunomodulatory antibodies targeting programmed death 1 and programmed death ligand 1 results in prolonged survival in Kras/p53-driven lung cancer. J Thorac Oncol. 2019;14(6):1046–60.PubMedPubMedCentralCrossRef Lee JW, Zhang Y, Eoh KJ, Sharma R, Sanmamed MF, Wu J, et al. The combination of MEK inhibitor with Immunomodulatory antibodies targeting programmed death 1 and programmed death ligand 1 results in prolonged survival in Kras/p53-driven lung cancer. J Thorac Oncol. 2019;14(6):1046–60.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Medina-Echeverz J, Hinterberger M, Testori M, Geiger M, Giessel R, Bathke B, et al. Synergistic cancer immunotherapy combines MVA-CD40L induced innate and adaptive immunity with tumor targeting antibodies. Nat Commun. 2019;10(1):5041.PubMedPubMedCentralCrossRef Medina-Echeverz J, Hinterberger M, Testori M, Geiger M, Giessel R, Bathke B, et al. Synergistic cancer immunotherapy combines MVA-CD40L induced innate and adaptive immunity with tumor targeting antibodies. Nat Commun. 2019;10(1):5041.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Ma HS, Poudel B, Torres ER, Sidhom JW, Robinson TM, Christmas B, et al. A CD40 agonist and PD-1 antagonist antibody reprogram the microenvironment of nonimmunogenic tumors to allow T-cell-mediated anticancer activity. Cancer Immunol Res. 2019;7(3):428–42.PubMedPubMedCentralCrossRef Ma HS, Poudel B, Torres ER, Sidhom JW, Robinson TM, Christmas B, et al. A CD40 agonist and PD-1 antagonist antibody reprogram the microenvironment of nonimmunogenic tumors to allow T-cell-mediated anticancer activity. Cancer Immunol Res. 2019;7(3):428–42.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Galvani E, Mundra PA, Valpione S, Garcia-Martinez P, Smith M, Greenall J, et al. Stroma remodeling and reduced cell division define durable response to PD-1 blockade in melanoma. Nat Commun. 2020;11(1):853.PubMedPubMedCentralCrossRef Galvani E, Mundra PA, Valpione S, Garcia-Martinez P, Smith M, Greenall J, et al. Stroma remodeling and reduced cell division define durable response to PD-1 blockade in melanoma. Nat Commun. 2020;11(1):853.PubMedPubMedCentralCrossRef
Metadaten
Titel
Evodiamine suppresses non-small cell lung cancer by elevating CD8+ T cells and downregulating the MUC1-C/PD-L1 axis
verfasst von
Ze-Bo Jiang
Ju-Min Huang
Ya-Jia Xie
Yi- Zhong Zhang
Chan Chang
Huan-Ling Lai
Wenjun Wang
Xiao-Jun Yao
Xing-Xing Fan
Qi-Biao Wu
Chun Xie
Mei-Fang Wang
Elaine Lai-Han Leung
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2020
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01741-5

Weitere Artikel der Ausgabe 1/2020

Journal of Experimental & Clinical Cancer Research 1/2020 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

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