scholarly journals Green Tea Polyphenol EGCG Attenuates MDSCs-mediated Immunosuppression through Canonical and Non-Canonical Pathways in a 4T1 Murine Breast Cancer Model

Nutrients ◽  
2020 ◽  
Vol 12 (4) ◽  
pp. 1042 ◽  
Author(s):  
Ping Xu ◽  
Feng Yan ◽  
Yueling Zhao ◽  
Xiangbo Chen ◽  
Shili Sun ◽  
...  

Several studies in the past decades have reported anti-tumor activity of the bioactive compounds extracted from tea leaves, with a focus on the compound epigallocatechin-3-gallate (EGCG). However, further investigations are required to unravel the underlying mechanisms behind the anti-tumor activity of EGCG. In this study, we demonstrate that EGCG significantly inhibits the growth of 4T1 breast cancer cells in vitro and in vivo. EGCG ameliorated immunosuppression by significantly decreasing the accumulation of myeloid-derived suppressor cells (MDSCs) and increasing the proportions of CD4+ and CD8+ T cells in spleen and tumor sites in 4T1 breast tumor-bearing mice. Surprisingly, a low dose of EGCG (0.5–5 μg/mL) effectively reduced the cell viability and increased the apoptosis rate of MDSCs in vitro. EGCG down-regulated the canonical pathways in MDSCs, mainly through the Arg-1/iNOS/Nox2/NF-κB/STAT3 signaling pathway. Moreover, transcriptomic analysis suggested that EGCG also affected the non-canonical pathways in MDSCs, such as ECM–receptor interaction and focal adhesion. qRT-PCR further validated that EGCG restored nine key genes in MDSCs, including Cxcl3, Vcan, Col4a1, Col8a1, Oasl2, Mmp12, Met, Itsnl and Acot1. Our results provide new insight into the mechanism of EGCG-associated key pathways/genes in MDSCs in the murine breast tumor model.

2021 ◽  
Vol 12 ◽  
Author(s):  
Jiafeng Gao ◽  
Yi-Nan Zhang ◽  
Jingwen Cui ◽  
Jiatong Zhang ◽  
Yuexiang Ming ◽  
...  

Dendritic cells (DCs) are the most potent professional antigen-presenting cells (APCs) that mediate T-cell immune responses. Breast cancer is one of the most commonly diagnosed diseases and its mortality rate is higher than any other cancer in both humans and canines. Plantain polysaccharide (PLP), extracted from the whole plant of Plantago asiatica L., could promote the maturation of DCs. In this research, we found that PLP could upregulate the maturation of DCs both in vitro and in vivo. PLP-activated DCs could stimulate lymphocytes’ proliferation and differentiate naive T cells into cytotoxic T cells. Tumor antigen-specific lymphocyte responses were enhanced by PLP and CIPp canine breast tumor cells lysate-pulsed DCs, and PLP and CIPp-cell-lysate jointly stimulated DCs cocultured with lymphocytes having the great cytotoxicity on CIPp cells. In the 4T1 murine breast tumor model, PLP could control the size of breast tumors and improve immunity by recruiting DCs, macrophages, and CD4+ and CD8+ T cells in the tumor microenvironment. These results indicated that PLP could achieve immunotherapeutic effects and improve immunity in the breast tumor model.


Oncotarget ◽  
2017 ◽  
Vol 9 (4) ◽  
pp. 4593-4606 ◽  
Author(s):  
Ka-Wai Tam ◽  
Chi-Tang Ho ◽  
Shih-Hsin Tu ◽  
Wen-Jui Lee ◽  
Ching-Shui Huang ◽  
...  

Oncotarget ◽  
2017 ◽  
Vol 8 (17) ◽  
pp. 28840-28853 ◽  
Author(s):  
Mohadeseh Hasanpourghadi ◽  
Ashok Kumar Pandurangan ◽  
Chandrabose Karthikeyan ◽  
Piyush Trivedi ◽  
Mohd Rais Mustafa

2019 ◽  
Vol 18 ◽  
pp. 153473541984804 ◽  
Author(s):  
Paola Lasso ◽  
Mónica Llano Murcia ◽  
Tito Alejandro Sandoval ◽  
Claudia Urueña ◽  
Alfonso Barreto ◽  
...  

Background: The tumor cells responsible for metastasis are highly resistant to chemotherapy and have characteristics of stem cells, with a high capacity for self-regeneration and the use of detoxifying mechanisms that participate in drug resistance. In vivo models of highly resistant cells allow us to evaluate the real impact of the immune response in the control of cancer. Materials and Methods: A tumor population derived from the 4T1 breast cancer cell line that was stable in vitro and highly aggressive in vivo was obtained, characterized, and determined to exhibit cancer stem cell (CSC) phenotypes (CD44+, CD24+, ALDH+, Oct4+, Nanog+, Sox2+, and high self-renewal capacity). Orthotopic transplantation of these cells allowed us to evaluate their in vivo susceptibility to chemo and immune responses induced after vaccination. Results: The immune response induced after vaccination with tumor cells treated with doxorubicin decreased the formation of tumors and macrometastasis in this model, which allowed us to confirm the immune response relevance in the control of highly chemotherapy-resistant ALDH+ CSCs in an aggressive tumor model in immunocompetent animals. Conclusions: The antitumor immune response was the main element capable of controlling tumor progression as well as metastasis in a highly chemotherapy-resistant aggressive breast cancer model.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 10101-10101
Author(s):  
J. Hartman ◽  
K. Lindberg ◽  
J. Inzunza ◽  
J. Wan ◽  
A. Ström ◽  
...  

10101 Background: Estrogens are well known stimulators of breast cancer cell growth in vitro as well as in vivo. Two different estrogen receptors exist, namely estrogen receptor (ER) α and β. ERα mediates the proliferative effect of estrogen in breast cancer cells and we have earlier shown that ERβ inhibits cell-cycle progression in vitro. Estrogens are well known stimulators of in vivo breast cancer cell growth as well as angiogenesis, and the effect is mediated through ERα. The function of ERβ in this context is not well understood. Methods: We have used ERα-positive T47D breast cancer cells stably transfected with a Tet/Off regulated ERβ expression vector system. The ERβ-inducible tumor cells are studied in vitro as well as in vivo. Results: By transplanting ERβ-inducible breast cancer cells into SCID-mice, we show that ERβ inhibits tumor growth and reduces the volume of established tumors. Furthermore, we show by immunohistochemistry, that the number of blood microvessels in the tumor periphery is decreased by ERβ expression, counteracting the well-known pro-angiogenic effect of ERα. By Western blot analysis on tumor extracts, we show that the concentration of the important pro-angiogenic growth factors VEGF and bFGF, normally expressed by breast tumor cells, is decreased in the ERβ-expressing tumors compared to the normal tumors. To exclude that the observed anti-angiogenic effect is just a result of reduced tumor growth, we incubated Tet/Off regulated ERβ expressing cells in vitro, during non-hypoxic conditions. We found that the expression of ERβ leads to decreased expression of VEGF and PDGFβ at the mRNA and protein-levels. In transient transfection assays, we found estrogen-ERα mediated up regulation of VEGF, PDGFβ and bFGF-promoter activities in T47D cells, and these activities were all suppressed following co-transfection with an ERβ-expression vector. Conclusions: We conclude that ERβ inhibits growth factor expression at transcriptional level in breast cancer cells; taken together, our data indicates that ERβ inhibits growth and angiogenesis of tumors formed by T47D breast cancer cells. This makes ERβ an interesting therapeutic target in breast cancer and perhaps treatment with the newly designed ERβ-selective ligands might work as a new anti-proliferative and anti-angiogenic therapy. No significant financial relationships to disclose.


2014 ◽  
Vol 349 (2) ◽  
pp. 120-127 ◽  
Author(s):  
Lucia Borriello ◽  
Matthieu Montès ◽  
Yves Lepelletier ◽  
Bertrand Leforban ◽  
Wang-Qing Liu ◽  
...  

2021 ◽  
Vol 12 ◽  
Author(s):  
Muhammad Tayyab Imtiaz ◽  
Fareeha Anwar ◽  
Uzma Saleem ◽  
Bashir Ahmad ◽  
Sundas Hira ◽  
...  

Background and objectives: Breast cancer is a heterogeneous disease that poses the highest incidence of morbidity among women and presents many treatment challenges. In search of novel breast cancer therapies, several triazine derivatives have been developed for their potential chemotherapeutic activity. This study aims to evaluate the N-nitroso-N-methyl urea (NMU)–induced anti–mammary gland tumor activity of 2,4,6 (O-nitrophenyl amino) 1,3,5-triazine (O-NPAT).Methods: The in silico modeling and in vitro cytotoxicity assay were performed to strengthen the research hypothesis. For in vivo experimentation, 30 female rats were divided into five groups. Group I (normal control) received normal saline. Group II (disease control) received NMU (50 mg/kg). Group III (standard control) was treated with tamoxifen (5 mg/kg). Groups IV and V received O-NPAT at a dose level of 30 and 60 mg/kg, respectively. For tumor induction, 3 intraperitoneal doses of NMU were given at a 3-week interval, whereas all treatment compounds were administered orally for 14 consecutive days. Biochemical and oxidative stress markers were estimated for all experimental animals. DNA strand breakage alongside inflammatory markers was also measured for the analysis of inflammation. The hormonal profile of progesterone and estrogen was also estimated.Results: The test compound presented a significant reduction in organ weight and restored the hepatic and renal enzymes. O-NPAT treatments enhanced the antioxidant enzyme level of catalase (CAT), superoxide dismutase (SOD), and total sulfhydryl (TSH), with a highly significant reduction in lactate dehydrogenase (LDH) and lipid peroxidation. Also, the decrease in fragmented DNA, hormonal levels (estradiol and progesterone), and inflammatory cytokines (IL-6 and TNF-α) justified the dosage efficacy further supported by histopathological findings.Conclusion: All results indicated the anti–breast tumor activity of O-NPAT and presented its possibility of exploitation for beneficial effects in breast cancer treatment.


2005 ◽  
Vol 94 (1-3) ◽  
pp. 111-121 ◽  
Author(s):  
Sébastien Maillard ◽  
Thibault Ameller ◽  
Juliette Gauduchon ◽  
Angélique Gougelet ◽  
Fabrice Gouilleux ◽  
...  

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 11-12
Author(s):  
Doris Mangelberger ◽  
Christian Augsberger ◽  
Karin Landgraf ◽  
Christina Heitmüller ◽  
Stefan Steidl

Introduction Tafasitamab (MOR208) is an Fc-enhanced, humanized, monoclonal antibody that targets CD19 and has shown promising clinical activity in patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL). CD19 is homogeneously expressed among different B-cell malignancies, and the binding of tafasitamab to CD19 directly mediates cell death, induces antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis. Aiming to potentiate the tafasitamab-mediated "eat me" signal, we tested a combination with a CD47-directed monoclonal antibody (mAb) to inhibit the CD47/SIRPα "don't eat me" signal and further enhance macrophage-mediated phagocytosis. Preclinical studies demonstrated that blocking the CD47/SIRPα checkpoint in combination with antibodies, such as rituximab, increased phagocytosis by macrophages, resulting in effective anti-tumor effects in non-Hodgkin lymphoma (NHL) (Chao, et al. 2010). Additionally, the combination of the anti-CD47, magrolimab, and the anti-CD20, rituximab, demonstrated beneficial outcomes for patients with refractory NHL (Advani, et al. 2019). Here, we present in vitro and in vivo data on the combinatory effect of tafasitamab and an anti-CD47 mAb in preclinical models of Burkitt's lymphoma (BL). Methods During in vitro studies, CD14+ monocytes were isolated from the whole blood of healthy volunteers and differentiated with 50 ng/mL M-CSF for 6 days. ADCP was analyzed by flow cytometry in co-culture experiments with Ramos cells (BL) after 3 hours of treatment with tafasitamab and anti-CD47 mAb (clone B6H12). In vivo, the combination of tafasitamab with an anti-CD47 mAb was tested in a Ramos disseminated survival and subcutaneous tumor model in SCID and NOD-SCID mice, respectively. In both models, tafasitamab was administered therapeutically twice a week either at 3 mg/kg (disseminated) or 10 mg/kg (subcutaneous) for max. 4 weeks. The anti-CD47 mAb was administered at 4 mg/kg three times per week. Main study readouts were to assess animal survival and any delays in tumor growth. Results The combination of tafasitamab + CD47/SIRPα checkpoint blockade enhanced ADCP activity of primary M2 macrophages on BL-derived Ramos cells, in comparison with the anti-CD47 mAb or tafasitamab monotherapies (Figure 1A). In vivo, a significant increase in anti-tumor activity was observed with the combination of tafasitamab + anti-CD47 mAb. In the Ramos disseminated survival model, the combination showed an increased life span (ILS) of >182% compared with tafasitamab monotherapy control, with an overall survival of all animals treated with the combination (15/15) until the end of the study (Day 99 post-cell injection). Additionally, pronounced anti-tumor efficacies were detected in the Ramos subcutaneous tumor model. Here, the combination resulted in a significant delay in tumor growth compared with the tafasitamab or anti-CD47 mAb monotherapies (ILS >175% tafasitamab and ILS >72% anti-CD47 mAb vs tafasitamab + B6H12) (Figure 1B). Conclusions The ADCP activity of primary macrophages was increased by combining tafasitamab with an anti-CD47 mAb in vitro, resulting in enhanced anti-tumor activity compared with tafasitamab or anti-CD47 mAb monotherapies in vivo. Overall, results indicate the combination of tafasitamab with a CD47/SIRPα checkpoint blockade may be a promising novel combination approach for lymphoma therapy. Disclosures Mangelberger: MorphoSys AG: Current Employment. Augsberger:MorphoSys AG: Current Employment. Landgraf:MorphoSys AG: Current Employment. Heitmüller:MorphoSys AG: Current Employment. Steidl:MorphoSys AG: Current Employment.


Sign in / Sign up

Export Citation Format

Share Document