|
|
Mechanism of action of triple therapy with radiotherapy and Gemcitabine combined with immune checkpoint inhibitors on the microenvironment of triple-negative breast cancer |
WU Miaomiao1 FANG Kun2 |
1.Tumor Epigenetics Lab, West Branch of the First Affiliated Hospital of University of Science and Technology of China Anhui Provincial Cancer Hospital West Branch of Anhui Provincial Hospital, Anhui Province, Hefei 230031, China; 2.Department of Biomedical Engineering, the First Affiliated Hospital of University of Science and Technology of China Anhui Provincial Hospital, Anhui Province, Hefei 230001, China |
|
|
Abstract Objective To investigate the effect of triple therapy of radiotherapy, Gemcitabine (Gem) combined with immune checkpoint inhibitors on the microenvironment of triple-negative breast cancer, and to reveal its possible mechanism. Methods A total of 29 SPF BALB/c mice aged five weeks and weighing 18-20 g were selected. On the first day of the experiment, about 1×106 4T1 cells were injected subcutaneously into the left abdomen to establish triple-negative breast cancer transplant tumor model. When the tumor volume increased to 100 mm3, the mice were divided into control group (five mice), anti-programmed death receptor ligand 1 (aPD-L1) group (six mice), radiotherapy (RT) +aPD-L1 group (six mice), Gem +aPD-L1 group (six mice), and triple group (six mice) by random number method. In addition to the control group, the other four groups were injected with drugs (Gem 5 mg/kg, aPD-L1 3 mg/kg) in the tumor at the 6th, 8th, and 10th days of the experiment. Twenty-four hours after each injection, RT (2 Gy) was administered to the RT+aPD-L1 group and the triple group. On the 30th day of the experiment, the animals were killed, the tumor weight of each group was weighed, and the main organs were stained with hematoxylin-eosin. The absolute percentage of CD3+CD8+ T cells in peripheral blood and tumor was detected by flow cytometry. The expression of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in peripheral blood and tumor were detected by enzyme-linked immunosorbent assay. The expressions of Ki-67, microvessel density(MVD)-CD31, and γH2AX were detected by immunohistochemistry, and apoptosis was evaluated by TUNEL detection. The expression of vascular endothelial growth factor (VEGF) and hypoxia inducible factor-1 α (HIF-1α) were detected by Western blot analysis. Results There was no significant difference in body weight among all groups (P>0.05). At the end of the experiment, the tumor weight of RT+aPD-L1 group and triple group were lower than those of aPD-L1 group, and those of triple group were lower than those of RT+aPD-L1 group (P<0.05). Hematoxylin-eosin staining showed no obvious damage to the heart, liver, spleen, and kidney in each group. The absolute percentage of CD3+CD8+ T cells , TNF-α, and IL-6 in peripheral blood of aPD-L1 group were higher than those of control group. Peripheral blood IL-6 in RT+aPD-L1 group was higher than that in aPD-L1 group, and absolute percentage of CD3+CD8+ T cells , IFN-γ, and IL-6 of peripheral blood in triple group were higher than those in aPD-L1 group. The absolute percentage of CD3+CD8+ T cells of peripheral blood in triple group was higher than that of RT+aPD-L1 group and Gem+aPD-L1 group, and IL-6 was higher than that of Gem+aPD-L1 group (P<0.05). The absolute percentage of CD3+CD8+ T cells , IFN-γ, TNF-α, and IL-6 of tumor in aPD-L1 group were higher than those in control group. The absolute percentages of CD3+CD8+ T cells of tumor in RT+aPD-L1 group, Gem+aPD-L1 group, and triple group were higher than those in aPD-L1 group, and TNF-α and IL-6 of tumor in triple group were higher than those in aPD-L1 group. The absolute percentage of CD3+CD8+ T cells and IL-6 of tumor in triple group were higher than those in RT+aPD-L1 group and Gem+aPD-L1 group, and IFN-γ was higher than those in RT+aPD-L1 group (P<0.05). Immunohistochemical staining and TUNEL detection showed that the expression of Ki-67 decreased and the apoptotic cells increased in the tumor tissues of triple group. MVD-CD31 in triple group was lower than that in aPD-L1 group, γH2AX was higher than that in aPD-L1 group and Gem+aPD-L1 group (P<0.05). VEGF in aPD-L1 group was lower than that in control group, VEGF in RT+aPD-L1 group, Gem+aPD-L1 group, and triple group was lower than that in aPD-L1 group, and HIF-1α in triple group was lower than that in aPD-L1 group and RT+aPD-L1 group (P<0.05). Conclusion The triple therapy of radiotherapy, Gem combined with immune checkpoint inhibitors can delay tumor growth, increase the absolute percentage of CD3+CD8+ T cells in peripheral blood and tumor, promote the release of inflammatory factors in serum and tumor, improve intratumor hypoxia and inhibit tumor microvascular growth in mouse triple-negative breast cancer transplant tumor model.
|
|
|
|
|
[1] Derakhshan F,Reis-Filho JS. Pathogenesis of Triple-Negative Breast Cancer [J]. Annu Rev Pathol,2022,17:181-204. [2] Li Y,Zhang H,Merkher Y,et al. Recent advances in therapeutic strategies for triple-negative breast cancer [J]. J Hematol Oncol,2022,15(1):121. [3] Ai L,Xu A,Xu J. Roles of PD-1/PD-L1 Pathway:Signaling,Cancer,and Beyond [J]. Adv Exp Med Biol,2020,1248:33-59. [4] Tang Q,Chen Y,Li X,et al. The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers [J]. Front Immunol,2022,13:964442. [5] Zhang Y,Yang Y,Chen Y,et al. PD-L1:Biological mechanism,function,and immunotherapy in gastric cancer [J]. Front Immunol,2022,13:1060497. [6] Lin M,Cai Y,Chen G,et al. A hierarchical tumor-targeting strategy for eliciting potent antitumor immunity against triple negative breast cancer [J]. Biomaterials,2023,296:122067. [7] Chen X,Feng L,Huang Y,et al. Mechanisms and Strategies to Overcome PD-1/PD-L1 Blockade Resistance in Triple- Negative Breast Cancer [J]. Cancers(Basel),2022,15(1):104. [8] Boustani J,Lecoester B,Baude J,et al. Anti-PD-1/Anti-PD- L1 Drugs and Radiation Therapy:Combinations and Optimization Strategies [J]. Cancers(Basel),2021,13(19):4893. [9] Chen C,Li A,Sun P,et al. Efficiently restoring the tumoricidal immunity against resistant malignancies via an immune nanomodulator [J]. J Control Release,2020,324:574-585. [10] Del Re M,Vivaldi C,Rofi E,et al. Gemcitabine Plus Nab- Paclitaxel Induces PD-L1 mRNA Expression in Plasma-Derived Microvesicles in Pancreatic Cancer [J]. Cancers(Basel),2021,13(15):3738. [11] Zhang Q,Han Z,Zhu Y,et al. Role of hypoxia inducible factor-1 in cancer stem cells(Review)[J]. Mol Med Rep,2021,23(1):17. [12] Zhao Q,Zhang L,He Q,et al. Targeting TRMT5 suppresses hepatocellular carcinoma progression via inhibiting the HIF-1α pathways [J]. J Zhejiang Univ Sci B,2023,24(1):50-63. [13] Pulaski BA,Ostrand-Rosenberg S. Reduction of Established Spontaneous Mammary Carcinoma Metastases following Immunotherapy with Major Histocompatibility Complex Class Ⅱ and B7.1 Cell-based Tumor Vaccines [J]. Cancer Res,1998,58(7):1486-1493. [14] Leun AM,Thommen DS,Schumacher TN. CD8+ T cell states in human cancer:insights from single-cell analysis [J]. Nat Rev Cancer,2020,20(4):218-232. [15] Lalos A,Tülek A,Tosti N,et al. Prognostic significance of CD8+ T-cells density in stage Ⅲ colorectal cancer depends on SDF-1 expression [J]. Sci Rep,2021,11(1):775. [16] VirassamyB,Caramia F,Savas P,et al. Intratumoral CD8+ T cells with a tissue-resident memory phenotype mediate local immunity and immune checkpoint responses in breast cancer [J]. Cancer Cell,2023,41(3):585-601.e8. [17] Wang X,Wu WKK,Gao J,et al. Autophagy inhibition enhances PD-L1 expression in gastric cancer [J]. J Exp Clin Cancer Res,2019,38(1):140. [18] Esparza Guerrero Y,Vazquez Villegas ML,Nava Valdivia CA,et al. Association of the STAT4 Gene rs7574865 Polymorphism with IFN-γ Levels in Patients with Systemic Lupus Erythematosus [J]. Genes(Basel),2023,14(3):537. [19] Dixit A,Sarver A,Zettervall J,et al. Targeting TNF-α- producing macrophages activates antitumor immunity in pancreatic cancer via IL-33 signaling [J]. JCI Insight,2022, 7(22):e153242. [20] Ra?觢ková M,Lacina L,Kejík Z,et al. The Role of IL-6 in Cancer Cell Invasiveness and Metastasis-Overview and Therapeutic Opportunities [J]. Cells,2022,11(22):3698. [21] Srivastava N,Usmani SS,Subbarayan R,et al. Hypoxia:syndicating triple negative breast cancer against various therapeutic regimens [J]. Front Oncol,2023,13:1199105. [22] Luo S,Jiang Y,Anfu Z,et al. Targeting hypoxia-inducible factors for breast cancer therapy:A narrative review [J]. Front Pharmacol,2022,13:1064661. [23] Liu Q,Guan C,Liu C,et al. Targeting hypoxia-inducible factor-1alpha:A new strategy for triple-negative breast can- cer therapy [J]. Biomed Pharmacother,2022,156:113861. [24] Lopes-Coelho F,Martins F,Pereira SA,et al. Anti-Angiogenic Therapy:Current Challenges and Future Perspectives [J]. Int J Mol Sci,2021,22(7):3765. [25] Bu MT,Chandrasekhar P,Ding L,et al. The roles of TGF- β and VEGF pathways in the suppression of antitumor immunity in melanoma and other solid tumors [J]. Pharmacol Ther,2022,240:108211. |
|
|
|