1.Risk factors for positive post-transplantation measurable residual disease in patients with acute lymphoblastic leukemia.
Yuewen WANG ; Guomei FU ; Lanping XU ; Yu WANG ; Yifei CHENG ; Yuanyuan ZHANG ; Xiaohui ZHANG ; Yanrong LIU ; Kaiyan LIU ; Xiaojun HUANG ; Yingjun CHANG
Chinese Medical Journal 2025;138(9):1084-1093
BACKGROUND:
The level of measurable residual disease (MRD) before and after transplantation is related to inferior transplant outcomes, and post-hematopoietic stem cell transplantation measurable residual disease (post-HSCT MRD) has higher prognostic value in determining risk than pre-hematopoietic stem cell transplantation measurable residual disease (pre-HSCT MRD). However, only a few work has been devoted to the risk factors for positive post-HSCT MRD in patients with acute lymphoblastic leukemia (ALL). This study evaluated the risk factors for post-HSCT MRD positivity in patients with ALL who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT).
METHODS:
A total of 1683 ALL patients from Peking University People's Hospital between January 2009 and December 2019 were enrolled to evaluate the cumulative incidence of post-HSCT MRD. Cox proportional hazard regression models were built for time-to-event outcomes. Multivariable analysis was performed to determine independent influencing factors from the univariable analysis.
RESULTS:
Both in total patients and in T-cell ALL or B-cell ALL, pediatric or adult, human leukocyte antigen-matched sibling donor transplantation or haploidentical SCT subgroups, positive pre-HSCT MRD was a risk factor for post-HSCT MRD positivity ( P <0.001 for all). Disease status (complete remission 1 [CR1] vs . ≥CR2) was also a risk factor for post-HSCT MRD positivity in all patients and in the B cell-ALL, pediatric, or haploidentical SCT subgroups ( P = 0.027; P = 0.003; P = 0.035; P = 0.003, respectively). A risk score for post-HSCT MRD positivity was developed using the variables pre-HSCT MRD and disease status. The cumulative incidence of post-HSCT MRD positivity was 12.3%, 25.1%, and 38.8% for subjects with scores of 0, 1, and 2-3, respectively ( P <0.001). Multivariable analysis confirmed the association of the risk score with the cumulative incidence of post-HSCT MRD positivity and relapse as well as leukemia-free survival and overall survival.
CONCLUSION
Our results indicated that positive pre-MRD and disease status were two independent risk factors for post-HSCT MRD positivity in patients with ALL who underwent allo-HSCT.
Humans
;
Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology*
;
Neoplasm, Residual
;
Hematopoietic Stem Cell Transplantation/methods*
;
Male
;
Female
;
Risk Factors
;
Adolescent
;
Adult
;
Child
;
Child, Preschool
;
Young Adult
;
Middle Aged
;
Infant
;
Transplantation, Homologous
;
Proportional Hazards Models
;
Retrospective Studies
2.Patient-derived xenograft model: Applications and challenges in liver cancer.
Shuangshuang DOU ; Yunfei HUO ; Minghui GAO ; Quanwei LI ; Buxin KOU ; Mengyin CHAI ; Xiaoni LIU
Chinese Medical Journal 2025;138(11):1313-1323
Liver cancer is one of the most common malignant tumors worldwide. Currently, the available treatment methods cannot fully control its recurrence and mortality rate. Establishing appropriate animal models for liver cancer is crucial for developing new treatment technologies and strategies. The patient-derived xenograft (PDX) model preserves the tumor's microenvironment and heterogeneity, which makes it advantageous for biological research, drug evaluation, personalized medicine, and other purposes. This article reviews the development, preparation techniques, application fields, and challenges of PDX models in liver cancer, providing insights for the research and exploration of PDX models in diagnostic and therapeutic strategies of liver cancer.
Liver Neoplasms/drug therapy*
;
Animals
;
Humans
;
Xenograft Model Antitumor Assays/methods*
;
Mice
;
Disease Models, Animal
3.The effects of resveratrol on osteosarcoma cells: Regulation of the interaction between JAK2/STAT3 signaling pathway and tumor immune microenvironment.
Xiaoli WANG ; Guoliang MA ; Ruidong LIU ; Ruixia QI ; Jiudei QI ; Yuguo REN
Chinese Journal of Cellular and Molecular Immunology 2025;41(5):420-427
Objective To investigate the effect of resveratrol on the tumor microenvironment in osteosarcoma. Methods A C57BL/6 xenograft mouse model was established and treated with resveratrol. Single-cell sequencing was performed to analyze changes in the tumor microenvironment. Immunohistochemistry was used to assess immune cell infiltration, while Western blotting was conducted to examine alterations in cellular signaling pathways. Results Resveratrol significantly inhibited the proliferation of LM8 osteosarcoma cells in C57BL/6 mice compared to the control group. Additionally, CD8+ T cell recruitment was enhanced. The Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway was notably downregulated in LM8 osteosarcoma cells following resveratrol treatment. Conclusion Resveratrol promotes CD8+ T cell infiltration by inhibiting the JAK2/STAT3 signaling pathway, suggesting its potential as a therapeutic agent in osteosarcoma treatment.
Osteosarcoma/genetics*
;
STAT3 Transcription Factor/genetics*
;
Resveratrol/pharmacology*
;
Animals
;
Janus Kinase 2/genetics*
;
Signal Transduction/drug effects*
;
Tumor Microenvironment/immunology*
;
Cell Line, Tumor
;
Mice, Inbred C57BL
;
Mice
;
Humans
;
Cell Proliferation/drug effects*
;
Bone Neoplasms/metabolism*
;
CD8-Positive T-Lymphocytes/drug effects*
;
Xenograft Model Antitumor Assays
4.Prognostic significance of molecular minimal residual disease before and after allogeneic hematopoietic stem cell transplantation in children with acute myeloid leukemia.
Xiu-Wen XU ; Hao XIONG ; Jian-Xin LI ; Zhi CHEN ; Fang TAO ; Yu DU ; Zhuo WANG ; Li YANG ; Wen-Jie LU ; Ming SUN
Chinese Journal of Contemporary Pediatrics 2025;27(6):675-681
OBJECTIVES:
To investigate the prognostic value of molecular minimal residual disease (Mol-MRD) monitored before and after allogeneic hematopoietic stem cell transplantation (HSCT) in pediatric acute myeloid leukemia (AML).
METHODS:
Clinical data of 71 pediatric AML patients who underwent HSCT between August 2016 and December 2023 were analyzed. Mol-MRD levels were dynamically monitored in MRD-positive patients, and survival outcomes were evaluated.
RESULTS:
No significant difference in the 3-year overall survival (OS) rate was observed between patients with pre-HSCT Mol-MRD ≥0.01% and <0.01% (77.3% ± 8.9% vs 80.4% ± 7.9%, P=0.705). However, patients with pre-HSCT Mol-MRD <1.75% had a significantly higher 3-year OS rate than those with Mol-MRD ≥1.75% (86.6% ± 5.6% vs 44.4% ± 16.6%, P=0.020). The median Mol-MRD level in long-term survivors was significantly lower than in non-survivors [0.61% (range: 0.04%-51.58%)] vs 10.60% (range: 1.90%-19.75%), P=0.035]. Concurrent flow cytometry-based MRD positivity was significantly higher in non-survivors (80% vs 24%, P=0.039). There was no significant difference in the 3-year overall survival rate between patients with Mol-MRD ≥0.01% and those with <0.01% at 30 days post-HSCT (P=0.527). For children with Mol-MRD <0.22% at 30 days post-HSCT, the 3-year overall survival rate was 80.4% ± 5.9%, showing no significant difference compared to those with molecular negativity (87.0% ± 7.0%) (P=0.523).
CONCLUSIONS
Patients with pre-HSCT Mol-MRD <1.75% or post-HSCT Mol-MRD <0.22% may achieve long-term survival outcomes comparable to Mol-MRD-negative cases through HSCT and targeted interventions.
Humans
;
Hematopoietic Stem Cell Transplantation
;
Neoplasm, Residual
;
Leukemia, Myeloid, Acute/genetics*
;
Child
;
Male
;
Female
;
Child, Preschool
;
Prognosis
;
Adolescent
;
Infant
;
Transplantation, Homologous
5.Establishment of a Bortezomib-Resistant Multiple Myeloma Xenotransplantation Mouse Model by Transplanting Primary Cells from Patients.
Yan-Hua YUE ; Yi-Fang ZHOU ; Ying-Jie MIAO ; Yang CAO ; Fei WANG ; Yue LIU ; Feng LI ; Yang-Ling SHEN ; Yan-Ting GUO ; Yu-Hui HUANG ; Wei-Ying GU
Journal of Experimental Hematology 2025;33(1):133-141
OBJECTIVE:
To explore the construction method of a resistant multiple myeloma (MM) patient-derived xenotransplantation (PDX) model.
METHODS:
1.0×107 MM patient-derived mononuclear cells (MNCs), 2.0×106 MM.1S cells and 2.0×106 NCI-H929 cells were respectively subcutaneously inoculated into NOD.CB17-Prkdcscid Il2rgtm1/Bcgen (B-NDG) mice with a volume of 100 μl per mouse to establish mouse model. The morphologic, phenotypic, proliferative and genetic characteristics of PDX tumor were studied by hematoxylin-eosin staining, immunohistochemical staining (IHC), cell cycle analysis, flow cytometry and fluorescence in situ hybridization (FISH). The sensitivity of PDX tumor to bortezomib and anlotinib monotherapy or in combination was investigated through cell proliferation, apoptosis and in vitro and in vivo experiments. The effects of anlotinib therapy on tumor blood vessel and cell apoptosis were analyzed by IHC, TUNEL staining and confocal fluorescence microscope.
RESULTS:
MM PDX model was successfully established by subcutaneously inoculating primary MNCs. The morphologic features of tumor cells from MM PDX model were similar to those of mature plasma cells. MM PDX tumor cells positively expressed CD138 and CD38, which presented 1q21 amplification, deletion of Rb1 and IgH rearrangement, and had a lower proliferative activity than MM cell lines. in vitro, PDX, MM.1S and NCI-H929 cells were treated by bortezomib and anlotinib for 24 hours, respectively. Cell viability assay showed that the IC50 value of bortezomib were 5 716.486, 1.025 and 2.775 nmol/L, and IC50 value of anlotinib were 5 5107.337, 0.706 and 5.13 μmol/L, respectively. Anlotinib treatment increased the apoptosis of MM.1S cells (P < 0.01), but did not affect PDX tumor cells (P >0.05). in vivo, there was no significant difference in PDX tumor growth between bortezomib monotherapy group and control group (P >0.05), while both anlotinib monotherapy and anlotinib combined with bortezomib effectively inhibited PDX tumor growth (both P < 0.05). The vascular perfusion and vascular density of PDX tumor were decreased in anlotinib treatment group (both P < 0.01). The apoptotic cells in anlotinib treatment group were increased compared with those in control group (P < 0.05).
CONCLUSION
Bortezomib-resistant MM PDX model can be successfully established by subcutaneous inoculation of MNCs from MM patients in B-NDG mice. This PDX model, which retains the basic biological characteristics of MM cells, can be used to study the novel therapies.
Animals
;
Bortezomib
;
Humans
;
Multiple Myeloma/pathology*
;
Mice
;
Apoptosis
;
Drug Resistance, Neoplasm
;
Cell Line, Tumor
;
Xenograft Model Antitumor Assays
;
Mice, Inbred NOD
;
Disease Models, Animal
;
Cell Proliferation
;
Transplantation, Heterologous
6.Tanshinone II A Facilitates Chemosensitivity of Osteosarcoma Cells to Cisplatin via Activation of p38 MAPK Pathway.
Da-Ming XIE ; Zhi-Yun LI ; Bing-Kai REN ; Rui GONG ; Dong YANG ; Sheng HUANG
Chinese journal of integrative medicine 2025;31(4):326-335
OBJECTIVE:
To examine the mechanism of action of tanshinone II A (Tan II A) in promoting chemosensitization of osteosarcoma cells to cisplatin (DDP).
METHODS:
The effects of different concentrations of Tan II A (0-80 µ mol/L) and DDP (0-2 µ mol/L) on the proliferation of osteosarcoma cell lines (U2R, U2OS, 143B, and HOS) at different times were examined using the cell counting kit-8 and colony formation assays. Migration and invasion of U2R and U2OS cells were detected after 24 h treatment with 30 µ mol/L Tan II A, 0.5 µ mol/L DDP alone, and a combination of 10 µ mol/L Tan II A and 0.25 µ mol/L DDP using the transwell assay. After 48 h of treatment of U2R and U2OS cells with predetermined concentrations of each group of drugs, the cell cycle was analyzed using a cell cycle detection kit and flow cytometry. After 48 h treatment, apoptosis of U2R and U2OS cells was detected using annexin V-FITC apoptosis detection kit and flow cytometry. U2R cells were inoculated into the unilateral axilla of nude mice and then the mice were randomly divided into 4 groups of 6 nude mice each. The 4 groups were treated with equal volume of Tan II A (15 mg/kg), DDP (3 mg/kg), Tan II A (7.5 mg/kg) + DDP (1.5 mg/kg), and normal saline, respectively. The body weight of the nude mice was weighed, and the tumor volume and weight were measured. Cell-related gene and signaling pathway expression were detected by RNA sequencing and Kyoto Encyclopedia of Genes and Genomes pathway analysis. p38 MAPK signaling pathway proteins and apoptotic protein expressions were detected by Western blot.
RESULTS:
In vitro studies have shown that Tan II A, DDP and the combination of Tan II A and DDP inhibit the proliferation, migration and invasion of osteosarcoma cells. The inhibitory effect was more pronounced in the Tan II A and DDP combined treatment group (P<0.05 or P<0.01). Osteosarcoma cells underwent significantly cell-cycle arrest and cell apoptosis by Tan II A-DDP combination treatment (P<0.05 or P<0.01). In vivo studies demonstrated that the Tan II A-DD combination treatment group significantly inhibited tumor growth compared to the Tan II A and DDP single drug group (P<0.01). Additionally, we found that the combination of Tan II A and DDP treatment enhanced the p38 MAPK signaling pathway. Western blot assays showed higher p-p38, cleaved caspase-3, and Bax and lower caspase-3, and Bcl-2 expressions with the combination of Tan II A and DDP treatment compared to the single drug treatment (P<0.01).
CONCLUSION
Tan II A synergizes with DDP by activating the p38/MAPK pathway to upregulate cleaved caspase-3 and Bax pro-apoptotic gene expressions, and downregulate caspase-3 and Bcl-2 inhibitory apoptotic gene expressions, thereby enhancing the chemosensitivity of osteosarcoma cells to DDP.
Abietanes/therapeutic use*
;
Osteosarcoma/enzymology*
;
Cisplatin/therapeutic use*
;
Humans
;
Cell Line, Tumor
;
Animals
;
Apoptosis/drug effects*
;
Mice, Nude
;
Cell Proliferation/drug effects*
;
Cell Movement/drug effects*
;
p38 Mitogen-Activated Protein Kinases/metabolism*
;
MAP Kinase Signaling System/drug effects*
;
Bone Neoplasms/enzymology*
;
Cell Cycle/drug effects*
;
Xenograft Model Antitumor Assays
;
Mice
;
Drug Resistance, Neoplasm/drug effects*
;
Neoplasm Invasiveness
;
Mice, Inbred BALB C
7.Suppression of Hepatocellular Carcinoma through Apoptosis Induction by Total Alkaloids of Gelsemium elegans Benth.
Ming-Jing JIN ; Yan-Ping LI ; Huan-Si ZHOU ; Yu-Qian ZHAO ; Xiang-Pei ZHAO ; Mei YANG ; Mei-Jing QIN ; Chun-Hua LU
Chinese journal of integrative medicine 2025;31(9):792-801
OBJECTIVE:
To evaluate the anti-hepatocellular carcinoma (HCC) activity of total alkaloids from Gelsemium elegans Benth. (TAG) in vivo and in vitro and to elucidate their potential mechanisms of action through transcriptomic analysis.
METHODS:
TAG extraction was conducted, and the primary components were quantified using high-performance liquid chromatography (HPLC). The effects of TAG (100, 150, and 200 µg/mL) on various tumor cells, including SMMC-7721, HepG2, H22, CAL27, MCF7, HT29, and HCT116, were assessed. Effects of TAG on HCC proliferation and apoptosis were detected by colony formation assays and cell stainings. Caspase-3, Bcl-2, and Bax protein levels were detected by Western blotting. In vivo, a tumor xenograft model was developed using H22 cells. Totally 40 Kunming mice were randomly assigned to model, cyclophosphamide (20 mg/kg), TAG low-dose (TAG-L, 0.5 mg/kg), and TAG high-dose (TAG-H, 1 mg/kg) groups, with 10 mice in each group. Tumor volume, body weight, and tumor weight were recorded and compared during 14-day treatment. Immune organ index were calculated. Tissue changes were oberseved by hematoxylin and eosin staining and immunohistochemistry. Additionally, transcriptomic and metabolomic analyses, as well as quatitative real-time polymerase chain reaction (RT-qPCR), were performed to detect mRNA and metabolite expressions.
RESULTS:
HPLC successfully identified the components of TAG extraction. Live cell imaging and analysis, along with cell viability assays, demonstrated that TAG inhibited the proliferation of SMMC-7721, HepG2, H22, CAL27, MCF7, HT29, and HCT116 cells. Colony formation assays, Hoechst 33258 staining, Rhodamine 123 staining, and Western blotting revealed that TAG not only inhibited HCC proliferation but also promoted apoptosis (P<0.05). In vivo experiments showed that TAG inhibited the growth of solid tumors in HCC in mice (P<0.05). Transcriptomic analysis and RT-qPCR indicated that the inhibition of HCC by TAG was associated with the regulation of the key gene CXCL13.
CONCLUSION
TAG inhibits HCC both in vivo and in vitro, with its inhibitory effect linked to the regulation of the key gene CXCL13.
Animals
;
Apoptosis/drug effects*
;
Liver Neoplasms/genetics*
;
Carcinoma, Hepatocellular/genetics*
;
Humans
;
Alkaloids/therapeutic use*
;
Gelsemium/chemistry*
;
Cell Line, Tumor
;
Cell Proliferation/drug effects*
;
Mice
;
Xenograft Model Antitumor Assays
8.Shuangshu Decoction inhibits growth of gastric cancer cell xenografts by promoting cell ferroptosis via the P53/SLC7A11/GPX4 axis.
Xinyuan CHEN ; Chengting WU ; Ruidi LI ; Xueqin PAN ; Yaodan ZHANG ; Junyu TAO ; Caizhi LIN
Journal of Southern Medical University 2025;45(7):1363-1371
OBJECTIVES:
To explore the mechanism of Shuangshu Decoction (SSD) for inhibiting growth of gastric cancer xenografts in nude mice.
METHODS:
Network pharmacology analysis was conducted to identify the common targets of SSD and gastric cancer cell ferroptosis, and bioinformatics analysis and molecular docking were used to validate the core targets. In the cell experiment, AGS cells were treated with SSD-medicated serum, Fer-1 (a ferroptosis inhibitor), or both, and the changes in cell viability, ferroptosis markers (ROS, Fe2+ and GSH), expressions of P53, SLC7A11 and GPX4, and mitochondrial morphology were examined. In a nude mouse model bearing gastric cancer xenografts, the effects of gavage with SSD, intraperitoneal injection of Fer-1, or their combination on tumor volume/weight, histopathology, and expressions of P53, SLC7A11 and GPX4 levels were evaluated.
RESULTS:
The active components in SSD (quercetin and wogonin) showed strong binding affinities to P53. In AGS cells, SSD treatment dose-dependently inhibited cell proliferation, increased ROS and Fe2+ levels, upregulated P53 expression, and downregulated the expressions of SLC7A11 and GPX4, but these effects were effectively attenuated by Fer-1 treatment. SSD also induced mitochondrial shrinkage and increased the membrane density, which were alleviated by Fer-1. In the tumor-bearing mouse models, gavage with SSD significantly reduced tumor size and weight, caused tumor cell necrosis, upregulated P53 and downregulated SLC7A11 and GPX4 expression in the tumor tissue, and these effects were obviously mitigated by Fer-1 treatment.
CONCLUSIONS
SSD inhibits gastric cancer growth in nude mice by inducing cell ferroptosis via the P53/SLC7A11/GPX4 axis.
Ferroptosis/drug effects*
;
Animals
;
Stomach Neoplasms/metabolism*
;
Tumor Suppressor Protein p53/metabolism*
;
Mice, Nude
;
Phospholipid Hydroperoxide Glutathione Peroxidase
;
Drugs, Chinese Herbal/pharmacology*
;
Humans
;
Amino Acid Transport System y+/metabolism*
;
Mice
;
Cell Line, Tumor
;
Cell Proliferation/drug effects*
;
Xenograft Model Antitumor Assays
9.Therapeutic mechanism of hederagenin, an active component in Guizhi Fuling Pellets, against cervical cancer in nude mice.
Yinfu ZHU ; Yiran LI ; Yi WANG ; Yinger HUANG ; Kunxiang GONG ; Wenbo HAO ; Lingling SUN
Journal of Southern Medical University 2025;45(7):1423-1433
OBJECTIVES:
To explore the therapeutic mechanism of Guizhi Fuling (GZFL) Pellets against cervical cancer.
METHODS:
Publicly available databases were used to identify the targets of GZFL Pellets and cervical cancer to construct the protein-protein interaction (PPI) network, followed by GO biological process and KEGG pathway enrichment analysis of the hub genes. The "Traditional Chinese Medicine-Active Ingredients-Targets-Pathways" network for GZFL Pellets in cervical cancer treatment was generated using Cytoscape v10.0.0, and molecular docking of the drug and potential targets was performed to predict the specific targets of active components in Guizhi Fuling Pellets. The inhibitory effects of hederagenin, an active ingredient in GZFL Pellets, was tested in cultured cervical cancer cells and in nude mice bearing cervical cancer xenografts.
RESULTS:
GZFL Pellets contain 338 active components targeting 247 action sites. A total of 10127 cervical cancer-related targets were obtained, and among them 195 were identified as potential therapeutic targets of GZFL Pellets for cervical cancer treatment, including the key targets of GABRA1, PTK2, JAK2, HTR3A, GSR, and IL-17. Molecular docking study showed low binding energies of the active components such as hederagenin, campesterol, and stigmasterol for protein-molecule interaction. GO enrichment analysis suggested that GZFL Pellets inhibited cervical cancer primarily by regulating responses to steroid hormones, oxidative stress, and lipopolysaccharides. Among the active components of GZFL Pellets, hederagenin was found to inhibit cervical cancer cells in vitro and significantly reduced STAT3 phosphorylation level in the cancer cells. In nude mice bearing cervical cancer xenografts, hederagenin effectively inhibited tumor growth rate without causing obvious adverse effects.
CONCLUSIONS
GZFL Pellets inhibit cervical cancer cell growth through its multiple active components that target different pathways. Among these components, hederagenin inhibits tumor cell growth possibly by directly binding to JAK2 protein to inhibit STAT3 phosphorylation.
Female
;
Animals
;
Uterine Cervical Neoplasms/pathology*
;
Mice, Nude
;
Humans
;
Mice
;
Oleanolic Acid/therapeutic use*
;
Drugs, Chinese Herbal/therapeutic use*
;
Molecular Docking Simulation
;
Xenograft Model Antitumor Assays
;
Cell Line, Tumor
;
STAT3 Transcription Factor/metabolism*
;
Protein Interaction Maps
;
Janus Kinase 2/metabolism*
10.Pirfenidone inhibits bladder cancer xenograft growth in mice by regulating regulatory T cells.
Hongbo ZHANG ; Mengyu YAN ; Jiandong ZHANG ; Peiwang SUN ; Rui WANG ; Yuanyuan GUO
Journal of Southern Medical University 2025;45(7):1513-1518
OBJECTIVES:
To investigate the inhibitory effect of pirfenidone (PFD) on growth of bladder cancer xenograft and its regulatory effect on Treg cells in tumor-bearing mice.
METHODS:
Thirty-two C57BL/6 mice bearing ectopic bladder tumors were randomized into control and PFD groups (n=16). In PFD group, PFD was administered orally at the daily dose of 500 mg/kg, and tumor growth and survival of the mice were monitored. After treatment for 21 days, the tumors and vital organs were harvested for analysis. Immunohistochemistry was used to assess CD3, CD4, CD8, and FOXP3 expressions in the tumors. Flow cytometry and RT-qPCR were used to analyze the percentage of CD4⁺CD25⁺FOXP3⁺ Treg cells and IL-2, IL-10, and IL-35 expressions in the tumors and spleens; organ damage of the mice was examined with HE staining.
RESULTS:
Compared with the control group, the PFD-treated mice exhibited significantly lower tumor growth rate with smaller tumor volumes at day 21, along with improved survival at day 28. Immunohistochemistry revealed no significant differences in the infiltration of CD3⁺ and CD8⁺ cells between the two groups, but the percentages of CD4⁺ and FOXP3⁺ cells were significantly lower in the tumors of PFD-treated mice. Flow cytometric analysis confirmed a decrease in CD4⁺CD25⁺FOXP3⁺ Treg cells in the tumors from PFD-treated mice, which also had reduced expression levels of IL-2, IL-10 and IL-35 mRNAs in the tumors. No significant differences were found in Treg cell populations or cytokine expressions in the spleen tissues between the two groups. HE staining showed obvious organ damage in neither of the groups.
CONCLUSIONS
PFD inhibits bladder cancer growth and enhances survival of tumor-bearing mice possibly by suppressing Treg cells in the tumor microenvironment.
Animals
;
Urinary Bladder Neoplasms/drug therapy*
;
Mice
;
T-Lymphocytes, Regulatory/metabolism*
;
Mice, Inbred C57BL
;
Interleukins/metabolism*
;
Interleukin-10/metabolism*
;
Cell Line, Tumor
;
Interleukin-2/metabolism*
;
Xenograft Model Antitumor Assays
;
Female

Result Analysis
Print
Save
E-mail