Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
BMC Med ; 22(1): 207, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769543

ABSTRACT

BACKGROUND: Tumor-infiltrating lymphocyte (TIL) therapy has been restricted by intensive lymphodepletion and high-dose intravenous interleukin-2 (IL-2) administration. To address these limitations, we conducted preclinical and clinical studies to evaluate the safety, antitumor activity, and pharmacokinetics of an innovative modified regimen in patients with advanced gynecologic cancer. METHODS: Patient-derived xenografts (PDX) were established from a local recurrent cervical cancer patient. TILs were expanded ex vivo from minced tumors without feeder cells in the modified TIL therapy regimen. Patients underwent low-dose cyclophosphamide lymphodepletion followed by TIL infusion without intravenous IL-2. The primary endpoint was safety; the secondary endpoints included objective response rate, duration of response, and T cell persistence. RESULTS: In matched patient-derived xenografts (PDX) models, homologous TILs efficiently reduced tumor size (p < 0.0001) and underwent IL-2 absence in vivo. In the clinical section, all enrolled patients received TIL infusion using a modified TIL therapy regimen successfully with a manageable safety profile. Five (36%, 95% CI 16.3-61.2) out of 14 evaluable patients experienced objective responses, and three complete responses were ongoing at 19.5, 15.4, and 5.2 months, respectively. Responders had longer overall survival (OS) than non-responders (p = 0.036). Infused TILs showed continuous proliferation and long-term persistence in all patients and showed greater proliferation in responders which was indicated by the Morisita overlap index (MOI) of TCR clonotypes between infused TILs and peripheral T cells on day 14 (p = 0.004) and day 30 (p = 0.004). Higher alteration of the CD8+/CD4+ ratio on day 14 indicated a longer OS (p = 0.010). CONCLUSIONS: Our modified TIL therapy regimen demonstrated manageable safety, and TILs could survive and proliferate without IL-2 intravenous administration, showing potent efficacy in patients with advanced gynecologic cancer. TRIAL REGISTRATION: NCT04766320, Jan 04, 2021.


Subject(s)
Interleukin-2 , Lymphocytes, Tumor-Infiltrating , Humans , Female , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , Interleukin-2/administration & dosage , Interleukin-2/therapeutic use , Animals , Aged , Adult , Mice , Genital Neoplasms, Female/therapy , Genital Neoplasms, Female/drug therapy , Treatment Outcome , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/therapeutic use
2.
Reprod Biol Endocrinol ; 21(1): 46, 2023 May 16.
Article in English | MEDLINE | ID: mdl-37194006

ABSTRACT

Efficient evaluation of the primordial follicle pool (PFP) of mammalian models is an essential subject in biomedical research relating to ovarian physiology and pathogenesis. Our recent study has identified a gene signature including Sohlh1, Nobox, Lhx8, Tbpl2, Stk31, Padi6, and Vrtn strongly correlated with ovarian reserve by using bioinformatics analysis. Aimed to investigate the validity of these candidate biomarkers for evaluating the PFP, we utilized an OR comparison model to decode the relationship between the numbers of PFP and candidate biomarkers in the present study. Our results suggest that these biomarkers Sohlh1, Nobox, Lhx8, Tbpl2, Stk31, Padi6, and Vrtn possess independent potential to evaluate the number of the PFP. And the combination of Sohlh1 and Lhx8 can be used as the optimal biomarkers for rapid assessment of the PFP in the murine ovary. Our findings provide a new perspective for evaluating the PFP of the ovary in animal studies and the clinic.


Subject(s)
Ovarian Follicle , Transcription Factors , Animals , Female , Mice , Basic Helix-Loop-Helix Transcription Factors/genetics , Ovarian Follicle/physiology , Ovary , Transcription Factors/genetics
3.
Biomolecules ; 13(2)2023 02 09.
Article in English | MEDLINE | ID: mdl-36830707

ABSTRACT

BACKGROUND: Ovarian cancer (OC) is one of the most malignant tumors in the female reproductive system, with a poor prognosis. Various responses to treatments including chemotherapy and immunotherapy are observed among patients due to their individual characteristics. Applicable prognostic markers could make it easier to refine risk stratification for OC patients. Autophagy is closely implicated in the occurrence and development of tumors, including OC. Whether autophagy -related genes can be used as prognostic markers for OC patients remains unclear. METHODS: The gene transcriptome data of 374 OC patients were downloaded from The Cancer Genome Atlas (TCGA) database. The correlation between the autophagy levels and outcomes of OC patients was identified through the single sample gene set enrichment analysis (ssGSEA). Recognized molecular markers of autophagy in different clinical specimens were detected by immunohistochemistry (IHC) assay. The gene set enrichment analysis (GSEA), ESTIMATE, and CIBERSORT analysis were applied to explore the correlation of autophagy with the tumor immune microenvironment (TIME). Single-cell RNA-sequencing (scRNA-seq) data from seven OC patients were included for characterizing cell-cell interaction patterns of autophagy-high or low tumor cells. Machine learning, Stepwise Cox regression and LASSO-Cox analysis were used to screen autophagy hub genes, which were used to establish an autophagy-related signature for prognosis evaluation. Four tumor immunotherapy cohorts were obtained from the GEO (Gene Expression Omnibus) database and the literature for autophagy risk score validation. RESULTS: The autophagy levels were closely related to the prognosis of the OC patients. Additionally, the autophagy levels were correlated with TIME status including immune score, and immune-cell infiltration. The scRNA-seq analysis found that tumor cells with high or low autophagy levels had different interactions with immune cells, especially macrophages. Eight autophagy-hub genes (ZFYVE1, AMBRA1, LAMP2, TRAF6, PDPK1, ATG2B, DAPK1 and TP53INP2) were screened for an autophagy-related signature. According to this signature, higher risk score was correlated with poor prognosis and better immunotherapy response in the OC patients. CONCLUSIONS: The autophagy-related signature is applicable to predict the prognosis and immune checkpoint inhibitors (ICIs) therapy efficiency in OC patients. It is possible to identify OC patients who will respond to ICIs therapy and have a favorable prognosis, although more verification is needed.


Subject(s)
Genes, Regulator , Ovarian Neoplasms , Humans , Female , Immunotherapy , Autophagy , Biological Assay , Tumor Microenvironment , Nuclear Proteins , Adaptor Proteins, Signal Transducing , Autophagy-Related Proteins , Vesicular Transport Proteins , 3-Phosphoinositide-Dependent Protein Kinases
4.
Int J Gen Med ; 16: 367-391, 2023.
Article in English | MEDLINE | ID: mdl-36756390

ABSTRACT

Background: CD276 (also known as B7-H3), a newly discovered immunoregulatory protein that belongs to the B7 family, is a significant and attractive target for cancer immunotherapy. Existing evidence demonstrates its pivotal role in the tumorigenesis of some cancers. However, there still lacks a systematic and comprehensive pan-cancer analysis of the role of CD276 in tumor immunology and prognosis. Methods: We explored and validated the mRNA and protein expression levels of CD276 in multiple tumors through public databases and clinical tissues specimens. The Univariate Cox regression analysis and Kaplan-Meier analysis were applied to assess the prognostic value of CD276. The correlation between CD276 expression and clinical characteristics and immunological features in diverse tumors was also explored. GSEA was performed to illuminate the biological function and involved pathways of CD276. Moreover, the CellMiner database was used to interpret the relationship between CD276 and multiple chemotherapeutic agents. CCK-8 assay was performed to validate the biological function of CD276 in vitro. Results: In general, CD276 was differentially expressed between most tumor tissues and their corresponding normal tissues. Higher expression levels of CD276 were associated with poorer survival outcomes in most tumor cohorts from TCGA. There was a close correlation between CD276 expression and clinical features, the infiltration levels of specific immune cells, immune subtypes, TMB, MSI, MMR, recognized immunoregulatory genes and drug sensitivity across diverse human cancers. The scRNA-seq data analysis further revealed that CD276 was mainly expressed on the tumor infiltrating macrophages. Additionally, in vitro experiments showed that knockdown of CD276 inhibited the proliferation of ovarian cancer (OV) and cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) cell lines. Conclusion: CD276 is a potent biomarker for predicting the prognosis and immunological features in some tumors, and it may play a critical role in the tumor immune microenvironment (TIME) through macrophage-associated signaling.

5.
J Transl Med ; 20(1): 201, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35538537

ABSTRACT

PURPOSE: As a common complication of epithelial ovarian cancer (EOC), malignant ascites contributes to the peritoneal metastasis of EOC. CircRNAs play essential roles in tumor metastasis. However, no circRNAs have been reported to be involved in EOC peritoneal metastasis via ascites. METHODS: Total of 22 samples from 9 EOC patients containing primary lesions (T), tumor cells from ascites (ASC), and metastatic lesions (M) were included for RNA sequencing to identify differentially expressed circRNAs and mRNAs among different tumors. Bioinformatic analyses, including single-sample Gene Set Enrichment Analysis and soft cluster analysis, were performed to find circRNAs potentially correlated with ascitic metastasis. Wound healing and transwell analysis were performed to evaluate tumor cells metastasis in vitro. Quantitative real-time PCR and western-blot were used for gene expression evaluation. RESULTS: According to transcriptomic analysis, ASC showed mesenchymal phenotype while T and M showed epithelial phenotype. 10 circRNAs were differentially expressed among ASC, T, and M. Among them, hsa_circ_0000497 and hsa_circ_0000918 were significantly up-regulated in ASC. Functional analysis showed that both hsa_circ_0000497 and hsa_circ_0000918 promoted metastasis of EOC via epithelial-mesenchymal transition (EMT) in vitro. The regulatory network construction identified 8 miRNAs and 19 mRNAs, and 7 miRNAs and 17 mRNAs as potential downstream target genes of hsa_circ_0000497 and hsa_circ_0000918, respectively, which may play pivotal roles in EOC ascitic metastasis. CONCLUSIONS: circRNAs (hsa_circ_0000497 and hsa_circ_0000918) contribute to metastasis of EOC via ascites by regulating EMT. These circRNAs may serve as novel potential therapeutic targets or prognostic biomarkers for EOC peritoneal metastasis.


Subject(s)
MicroRNAs , Ovarian Neoplasms , Peritoneal Neoplasms , Ascites/genetics , Carcinoma, Ovarian Epithelial/genetics , Female , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Ovarian Neoplasms/genetics , Peritoneal Neoplasms/genetics , RNA, Circular/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Cancer Cell Int ; 22(1): 15, 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35012539

ABSTRACT

BACKGROUND: Accumulating evidence has revealed that aberrant microRNA (miRNA) expression can affect the development of chemotherapy drug resistance by modulating the expression of relevant target proteins. Emerging evidence has demonstrated that miR-133a participates in the tumorigenesis of various cancers. However, whether miR-133a is associated with cisplatin resistance in ovarian cancer remains unclear. OBJECTIVE: To investigate the role of miR-133a in the development of cisplatin resistance in ovarian cancer. METHODS: MiR-133a expression in cisplatin-resistant ovarian cancer cell lines was assessed by reverse-transcription quantitative PCR (RT-qPCR). A cell counting kit-8 (CCK-8) assay was used to evaluate the viability of tumour cells treated with cisplatin in the presence or absence of miR-133a. A luciferase reporter assay was used to analyse the binding of miR-133a with the 3' untranslated region (3'UTR) of YES proto-oncogene 1 (YES1). The YES1 expression level was analysed using a dataset from the International Cancer Genome Consortium (ICGC) and assessed by RT-qPCR and western blotting in vitro. The roles and mechanisms of YES1 in cell functions were further probed via gain- and loss-of-function analysis. RESULTS: The expression of miR-133a was significantly decreased in cisplatin-resistant ovarian cancer cell lines (A2780-DDP and SKOV3-DDP), and the overexpression of the miR-133a mimic reduced cisplatin resistance in A2780-DDP and SKOV3-DDP cells. Treatment with the miR-133a inhibitor increased cisplatin sensitivity in normal A2780 and SKOV3 cells. MiR-133a binds the 3'UTR of YES1 and downregulates its expression. Bioinformatics analysis revealed that YES1 expression was upregulated in recurrent cisplatin-resistant ovarian cancer tissue, and in vitro experiments also verified its upregulation in cisplatin-resistant cell lines. Furthermore, we discovered that miR-133a downregulated the expression of YES1 and thus inhibited cell autophagy to reduce cisplatin resistance. Yes1 knockdown significantly suppressed the cisplatin resistance of ovarian cancer cells by inhibiting autophagy in vitro. Xenograft tumour implantation further demonstrated that Yes1 overexpression promoted ovarian tumour development and cisplatin resistance. CONCLUSIONS: Our results suggest that the miR-133a/YES1 axis plays a critical role in cisplatin resistance in human ovarian cancer by regulating cell autophagy, which might serve as a promising therapeutic target for ovarian cancer chemotherapy treatment in the future.

7.
J Cancer ; 11(15): 4413-4420, 2020.
Article in English | MEDLINE | ID: mdl-32489460

ABSTRACT

Background: Immunotherapy including immune checkpoint blockade, cancer vaccines, and adoptive cell therapy. However, no immune therapies support ovarian cancer. It is not clear whether the neutrophils, the component of the immune system derived from umbilical cord blood play a role in inhibiting the progression of ovarian cancer. Methods: We investigate the impact of LPS and IL-8 activated neutrophils derived from umbilical cord blood(UCB)on ovarian cancer progression. After co-culture LPS and IL-8 activated UCB-derived neutrophils with ovarian cancer cell line SKOV3 and OVCAR3, CCK8, Transwell assay, and Flow Cytometry was performed to detect cell proliferation, migration, invasion, and apoptosis of ovarian cancer cell lines SKOV3 and OVCAR3. Furthermore, RT-PCR and western blotting assay were used to analyze the mechanism of metastasis and apoptosis of ovarian cancer cell lines respectively to support previous function experiments. Results: We demonstrate LPS and IL-8 activated neutrophils derived from umbilical cord blood inhibit proliferation, invasion migration and promote apoptosis of SKOV3 and OVCAR3. Meanwhile, LPS and IL-8 activated UCB-derived neutrophils significantly decreased BAX and increased BCL2 expression in SKOV3 and OVCAR3 which account for the mechanism of apoptosis. Moreover, LPS and IL-8 activated UCB derived neutrophils significantly up-regulated E-cadherin and downregulated N-cadherin, MMP2 expression in SKOV3 and OVCAR3. Conclusion: Taken together, these results approved that LPS and IL-8 activated neutrophils from UCB may be the novel strategy in immune therapy for ovarian cancer.

8.
Can J Cardiol ; 32(2): 151-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26454467

ABSTRACT

BACKGROUND: Iron deficiency (ID) is a common comorbidity in patients with heart failure (HF) and has been associated with increased mortality and hospitalizations. However, the benefit and safety of iron supplementation in treating HF and ID in randomized controlled trials (RCTs) are inconclusive. We therefore performed a meta-analysis to overcome this limitation. METHODS: PubMed, the Cochrane Library, and ClinicalTrials.gov were systematically searched for eligible trials up to December 31, 2014. We also searched the references of all relevant studies and reviews for more trials. Only RCTs reporting the clinical impact of iron therapy in patients with HF with ID compared with no iron treatment were enrolled in our meta-analysis. RESULTS: Five clinical trials comprising a total of 907 patients were finally included. Compared with placebo or no treatment, additional iron therapy was associated with a significantly reduced rate of hospitalization for HF (odds ratio [OR], 0.28; 95% confidence interval [CI], 0.16-0.49), though all-cause mortality was not significantly different (OR, 0.81; 95% CI, 0.42-1.57). In 4 studies where these endpoints were combined, the incidence of hospitalization for HF and death was lowered in the iron supplementation group (OR, 0.47; 95% CI, 0.29-0.76). There was no increase in the risk of adverse events. CONCLUSIONS: Iron supplementation significantly reduced the risk of (a) hospitalization for HF and (b) the combined endpoint of hospitalization for HF and death, without increasing the risk of adverse events in patients with symptomatic systolic HF and ID. However, the current data are inadequate to make a clear determination upon mortality.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Heart Failure/complications , Iron Compounds/administration & dosage , Nutritional Support/methods , Administration, Oral , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/complications , Global Health , Heart Failure/blood , Heart Failure/mortality , Humans , Survival Rate
9.
J Hum Genet ; 60(11): 709-16, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26334104

ABSTRACT

To examine microRNA-133a (miR-133a) endogenous expression in cardiomyocytes after ischemia-reperfusion (I/R) injury and study the effects of miR-133a overexpression on I/R injury-induced cardiomyocyte apoptosis. Dual-Luciferase Reporter Assay detected dynamic expression of miR-133a. In an in vitro hypoxia-reoxygenation (HR) injury model and an in vivo rat model of I/R injury, rat cardiomyocytes were transfected with miR-133a mimic to test the effects of miR-133a overexpression on apoptosis. MiR-133a and Death Associated Protein Kinase 2 (DAPK2) mRNA expression was measured using real-time-PCR, and DAPK2 protein expression was detected by western blotting. Annexin V-fluorescein isothiocyanate/propidium iodide (PI) double-staining measured the apoptosis rate in H9C2 cells and transferase dUTP nick end labeling assay quantified the cardiomyocyte apoptosis rate in tissues obtained from in vivo the rat model. DAPK2 is a target of miR-133a. Both in vitro and in vivo results confirmed that after expression of miR-133a mimics, miR-133a levels increased, which was accompanied by decrease in DAPK2 mRNA and protein expression. In H9C2 cells, HR injury caused a sharp decrease in miR-133a expression and a significant upregualtion of DAPK2 mRNA and protein levels. However, exogenous miR-133a expression led to a significant reduction in DAPK2 mRNA and protein levels despite HR injury. Similar results were obtained from in vivo I/R injury model. After HR injury or I/R injury the apoptosis rate of myocardial cells was highly elevated and decreased significantly only after transfection of miR-133a into cardiomyocytes. MiR-133a overexpression may inhibit I/R injury-mediated cardiomyocyte apoptosis by targeting DAPK2, leading to reduced DAPK2 protein, thus miR-133a may potentially have a high therapeutic value in I/R injury.


Subject(s)
Death-Associated Protein Kinases/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Reperfusion Injury/prevention & control , Animals , Apoptosis/genetics , Cell Line , Death-Associated Protein Kinases/genetics , Disease Models, Animal , Female , Gene Targeting , Models, Cardiovascular , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Up-Regulation
10.
Am J Cardiol ; 116(10): 1521-6, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26385518

ABSTRACT

Rosuvastatin and atorvastatin both are high-intensity statins. However, which statin is more effective for the reversion of coronary atherosclerotic plaques remains inconclusive. We, therefore, conducted a meta-analysis to provide further evidence for proper statin selection. Pubmed, The Cochrane Library, Embase, Chinese BioMedicine, and China National Knowledge Infrastructure databases were systematically searched for eligible publications. We also manually reviewed the references from all relevant literature for more trials. Only studies that met our predefined inclusion criteria up to March 31, 2015, were enrolled. Five randomized controlled trials, 4 published in English and 1 in Chinese, were finally included in our study with a total of 1,556 participants, of whom 772 were in the rosuvastatin group and 784 in the atorvastatin group. The dose ratios of rosuvastatin versus atorvastatin were 1:2 in all included trials. Pooling across the studies demonstrated that compared with atorvastatin, rosuvastatin administration further reduced the total atheroma volume (weighted mean difference [WMD] -1.61 mm(3), 95% confidence interval [CI] -2.70 to -0.52; p = 0.004) and percent atheroma volume (WMD -0.34%, 95% CI -0.64 to -0.03; p = 0.03) and improved the lumen volume more significantly (WMD 2.10 mm(3), 95% CI 0.04 to 4.17; p = 0.046). The comparative regression of plaques was not different across subgroups. In conclusion, rosuvastatin is superior to atorvastatin in the reversion of coronary atherosclerotic plaques.


Subject(s)
Atorvastatin/administration & dosage , Coronary Vessels/diagnostic imaging , Plaque, Atherosclerotic/drug therapy , Rosuvastatin Calcium/administration & dosage , Coronary Artery Disease/drug therapy , Disease Progression , Dose-Response Relationship, Drug , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Plaque, Atherosclerotic/diagnostic imaging , Treatment Outcome , Ultrasonography, Interventional
SELECTION OF CITATIONS
SEARCH DETAIL
...