Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Biomol Biomed ; 2024 07 14.
Article in English | MEDLINE | ID: mdl-39036926

ABSTRACT

Immune checkpoint inhibitors (ICIs) enhance the tumor-killing ability of T-cells in non-small cell lung cancer (NSCLC), improving overall survival (OS) and revolutionizing treatment for advanced stages. However, challenges remain, such as low response rates and the lack of effective markers for selecting candidates. This study evaluated the impact of hemoglobin, albumin, and platelet (HALP), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio (PLR) on the efficacy of immunotherapy and survival outcomes in advanced NSCLC. Additionally, it aimed to develop a nomogram based on these parameters. Clinical and hematological data from NSCLC patients who received immunotherapy were analyzed. Efficacy was assessed using the immune Response Evaluation Criteria in Solid Tumors (iRECIST), and progression-free survival (PFS) and OS were evaluated. Prediction models incorporated baseline and post-treatment HALP, NLR, and PLR values. The 203 patients had a median follow-up of 16 months, a median PFS (mPFS) of seven months (6.0­8.0), while the median OS (mOS) was not reached (24.0­not available). Pretreatment PLR (PLR0) was associated with a higher disease control rate (DCR) (odds ratio [OR] = 0.258), while initial immunotherapy and NLR after four treatment cycles (NLR4C) significantly improved the objective response rate (ORR). Cox regression analysis showed that pretreatment HALP (HALP0), HALP after four cycles of treatment (HALP4C), and pretreatment NLR (NLR0) significantly impacted PFS. Additionally, HALP0, NLR0, and PLR after four treatment cycles (PLR4C) were associated with OS. The C-indices for PFS and OS were 0.823 and 0.878, respectively, indicating good predictive accuracy. HALP, NLR, and PLR at various time points effectively predicted immunotherapy response in advanced NSCLC patients, with low HALP combined with high NLR and PLR indicating a poor prognosis. These findings could serve as the basis for stratified randomized controlled trials (RCTs) in the future.

2.
Invest New Drugs ; 39(5): 1411-1418, 2021 10.
Article in English | MEDLINE | ID: mdl-33765213

ABSTRACT

Primary acinar soft part sarcoma of the lung (ASPS) is a rare malignancy with unique cellular structure and clinical and genetic characteristics. Most patients do not exhibit clear clinical symptoms, with only a few developing respiratory symptoms. The typical histological characteristics are acinoid or organ-like structures. Immunofluorescence in situ hybridization suggests a rearrangement of the transcription factor E3 gene. Patients respond poorly to chemotherapy and are, thus, primarily treated with surgery and targeted therapy. We report herein a unique case of primary alveolar soft part sarcoma of the lung. The patient was a 24-year-old man with metastases to multiple organs, such as the brain, lungs, pancreas, and liver. The craniocerebral lesions attained partial remission after whole-brain radiotherapy and targeted combined immunotherapy, and other distant metastases completely disappeared after targeted combined immunotherapy (anlotinib and camrelizumab), indicating significant treatment efficacy. Anlotinib is an oral multi-target tyrosine kinase inhibitor (TKI) that exerts its anti-tumor effects by acting on various kinases. Camrelizumab is a humanized immunoglobulin G4 monoclonal antibody that can target PD-1 to block the interaction between PD-L1 and programmed death ligand 2, ultimately causing an anti-tumor effect. This is the first report of successful use of anlotinib combined with camrelizumab in the treatment of advanced primary ASPS. The treatment benefit provides preliminary evidence that targeted therapy, combined with immunotherapy, may be a safe and effective approach to treat primary pulmonary ASPS patients, thus warranting further investigation.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Indoles/therapeutic use , Lung Neoplasms/drug therapy , Quinolines/therapeutic use , Sarcoma, Alveolar Soft Part/drug therapy , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols , Humans , Indoles/administration & dosage , Indoles/adverse effects , Lung Neoplasms/pathology , Male , Neoplasm Metastasis , Quinolines/administration & dosage , Quinolines/adverse effects , Young Adult
4.
Exp Cell Res ; 382(1): 111450, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31152708

ABSTRACT

The metastasis-associated gene 1 (MTA1) has previously been recognized as an oncogene in many tumors, and aberrant MTA1 expression has been related to invasion and migration; however, its role and underlying molecular mechanism in oral squamous carcinoma (OSCC) remain largely unexplored. In this work, we determined the expression of MTA1 in OSCC tissues and cell lines. The effect of MTA1 on metastasis and the role of MTA1 in the epithelial-to-mesenchymal transition (EMT) of OSCC cells were evaluated by assays both in vitro and in vivo. We also identified the key Hedgehog signaling pathway-related protein involved in the MTA1-induced EMT. We found that MTA1 expression was upregulated and positively related to the metastasis in OSCC tissues and cell lines. MTA1 overexpression promoted OSCC invasion, migration, and induced EMT, while its silencing had the opposite effect both in vitro and in vivo. Additionally, our data further revealed the relevant molecular mechanism, Hedgehog(Hh) signaling pathway contributed to the effect of MTA1 on the aggressive phenotypes of OSCC cells.These findings indicate that MTA1 enhances OSCC cells invasion and migration by inducing EMT via the Hedgehog signaling pathway, which suggests MTA1 may be an effective anti-OSCC therapeutic target.


Subject(s)
Carcinoma, Squamous Cell/pathology , Epithelial-Mesenchymal Transition/physiology , Hedgehog Proteins/physiology , Mouth Neoplasms/pathology , Neoplasm Proteins/physiology , Repressor Proteins/physiology , Signal Transduction/physiology , Trans-Activators/physiology , Zinc Finger Protein GLI1/physiology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Cadherins/biosynthesis , Cadherins/genetics , Cell Line, Tumor , Cell Movement , Female , Heterografts , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Trans-Activators/antagonists & inhibitors , Trans-Activators/biosynthesis , Trans-Activators/genetics , Up-Regulation
5.
Gene ; 704: 91-96, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-30965130

ABSTRACT

LncRNA CASC11 is a recently identified oncogenic lncRNA in colorectal cancer. This study aimed to investigate the role of lncRNA CASC11 in small cell lung cancer (SCLC). In the present study, expression levels of CASC11 and TGF-ß1 were found to be positively and significantly correlated with the percentage of CDD133+ cells of SCLC cell lines. Plasma CASC11 and TGF-ß1 were upregulated and positively correlated in SCLC patients, but not in healthy controls. Upregulation of plasma CASC11 and TGF-ß1 predicted poor survival of SCLC patients. Overexpression of CASC11 and TGF-ß1 also resulted in the increased percentage of CDD133+ cells of SCLC cell lines, while TGF-ß inhibitor attenuated the effects of CASC11 overexpression. CASC11 overexpression mediated the upregulation of TGF-ß1 in SCLC cells, while treatment with exogenous TGF-ß1 showed no significant effect on CASC11. Therefore, lncRNA CASC11 promotes TGF-ß1, increases cancer cell stemness and predicts postoperative survival in SCLC.


Subject(s)
Lung Neoplasms/diagnosis , Lung Neoplasms/surgery , Neoplastic Stem Cells/physiology , RNA, Long Noncoding/physiology , Small Cell Lung Carcinoma/diagnosis , Small Cell Lung Carcinoma/surgery , Transforming Growth Factor beta1/genetics , Adult , Aged , Biomarkers, Tumor/physiology , Case-Control Studies , Cell Proliferation/genetics , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Postoperative Period , Prognosis , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Survival Analysis , Treatment Outcome , Up-Regulation/genetics
6.
J Exp Clin Cancer Res ; 32(1): 54, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23941622

ABSTRACT

BACKGROUND: The prognostic value of metastasis-associated gene 1 (MTA1) in nasopharyngeal carcinoma (NPC) has been suggested. However, there is still no direct evidence that MTA1 promotes NPC growth in vivo. In this study, we aimed to investigate the function of MTA1 in the regulation of NPC cell proliferation and tumorigenesis in vitro and in vivo. METHODS: Stable MTA1 knockdown or overexpression NPC cell lines were employed. The effects of MTA1 depletion or overexpression on cell proliferation, colony formation, cell cycle progression were examined by MTT, colony formation and flow cytometry assay. The effects of MTA1 depletion on tumor growth in vivo were examined in mouse xenograft model. RESULTS: MTA1 knockdown or overexpression drastically changed the proliferation, colony formation and cell cycle of NPC cells in vitro. MTA1 depletion significantly suppressed NPC tumorigenesis in vivo. CONCLUSION: MTA1 promotes NPC cell proliferation via enhancing G1 to S phase transition, leading to increased tumor growth. Targeting MTA1 is a promising approach to reduce tumor burden of NPC.


Subject(s)
Histone Deacetylases/metabolism , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Repressor Proteins/metabolism , Animals , Carcinoma , Cell Cycle/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Disease Models, Animal , Female , Gene Knockdown Techniques , Heterografts , Histone Deacetylases/deficiency , Histone Deacetylases/genetics , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Repressor Proteins/deficiency , Repressor Proteins/genetics , Trans-Activators
7.
Int J Biochem Cell Biol ; 45(7): 1439-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23618874

ABSTRACT

Nasopharyngeal carcinoma (NPC) is prone to appearing regional lymph node and distant metastasis. And its underlying mechanism is unclear. Recent study suggests that overexpression of metastasis-associated gene 1 (MTA1) was independently associated with poorer distant metastasis-free survival in NPC. However, it is still lack of direct evidence that MTA1 is responsible for aggressive phenotypes of NPC. Using stably transfected MTA1 knockdown or overexpression cells, we discovered the function of MTA1 in actin cytoskeleton reorganization and metastasis processing of NPC in this study. For the first time, our data demonstrate two tumor relevant molecular mechanisms, i.e. Rho GTPases and Hedgehog signaling both contribute to the effect of MTA1 on the aggressive phenotypes of NPC cells. In summary, the novel findings in this work provide further insight into the function of MTA1 and the molecular mechanism in the progression of NPC. Our results indicate that MTA1 might serve as a potential therapeutic target for advanced NPC.


Subject(s)
Actin Cytoskeleton/metabolism , Hedgehog Proteins/metabolism , Histone Deacetylases/metabolism , Nasopharyngeal Neoplasms/metabolism , Repressor Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Carcinoma , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Humans , Lymphatic Metastasis , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/mortality , Neoplasm Invasiveness , Repressor Proteins/genetics , Signal Transduction , Survival , Trans-Activators , Up-Regulation
8.
Oncol Rep ; 28(1): 218-24, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22576802

ABSTRACT

Metastasis-associated gene 1 (MTA1) is involved in the carcinogenesis and metastasis of many human carcinomas. However, its exact role in non-small cell lung cancer (NSCLC) is still unclear. Using immunohistochemistry analysis, we recently identified MTA1 to be associated with the progression of NSCLC. Here, we carried out further analysis on the effect of MTA1 knockdown in an NSCLC cell line on cell functions and the global microRNA (miRNA) expression profile. We succeeded in establishing the MTA1 knockdown NSCLC cell line using RNA interference (RNAi), and found that the silencing of MTA1 resulted in the effective inhibition of the invasive ability of NSCLC cells, but not of the cell growth in vitro. We performed an miRNA microarray analysis and demonstrated for the first time that MTA1 knockdown significantly changed the expression of some miRNAs in NSCLC cells. Among them, some have a well-characterized association with cancer progression, e.g. miR-125b, miR-210, miR-103, miR-194 and miR-500. In summary, it is evident from our results that MTA1 functions in regulating the invasive phenotype of lung cancer cells and this regulation may be through altered miRNA expression. The interaction between MTA1 and the miRNAs which contributes to lung cancer is worthy of further investigation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/secondary , Histone Deacetylases/genetics , Lung Neoplasms/pathology , MicroRNAs/metabolism , RNA Interference , Repressor Proteins/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Gene Expression Profiling , Gene Knockdown Techniques , Histone Deacetylases/metabolism , Humans , Lung Neoplasms/metabolism , MicroRNAs/genetics , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Phenotype , Real-Time Polymerase Chain Reaction , Repressor Proteins/metabolism , Trans-Activators , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL