Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cancer Biomark ; 40(2): 171-184, 2024.
Article in English | MEDLINE | ID: mdl-38517779

ABSTRACT

INTRODUCTION: GINS2 exerts a carcinogenic effect in multiple human malignancies, while it is still unclear that the potential roles and underlying mechanisms of GINS2 in HNSCC. METHODS: TCGA database was used to screen out genes with significant differences in expression in HNSCC. Immunohistochemistry and qRT-PCR were used to measure the expression of GINS2 in HNSCC tissues and cells. GINS2 was detected by qRT-PCR or western blot after knockdown or overexpression. Celigo cell counting, MTT, colony formation, and flow cytometric assay were used to check the ability of proliferation and apoptosis. Bioinformatics and microarray were used to screen out the downstream genes of GINS2. RESULTS: GINS2 in HNSCC tissues and cells was up-regulated, which was correlated with poor prognosis. GINS2 gene expression was successfully inhibited and overexpressed in HNSCC cells. Knockdown of GINS2 could inhibit proliferation and increase apoptosis of cells. Meanwhile, overexpression of GINS2 could enhance cell proliferation and colony formation. Knockdown of RRM2 may inhibit HNSCC cell proliferation, while overexpression of RRM2 rescued the effect of reducing GINS2 expression. CONCLUSION: Our study reported the role of GINS2 in HNSCC for the first time. The results demonstrated that in HNSCC cells, GINS2 promoted proliferation and inhibited apoptosis via altering RRM2 expression. Therefore, GINS2 might play a carcinogen in HNSCC, and become a specific promising therapeutic target.


Subject(s)
Apoptosis , Cell Proliferation , Chromosomal Proteins, Non-Histone , Gene Expression Regulation, Neoplastic , Ribonucleoside Diphosphate Reductase , Squamous Cell Carcinoma of Head and Neck , Humans , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cell Proliferation/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Cell Line, Tumor , Apoptosis/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/metabolism , Disease Progression , Prognosis , Female , Male , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism
2.
Int J Radiat Oncol Biol Phys ; 118(5): 1347-1370, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38092257

ABSTRACT

Radiation therapy is a primary treatment for cancer, but radioresistance remains a significant challenge in improving efficacy and reducing toxicity. Accumulating evidence suggests that deubiquitinases (DUBs) play a crucial role in regulating cell sensitivity to ionizing radiation. Traditional small-molecule DUB inhibitors have demonstrated radiosensitization effects, and novel deubiquitinase-targeting chimeras (DUBTACs) provide a promising strategy for radiosensitizer development by harnessing the ubiquitin-proteasome system. This review highlights the mechanisms by which DUBs regulate radiosensitivity, including DNA damage repair, the cell cycle, cell death, and hypoxia. Progress on DUB inhibitors and DUBTACs is summarized, and their potential radiosensitization effects are discussed. Developing drugs targeting DUBs appears to be a promising alternative approach to overcoming radioresistance, warranting further research into their mechanisms.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Antineoplastic Agents/therapeutic use , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Neoplasms/radiotherapy , Neoplasms/drug therapy , Deubiquitinating Enzymes/metabolism , Radiation Tolerance
3.
J Radiat Res ; 65(1): 10-27, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-37981296

ABSTRACT

Ionizing radiation (IR) induces ferroptosis in head and neck squamous cell carcinoma (HNSCC). But, it remains unclear whether ferroptosis affects the prognosis of HNSCC patients after receiving radiotherapy. This study aims to develop a ferroptosis signature to predict the radiosensitivity and prognosis of HNSCC. Ferroptosis-related genes, clinical data and RNA expression profiles were obtained from the FerrDb database, The Cancer Genome Atlas and GEO database. Prognostic genes were identified by random survival forest, univariate Cox regression, Kaplan-Meier and ROC analyses. Principal component analysis, multivariate Cox regression, nomogram and DCA analyses were conducted to estimate its predictive ability. Functional enrichment and immune-related analyses were performed to explore potential biological mechanisms and tumor immune microenvironment. The effect of the hub gene on ferroptosis and radiosensitivity was verified using flow cytometry, quantitative real-time PCR and clonogenic survival assay. We constructed a ferroptosis-related signature, including IL6, NCF2, metadherin (MTDH) and CBS. We classified patients into high-risk (HRisk) and low-risk groups according to the risk scores. The risk score was confirmed to be an independent predictor for overall survival (OS). Combining the clinical stage with the risk score, we established a predictive nomogram for OS. Furthermore, pathways related to tumorigenesis and tumor immune suppression were mainly enriched in HRisk. MTDH was verified to have a potent effect on IR-induced ferroptosis and consequently promoted radiosensitivity. We constructed a ferroptosis-related signature to predict radiosensitivity and OS in HNSCC patients. MTDH was identified as a promising therapeutic target in radioresistant HNSCC patients.


Subject(s)
Ferroptosis , Head and Neck Neoplasms , Humans , Prognosis , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Ferroptosis/genetics , Transcription Factors , Radiation Tolerance/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/radiotherapy , Tumor Microenvironment , Membrane Proteins/genetics , RNA-Binding Proteins
4.
IUBMB Life ; 75(9): 702-716, 2023 09.
Article in English | MEDLINE | ID: mdl-36973940

ABSTRACT

The long non-coding RNA (lncRNA)-microRNA (miRNA) interaction network plays a crucial part in the pathogenesis of nasopharyngeal carcinoma (NPC). Here, we discovered a relationship between LINC01376 and miR-4757 in NPC tumor development. First, LINC01376 was abnormally overexpressed in NPC tissues and cells, and its elevated expression was associated with advanced clinical stage and shorter distant metastasis-free survival time. Moreover, biological experiments showed that LINC01376 facilitated the proliferative, invasive, and migratory abilities of NPC cells in vitro and in vivo. Mechanistically, bioinformatics and RT-qPCR assays revealed that LINC01376 knockdown upregulated the expression level of downstream miR-4757, including miR-4757 primary transcript (pri-miR-4757) and mature miR-4757. Furthermore, LINC01376 competitively sponged the transcription factor SP1 and reduced its enrichment in the upstream promoter region of miR-4757 to repress miR-4757 expression. Finally, insulin-like growth factor 1(IGF1) was identified as the target of miR-4757. Rescue experiments indicated that LINC01376 accelerated NPC cell proliferation, migration, and invasion through the miR-4757-5p/IGF1 axis. In conclusion, the SP1/miR-4757/IGF1 axis, which is regulated by LINC01376 in NPC deterioration and metastasis, is expected to provide new insights into the molecular mechanism of NPC carcinogenesis.


Subject(s)
MicroRNAs , Nasopharyngeal Neoplasms , RNA, Long Noncoding , Humans , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/metabolism , Insulin-Like Growth Factor I/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Cell Proliferation/genetics , Cell Movement/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
5.
J Gastrointest Oncol ; 13(3): 912-922, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35837168

ABSTRACT

Background: The goal of the current research was to investigate circATXN7 expression in esophageal cancer (EC) and its impact on the proliferation, migration, and invasion of EC cells. Methods: Determination of circATXN7 expression in esophageal cancer tissues and adjacent tissueswas carried out using quantitative reverse transcription polymerase chain reaction (qRT-PCR), and we further analyzed the correlation between patients' clinical characteristics and circATXN7 expression. EC cell lines (EC-9706, Eca-109, TE-1, KYSE-30, and KYSE-150) and normal esophageal cell line (HET-1A) were cultured, and circATXN7 expression was detected by qRT-PCR. The lowest circATXN7-containing Eca-109 cells were selected to be transfected with an overexpressing lentiviral vector (circATXN7). EC-9706 cells with the highest expression of circATXN7 were selected for transfection with knockdown vectors [short hairpin RNA (shRNA)#1 and shRNA#2] of the circATXN7 sequence. Cell proliferation was determined via MTT assay. The formation of cell clones was investigated via colony formation assay. Transwell migration assay was utilized to determine cell migration and invasion ability. Results: Significantly higher levels of circATXN7 were observed in EC tissues compared with paracancerous tissues (P<0.01), and circATXN7 expression level showed a significant correlation with the tumor/lymph nodes/metastasis (TNM) stage and metastasis of lymph nodes (P<0.05). Among all esophageal cell lines, EC-9706 had the highest expression level and Eca-109 had the lowest expression level. The MTT assay revealed that circATXN7 overexpression could significantly promote the proliferation of Eca-109 cells, while circATXN7 knockdown was capable of significantly inhibiting EC cell proliferation. The colony formation experiments revealed a significant increase in the number of clones in the circATXN7 overexpression model and a significant decrease in the circATXN7 knockdown model. The results of transwell migration experiments suggested that circATXN7 overexpression could promote EC cell invasion and migration, while knockdown of circATXN7 expression was associated with significant inhibition of the invasion and migration of these cells. Conclusions: CircATXN7 exerted a critical role in the incidence and progression of EC. This study identified a novel molecular target and established a theoretical basis for the early detection and treatment of EC.

6.
J Biomed Res ; 36(4): 221-230, 2022 May 28.
Article in English | MEDLINE | ID: mdl-35715194

ABSTRACT

It is difficult for physicians to identify patients with metastatic nasopharyngeal carcinoma (NPC) who are sensitive to local treatment of metastases. Here, we aimed to establish a prognostic model for survival and individualize treatments for patients with metastatic NPC. Data were collated from 240 NPC patients diagnosed with metachronous metastasis between 2006 and 2020 who received palliative chemotherapy with or without local treatment. Multivariable Cox regression was implemented to construct a nomogram which had a concordance index of 0.764 when predicting 1-, 3-, and 5-year overall survival (OS). We then classified patients according to risk, creating low- and high-risk groups using the nomogram. Differences in OS between the two groups were significant ( P<0.001). In the low-risk group, the OS for patients who received local treatment was longer than those without ( P=0.009). This novel nomogram shows good performance in classifying patients according to risk and may also be a promising tool for determining who responds best to local treatment. Further validation using external center data is warranted.

7.
J Thorac Dis ; 13(5): 3061-3069, 2021 May.
Article in English | MEDLINE | ID: mdl-34164197

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) are firmly identified with the event and improvement of tumors. Therefore, elucidating the functions and mechanisms of related lncRNAs is significant for understanding the occurrence and advancement of tumors. The recently discovered lncRNA TUC338 has been shown to play the role of an oncogene in an assortment of tumors. Be that as it may, the articulation and elements of lncRNA TUC338 in esophageal cancer are as yet hazy. This investigation plans to explain the capacities and related molecular mechanisms of lncRNA TUC338 in esophageal malignancy. METHODS: Firstly, the expression of TUC338 in 50 instances of esophageal disease tissues and nearby tissues was detected by fluorescence reckonable PCR, and correlations with the clinic pathological characteristics of patients was further analyzed. Then, a lentiviral interference vector was designed and transfected into an esophageal cancer cell line, and knockdown was verified by fluorescence quantitative PCR. The effects of TUC338 knockdown on the proliferation, clone formation, and migration and infringement of esophageal malignancy cells were tested utilizing the CCK-8 assay, clone formation experiments, and Transwell experiments. Western blot detected the expression of invasion-related proteins. RESULTS: Fluorescence reckonable PCR exhibit that TUC338 was exceptionally communicated in esophageal cancer tissues, and was significantly related with metastasis and TNM stage in tolerant. Functional experiments showed that in esophageal disease cell lines, knocking down the declaration of TUC338 significantly inhibited cell multiplication, clone development, and intrusion and movement. Further experiments on molecular mechanisms showed that knocking down TUC338 inhibited statement of N-cadherin and vimentin in cells. CONCLUSIONS: TUC338 is exceptionally communicated in esophageal malignancy tissues and can regulate cell proliferation and invasion.

8.
Int J Biol Sci ; 17(8): 1963-1978, 2021.
Article in English | MEDLINE | ID: mdl-34131399

ABSTRACT

Nasopharyngeal carcinoma (NPC) is known for its potential to progress to the lymph nodes and distant metastases at an early stage. As an important regulator in tumorigenesis biological processes, the functions of lncRNA in NPC tumor development remain largely unclear. In this research, the expression of EPB41L4A-AS2 in NPC tissues and cells was analyzed via real-time quantitative polymerase chain reaction (qRT-PCR). CCK8, colony formation, and EDU experiments were used to determine the viability of NPC cells. Transwell and wound healing assays were performed to test NPC cell migration and invasion. RNA pull-down and mass spectrometry analysis were used to identify potential binding proteins. Then, a popliteal lymph node metastasis model was established to test NPC metastasis. EPB41L4A-AS2 is repressed by transforming growth factor-beta, which is downregulated in NPC cells and tissue. It is associated with the presence of distant metastasis and adverse outcomes. The univariate and multivariate survival assays confirmed that EPB41L4A-AS2 expression was an independent predictor of progression-free survival (PFS) in patients with NPC. Biological analyses showed that overexpression of EPB41L4A-AS2 reduced the metastasis and invasion of NPC in vitro and in vivo, but had no significant effect on cell proliferation. Mechanistically, in the nucleus we identified that EPB41L4A-AS2 relies on binding to YBX1 to reduce the stability of Snail mRNA to enhance the expression of E-cadherin and reverse the progression of epithelial-to-mesenchymal transition (EMT). In the cytoplasm, we found that EPB41L4A-AS2 blocked the invasion and migration of NPC cells by promoting LATS2 expression via sponging miR-107. In a whole, the findings of this study help to further understand the metastasis mechanism of NPC and could help in the prevention and treatment of NPC metastasis.


Subject(s)
Cytoskeletal Proteins , Lymphatic Metastasis , Membrane Proteins , MicroRNAs/metabolism , Nasopharyngeal Carcinoma , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Y-Box-Binding Protein 1/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Down-Regulation , Drug Discovery , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Lymphatic Metastasis/prevention & control , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transforming Growth Factors
9.
Am J Cancer Res ; 9(4): 779-790, 2019.
Article in English | MEDLINE | ID: mdl-31106003

ABSTRACT

Recently, long non-coding RNAs (lncRNAs) have been reported as the vital regulators of various cancers including nasopharyngeal carcinoma (NPC). An increasing number of studies have suggested that lncRNA LINC01133 is dysregulated and involved in human carcinogenesis. However, the roles of LINC01133 in NPC remain largely unknown. In this work, we demonstrated that LINC01133 was significantly downregulated in NPC tissues and cell lines. Loss and gain of function experiments provided evidence that LINC01133 inhibited NPC cell proliferation, invasion and migration both in vitro and in vivo. Besides, Fluorescence in situ hybridization (FISH) assay was performed to determine the localization of LINC01133 and LINC01133 was observed mainly distributed in the nucleus. Importantly, RNA pull-down and RIP assays showed that LINC01133 directly combined with YBX1, and YBX1 can partly reverse the repression of NPC cell proliferation, migration, and invasion caused by LINC01133. Collectively, our exploration indicate that LINC01133 inhibits the malignant-biological behavior of NPC cells by binding to YBX1, thereby suggesting a novel biomarker for the NPC prognosis and treatment.

10.
Artif Cells Nanomed Biotechnol ; 47(1): 833-843, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30862190

ABSTRACT

We aimed to explore the mediating role of Notch signal in macrophage polarization. This signal was knocked out from macrophages of Lyz2 cre and RBP-J flox mice. Bone marrow-derived macrophages (BMDMs) were isolated and polarized. The expressions of polarization markers in BMDMs 24 h after transfection were detected by qPCR. After Notch knockout, the expressions of M1 markers decreased but those of M2 markers increased significantly. MiR-125a/miR-99b and Spaca6 were highly and lowly expressed upon M1 and M2 polarizations, respectively. The expressions of experimental group were significantly lower than those of control group. Overexpression of miR-125a significantly promoted the expressions of M1 markers, whereas inhibited those of M2 markers. NO release in the culture supernatant of miR-125a overexpression group significantly exceeded that of control group. Transfection with miR-125a inhibitor significantly down-regulated IL-12 expression but up-regulated MR expression in BMDMs. The supernatant secreted by M1 macrophages significantly facilitated BS524 cell apoptosis, which was enhanced after miR-125a overexpression. The TNF-α expression of miR-99b overexpression group increased whereas that of MR decreased significantly. The miR-125a/miR-99b cluster contained an RBP-J specific recognition site in the first intron of initial transcript. The Notch signalling pathway promoted macrophage polarization into M1 phenotype by up-regulating miR-125a/miR-99b expression.


Subject(s)
Gene Expression Regulation , Macrophages/immunology , MicroRNAs/genetics , Receptors, Notch/metabolism , Signal Transduction , Animals , Biomarkers/metabolism , Cell Line, Tumor , Gene Knockout Techniques , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Macrophage Activation/genetics , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout , MicroRNAs/antagonists & inhibitors , Phenotype , Receptors, Notch/genetics , Seminal Plasma Proteins/genetics
11.
Cancer Med ; 8(3): 1197-1208, 2019 03.
Article in English | MEDLINE | ID: mdl-30741461

ABSTRACT

Metastasis and invasion are the primary causes of malignant progression in esophageal squamous cell carcinoma (ESCC). Epithelial-mesenchymal transition (EMT) is crucial step of acquisition of "stemness" properties in tumor cells. However, the mechanism of esophageal cancer metastasis remains unclear. This research was designed to explore the role and mechanism of SMAD4 and miR-130a-3p in the progression of transforming growth factor-ß (TGF-ß)-induced EMT in vivo and in vitro. The expression of miR-130a-3p in ESCC cell line and normal esophageal epithelial cell was determined by RT-qPCR. The protein expression levels of TGF-ß-induced changes in EMT were analyzed by western blotting and immunofluorescence. Dual-luciferase report assays were used to validate the regulation of miR-130a-3p-SMAD4 axis. The effect of miR-130a-3p and SMAD4 in TGF-ß-induced migration, invasion in the ESCC cell line EC-1 was investigated by wound healing assays and Transwell assays. Here we found that knocked down SMAD4 could partially reverse TGF-ß-induced migration, invasion, and EMT progression in the ESCC cell line EC-1. miR-130a-3p, which directly targets SMAD4, is down-regulated in ESCC. miR-130a-3p inhibits the migration and invasion of EC-1 cells both in vitro and in vivo. Finally, miR-130a-3p inhibits TGF-ß-induced EC-1 cell migration, invasion, and EMT progression in a SMAD4-dependent way. In conclusion, this study provides new insights into the mechanism underlying ESCC metastasis. The TGF-ß/miR-130a-3p/SMAD4 pathway could be potential targets for clinical treatment of ESCC.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Smad4 Protein/genetics , Transforming Growth Factor beta1/genetics , 3' Untranslated Regions , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Gene Knockdown Techniques , Heterografts , Humans , Male , Mice , Smad4 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
12.
Biomed Pharmacother ; 99: 688-696, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29710466

ABSTRACT

Transforming growth factor beta (TGF-?), a pleiotropic cytokine, promotes cell proliferation and migration in multiple cancers, including nasopharyngeal carcinoma (NPC). microRNA-124 (miR-124) becomes downregulated in NPC and inhibits the tumorigenesis of this disease. However, the role of miR-124 in TGF-?-induced NPC development remains unknown. In this study, constant TGF-? stimulation repressed miR-124 expression, whereas miR-124 overexpression antagonized TGF-?-promoted NPC cell growth and migration. miR-124 overexpression decreased p-SMAD2/3, SMAD4, and p-ERK levels, indicating that ectopic miR-124 overexpression inhibited SMAD and non-SMAD pathways. Pro-oncogenic lncRNA MALAT1 was targeted by miR-124 that regulated ERK/MAPK by targeting MALAT1 independent of the SMAD signaling pathway. In conclusion, our work clarified the significant role of miR-124 in TGF-? signaling pathways independent of the SMAD signaling pathway and showed the potential of miR-124 as a new therapeutic target against NPC.


Subject(s)
Carcinoma/pathology , MicroRNAs/genetics , Nasopharyngeal Neoplasms/pathology , RNA, Long Noncoding/genetics , Transforming Growth Factor beta/metabolism , Carcinoma/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System/genetics , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Signal Transduction/genetics , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Smad4 Protein/metabolism
13.
Onco Targets Ther ; 11: 997-1004, 2018.
Article in English | MEDLINE | ID: mdl-29520150

ABSTRACT

Extranodal NK/T-cell lymphoma is closely associated with Epstein-Barr virus (EBV) infection. However, the prognostic value of EBV-DNA in extranodal NK/T-cell lymphoma remains unclear. Thus, we conducted a meta-analysis to estimate its prognostic significance. PubMed, EMBASE, and Web of Science were used to search for studies conducted until June 12, 2017. The pooled hazard ratio (HR) and its 95% confidence interval (CI) were calculated to evaluate the prognostic value of pretreatment EBV-DNA on the overall survival of extranodal NK/T-cell lymphoma. Seven eligible studies on 356 patients with extranodal NK/T-cell lymphoma were pooled for this meta-analysis. Results suggested that the pretreatment EBV-DNA positivity was significantly correlated with the overall survival of extranodal NK/T-cell lymphoma (pooled HR =3.78, 95% CI: 1.52-9.40, p=0.004; heterogeneity test: I2=52%, p=0.05). Subgroup analyses stratified by sample type, survival analysis mode, and HR origin showed that patients with positive pretreatment EBV-DNA had poorer prognosis than those with negative pretreatment EBV-DNA. Moreover, the cut-off value (HR =1.66; 95% CI: 0.73-3.73; p=0.22) might account for the heterogeneity. No significant publication bias was observed. Pretreatment EBV-DNA positivity can predict poor prognosis for patients with extranodal NK/T-cell lymphoma. Future large-scale studies based on prognostic significance of EBV-DNA for patients with extranodal NK/T-cell lymphoma are necessary.

SELECTION OF CITATIONS
SEARCH DETAIL
...