Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Int Immunopharmacol ; 132: 112054, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38608477

ABSTRACT

γδT cells are unconventional T cells only accounting for 1-5 % of circulating T lymphocytes. Their potent anti-tumor capability has been evidenced by accumulating studies. However, the prognostic value of γδT cells remains not well documented in head and neck squamous cell carcinoma (HNSCC). In this study, we utilized the TCGA HNSCC database to evaluate the infiltration of γδT cells and the association between γδT cells and clinicopathological factors by related gene signature, which were then validated by a total of 100 collected tumor samples from HNSCC patient cohort. Heterogeneity and functional characteristics of distinct infiltrating γδT cell profiles in HNSCC were then investigated based on the scRNA-seq data from the GEO database. We found higher γδT cell gene signature score was significantly associated with longer survival. Cox regression models showed that γδT cell gene signature could serve as an independent prognostic indicator for HNSCC patients. A high level of γδT cell-related gene signature was positively correlated with the infiltration of tumor-infiltrating lymphocytes and immune score. Through scRNA-seq analysis, we identified that γδ+ Trm cells and γδ+ CTL cells possessed anti-tumor and immunoregulatory properties. Notably, we found a significant association between the presence of these cells and improved survival outcomes. In our cell-cell communication analyses, we identified that γδT cells have the potential to eliminate tumor cells through the secretion of interferon-gamma and granzyme. Collectively, the infiltration of γδT cells may serve as a promising prognostic tool, prompting the consideration of treatment options for patients with HNSCC.


Subject(s)
Head and Neck Neoplasms , Lymphocytes, Tumor-Infiltrating , Squamous Cell Carcinoma of Head and Neck , Humans , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/mortality , Lymphocytes, Tumor-Infiltrating/immunology , Prognosis , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/mortality , Female , Male , Middle Aged , Transcriptome , Intraepithelial Lymphocytes/immunology , Gene Expression Regulation, Neoplastic , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Aged
2.
Biomolecules ; 14(2)2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38397469

ABSTRACT

The role of Pleckstrin homology-like domain family B member 2 (PHLDB2) in the regulation of cell migration has been extensively studied. However, the exploration of PHLDB2 in head and neck squamous cell carcinoma (HNSCC) is still limited in terms of expression, function, and therapeutic potential. In this study, we discovered an upregulation of PHLDB2 expression in HNSCC tissues which was correlated with a negative prognosis in patients with HNSCC. Additionally, we determined that a high level of expression of PHLDB2 is crucial for maintaining cell migration through the regulation of the epithelial-mesenchymal transition (EMT). Furthermore, we demonstrated that the ablation of PHLDB2 in tumor cells inhibited tumorigenicity in a C3H syngeneic tumor-bearing mouse model. Mechanistically, PHLDB2 was found to be involved in the regulation of T cell anti-tumor immunity, primarily by enhancing the activation and infiltration of CD8+ T cells. In light of these findings, PHLDB2 emerges as a promising biomarker and therapeutic target for interventions in HNSCC.


Subject(s)
Head and Neck Neoplasms , Animals , Mice , Humans , Squamous Cell Carcinoma of Head and Neck/genetics , Head and Neck Neoplasms/genetics , CD8-Positive T-Lymphocytes , Mice, Inbred C3H , Epithelial-Mesenchymal Transition/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Carrier Proteins
3.
Oral Dis ; 28(8): 2131-2138, 2022 Nov.
Article in English | MEDLINE | ID: mdl-33772979

ABSTRACT

OBJECTIVES: To identify independent factors for head and neck cancer (HNC) patients with carotid blowout syndrome (CBS) and construct a nomogram to predict risk of CBS preoperatively based on computed tomography angiography (CTA) imaging. SUBJECT AND METHODS: From January 2010 to July 2020, 73 HNC patients who had surgery in hospitalization and underwent CTA examination for head and neck region were included in this study. Vascular alterations and the relationship between carotid artery (CA) and tumor were evaluated in CTA. Clinical and CTA imaging features were distinguished by logistic regression analysis and used to perform receiver operating curve analysis. Nomogram was created to predict risk of CBS and assessed by concordance index (C-index) and calibration curve. RESULTS: Three independent risk factors were identified, including radical neck dissection, CA surrounded by tumor, and CA invaded by tumor without clear boundary. Area under curve of the combination of 3 variables was 0.836 (95% CI, 0.72-0.952, p < 0.001). The C-index of nomogram was 0.84 (95% CI, 0.73-0.94), and the calibration plot showed a good fitting between prediction and observation. CONCLUSIONS: We established a useful nomogram based on CTA imaging, which showed a satisfied efficacy for evaluating risk of CBS in HNC patients preoperatively.


Subject(s)
Carotid Artery Diseases , Head and Neck Neoplasms , Nomograms , Carotid Arteries/diagnostic imaging , Carotid Artery Diseases/diagnostic imaging , Computed Tomography Angiography , Head and Neck Neoplasms/diagnostic imaging , Humans , Retrospective Studies , Risk Factors , Syndrome
4.
J Magn Reson Imaging ; 56(1): 196-209, 2022 07.
Article in English | MEDLINE | ID: mdl-34888985

ABSTRACT

BACKGROUND: Adequate safe margin in tongue cancer radical surgery is one of the most important prognostic factors. However, the role of peritumoral tissues in predicting lymph node metastasis (LNM) and prognosis using radiomics analysis remains unclear. PURPOSE: To investigate whether magnetic resonance imaging (MRI)-based radiomics analysis with peritumoral extensions contributes toward the prediction of LNM and prognosis in tongue cancer. STUDY TYPE: Retrospective. POPULATION: Two hundred and thirty-six patients (38.56% female) with tongue cancer (training set, N = 157; testing set, N = 79; 37.58% and 40.51% female for each). FIELD STRENGTH/SEQUENCE: 1.5 T; T2-weighted turbo spin-echo images. ASSESSMENT: Radiomics models (Rprim , Rprim+3 , Rprim+5 , Rprim+10 , Rprim+15 ) were developed with features extracted from the primary tumor without or with peritumoral extensions (3, 5, 10, and 15 mm, respectively). Clinicopathological characteristics selected from univariate analysis, including MRI-reported LN status, radiological extrinsic lingual muscle invasion, and pathological depth of invasion (DOI) were further incorporated into radiomics models to develop combined radiomics models (CRprim , CRprim+3 , CRprim+5 , CRprim+10 , CRprim+15 ). Finally, the model performance was validated in the testing set. DOI was measured from the adjacent normal mucosa to the deepest point of tumor invasion. STATISTICAL TESTS: Chi-square test, regression analysis, receiver operating characteristic curve (ROC) analysis, decision analysis, spearman correlation analysis. The Delong test was used to compare area under the ROC (AUC). P < 0.05 was considered statistically significant. RESULTS: Of all the models, the CRprim+10 reached the highest AUC of 0.995 in the training set and 0.872 in the testing set. Radiomics features were significantly correlated with pathological DOI (correlation coefficients, -0.157 to -0.336). The CRprim+10 was an independent indicator for poor disease-free survival (hazard ratio, 5.250) and overall survival (hazard ratio, 17.464) in the testing set. DATA CONCLUSION: Radiomics analysis with a 10-mm peritumoral extension had excellent power to predict LNM and prognosis in tongue cancer.


Subject(s)
Tongue Neoplasms , Female , Humans , Lymph Nodes/pathology , Lymphatic Metastasis/diagnostic imaging , Lymphatic Metastasis/pathology , Magnetic Resonance Imaging/methods , Male , Prognosis , Retrospective Studies , Tongue Neoplasms/diagnostic imaging , Tongue Neoplasms/pathology
5.
Pharmazie ; 75(10): 494-499, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33305724

ABSTRACT

Platinum (Pt)-based drugs are routinely used to treat oral cancer (OC), but occurrence of therapeutic resistance remains a formidable challenge in cancer treatment. We sought to explore the cytotoxicity of non-classical Pt-based compounds, and compared the efficacy and anticancer activity of 56MESS with cisplatin in OC. Drug sensitivity of seven non-classical Pt-based compounds as well as cisplatin were determined by CCK-8 assay. Comparison of cytotoxic effects between 56MESS, phenanthriplatin and cisplatin was performed on six different OC cell lines. The anticancer effects of 56MESS was further measured both in vitro and in vivo. Additionally, the biological role of FACL4 and its relationship with 56MESS-induced growth inhibition were investigated. Two out of seven Pt-based compounds displayed a significant cytotoxic effect. 56MESS was chosen as the most potent compound due to its highly selective cytotoxic activity. 56MESS particularly caused G2/M phase arrest, while failed to induce apoptosis. In vivo, 56MESS had a higher cytotoxic capacity than cisplatin. Overexpression of FACL4 rescued 56MESS-induced growth inhibition in OC cells. Overall, 56MESS is a highly selective and potent chemotherapeutic drug superior to cisplatin, and thus may be considered as a promising anticancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Coenzyme A Ligases/genetics , Mouth Neoplasms/drug therapy , Organoplatinum Compounds/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Phenanthridines/pharmacology , Xenograft Model Antitumor Assays
6.
Theranostics ; 10(3): 1433-1453, 2020.
Article in English | MEDLINE | ID: mdl-31938073

ABSTRACT

Elucidation of the molecular mechanisms governing aggressiveness of HNSCC may provide clinical therapeutic strategies for patients. In this study, a novel hub miR-204-5p functioning as tumor suppressor has been identified and explored in HNSCC. Methods: A novel hub miR-204-5p was identified based on miRNA microarray, bioinformatics analysis and validated in different HNSCC patient cohorts. The functional role of miR-204-5p and its downstream and upstream regulatory machinery were investigated by gain-of-function and loss-of-function assays in vitro and in vivo. Interactions among miR-204-5p and SNAI2/SUZ12/HDAC1/STAT3 complex were examined by a series of molecular biology experiments. Then, the clinical relevance of miR-204-5p and its targets were evaluated in HNSCC samples. HNSCC patient-derived xenograft (PDX) model was used to assess the therapeutic value of miR-204-5p. Results: We reveal that miR-204-5p as a tumor suppressor is commonly repressed in HNSCC, which can inhibit tumor growth, metastasis and stemness. Mechanically, miR-204-5p suppresses epithelial-mesenchymal transition (EMT) and STAT3 signaling by targeting SNAI2, SUZ12, HDAC1 and JAK2. Among these targets, we further showed that SNAI2, SUZ12, and HDAC1 form a repressive complex on CDH1 promoter to maintain EMT in HNSCC. In turn, the SNAI2/SUZ12/HDAC1 complex interacts with STAT3 on miR-204-5p-regulatory regions to suppress the transcription of miR-204-5p. Moreover, we also show that decrease of miR-204-5p indicates a poor prognosis in HNSCC patients and administration of agomiR-204-5p inhibits tumor growth and metastasis in HNSCC PDX models. Conclusion: miR-204-5p-SNAI2/SUZ12/HDAC1/STAT3 regulatory circuit has a critical role in maintaining aggressiveness of HNSCC, suggesting that miR-204-5p might serve as a promising therapeutic target for clinical intervention.


Subject(s)
Genes, Tumor Suppressor , Head and Neck Neoplasms/metabolism , Lymphatic Metastasis/pathology , MicroRNAs/physiology , Squamous Cell Carcinoma of Head and Neck/metabolism , Adult , Aged , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , STAT3 Transcription Factor/metabolism
7.
Front Oncol ; 10: 624752, 2020.
Article in English | MEDLINE | ID: mdl-33732637

ABSTRACT

OBJECTIVES: Recently long non-coding RNAs (lncRNAs) have emerged as novel gene regulators involved in tumorigenic processes, including oral squamous cell carcinoma (OSCC). Here, we identified a differentiation-related lncRNA, terminal differentiation-induced non-coding RNA (TINCR). However, its biological function and clinicopathological significance in OSCC still remain unclear. METHODS: The lncRNA expression profiles in OSCC tissues and paired adjacent non-tumor tissues (NATs) from 10 patients were detected by lncRNA microarrays. Weighted gene co-expression network analysis (WGCNA) and gene ontology (GO) enrichment were performed to identify the most significant module and module functional annotation, respectively. Potential differentiation-related lncRNAs were screened by differential expression analysis. TINCR was further confirmed in OSCC cell lines and tissues of another patient cohort by using qRT-PCR. The correlation between the TINCR expression level and clinicopathological characteristics was analyzed. The effects of TINCR on cell differentiation, migration and invasion were assessed by knockdown or knock-in in vitro and in vivo. RESULTS: WGCNA and GO enrichment analysis showed that one co-expression network was significantly enriched for epithelial cell differentiation, among which, TINCR was significantly downregulated. qRT-PCR analyses validated down-regulation of TINCR in tumor tissues compared with paired NATs, and its expression was closely correlated with pathological differentiation and lymph node metastasis in patients with OSCC. Patients with lower TINCR expression levels had worse survival. Cell function experiments showed that TINCR played a crucial role in epithelial differentiation. Both TINCR and epithelial differentiation-associated genes, including IVL and KRT4, were significantly upregulated during OSCC cell calcium-induced differentiation but were reduced when cell dedifferentiation occurred in tumor spheres. Overexpression of TINCR dramatically suppressed cell dedifferentiation, migration and invasion in vitro, while knockdown of TINCR had the opposite effects. Upregulation of TINCR significantly elevated the expression of terminal differentiation genes and repressed tumor growth in vivo. Moreover, TINCR significantly suppressed the activation of JAK2/STAT3 signaling in OSCC cells. CONCLUSION: Our study suggests that TINCR functions as a tumor suppressor by inducing cell differentiation through modulating JAK2/STAT3 signaling in OSCC. TINCR may serve as a prognostic biomarker and therapeutic target for OSCC.

8.
Histopathology ; 74(5): 766-779, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30444275

ABSTRACT

AIMS: Tumour budding and invasive depth can predict survival of patients with tongue squamous cell carcinoma (TSCC), while the prognostic value of tumour microenvironment (TME) remains unknown. Here, both characteristics of the tumour and its microenvironment were examined and a novel prognostic model has been proposed. METHODS AND RESULTS: A total of 246 patients with TSCC were included. Using H&E-stained sections, pathological parameters of tumour and the TME were assessed. Inflammatory response (i), tumour budding (B) and invasive depth (D) were combined as iBD score. The association between these variables and the patient survival was determined. Both tumour budding and inflammatory status were independent variables for predicting overall survival (OS) and disease-free survival (DFS) of TSCC patients. Invasive depth was correlated with differentiation, T classification, lymph node metastasis, clinical stage and recurrence (P < 0.05). The novel iBD model was strongly correlated with T classification, lymph node metastasis, clinical stage and recurrence, and showed clear distinction of scores 0, 1 and 2. High iBD score had a strong association with reduced OS and DFS (P < 0.01). CONCLUSIONS: The iBD scoring model is strongly associated with lymph node metastasis and recurrence in TSCC and could be a promising survival predictor for TSCC patients.


Subject(s)
Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Tongue Neoplasms/mortality , Tongue Neoplasms/pathology , Tumor Microenvironment , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/surgery , China , Disease-Free Survival , Female , Follow-Up Studies , Hospitals, University , Humans , Kaplan-Meier Estimate , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness/diagnosis , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Proportional Hazards Models , Research Design , Tongue Neoplasms/surgery , Young Adult
9.
Int J Mol Med ; 41(1): 147-154, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29138797

ABSTRACT

Tongue squamous cell carcinoma (TSCC) is more aggressive than other cancers in the head and neck region because of its potential for metastasis. Recently, ß2­adrenergic receptor (ß2­AR) has been reported to be a potential promoter in various types of solid cancer. However, the role of ß2­AR and its effect on TSCC is not well documented. Histological staining, western blot analysis, migration and invasion assay were used. In this study, the expression of ß2­AR was increased in TSCC tissue compared with adjacent non­cancerous epithelium. Further analysis demonstrated that increased expression of ß2­AR was correlated with differentiation, lymph node metastasis and reduced overall survival rate in patients with TSCC. In vitro studies confirmed that activation of ß2­AR can promote epithelial mesenchymal transition in TSCC by initiating an interleukin­6/Stat3/Snail1 pathway. These results suggest that ß2­AR has an oncogenic role in TSCC and may be a potential therapeutic target in TSCC.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Squamous Cell/genetics , Receptors, Adrenergic, beta-2/genetics , Tongue Neoplasms/genetics , Aged , Carcinoma, Squamous Cell/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Disease-Free Survival , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/genetics , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Middle Aged , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Snail Family Transcription Factors/genetics , Tongue Neoplasms/pathology
10.
Oncol Rep ; 38(4): 2051-2061, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28849187

ABSTRACT

Accumulating data suggest that microRNAs (miRNAs) play a pivotal role in the regulation of tumor cell sensitivity to chemotherapeutic agents. Although the roles of a few miRNAs have been identified in cisplatin resistance, little is known in regards to the concerted contribution of miRNA­mediated biological networks. In the present study, we demonstrated that microRNA-218 (miR-218) was significantly upregulated in cisplatin-resistant oral cancer cells. The results of cell viability and apoptosis assay showed that ectopic expression of miR-218 induced cell survival and resistance to cisplatin, whereas suppression of miR-218 caused apoptosis and enhanced sensitivity to cisplatin. Moreover, we identified PPP2R5A as a new direct target of miR-218 by using the dual luciferase reporter assay. Overexpression of miR-218 led to inhibition of PPP2R5A expression, whereas knockdown of miR-218 increased PPP2R5A levels. Introduction of PPP2R5A abrogated miR­218-mediated cell survival and drug resistance. Furthermore, suppression of miR-218 or PPP2R5A significantly promoted or reduced cisplatin-induced apoptosis, respectively. Finally, PPP2R5A overexpression or ß-catenin knockdown inhibited miR-218-mediated Wnt activation and partially restored cell sensitivity. Our data revealed a molecular link between miR-218 and PPP2R5A/Wnt signaling and implicates miR-218 as a potential target for oral cancer therapy.


Subject(s)
Cisplatin/pharmacology , MicroRNAs/metabolism , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Protein Phosphatase 2/metabolism , Wnt Signaling Pathway , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Knockdown Techniques , Humans , MicroRNAs/biosynthesis , MicroRNAs/genetics , Mouth Neoplasms/genetics , Up-Regulation
11.
Oncotarget ; 8(13): 21954-21973, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423539

ABSTRACT

TP63 acts as a master regulator in epithelia development and in the progression of various cancers, but its role in oral cancer pathogenesis remains unknown. This study aimed to explore the role of TP63 in the progression of oral squamous cell carcinoma (OSCC). This study shows that ΔNp63, the predominant isoform of TP63, is significantly upregulated in OSCC tissues and cell lines compared with their normal counterparts, and its expression is closely correlated with pathological differentiation, lymph node metastasis and clinical stage in patients with OSCC. The overexpression of ΔNp63 promotes growth, metastasis and stem-like properties in OSCC cells, and ΔNp63 depletion significantly represses OSCC cellular phenotypes in vitro and in vivo. The ΔNp63 isoform transcriptionally suppresses miR-138-5p expression; restoration of miR-138-5p expression partially abolishes the effect of upregulating ΔNp63. This study also demonstrates that miR-138-5p directly targets ΔNp63, resulting in crosstalk with ΔNp63. The correlation between ΔNp63 and miR-138-5p was further validated in OSCC tissues and was found to be significantly associated with the prognosis of patients with OSCC. Therefore, our data reveal that the interplay between ΔNp63 and miR-138-5p promotes OSCC progression by regulating cell growth, metastasis and stemness.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Proliferation , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Mouth Neoplasms/pathology , Neoplastic Stem Cells/pathology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Movement , Female , Humans , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Neoplasm Staging , Neoplastic Stem Cells/metabolism , Prognosis , Survival Rate , Transcription Factors/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
12.
Oncotarget ; 7(40): 65744-65757, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27582550

ABSTRACT

We aimed to determine the specific miRNA profile of tumor budding cells and investigate the potential role of miR-320a in invasion and metastasis of tongue squamous cell carcinoma (TSCC). We collected tumor budding cells and paired central tumor samples from five TSCC specimens with laser capture microdissection and examined the specimens using a miRNA microarray. The specific miRNA signature of tumor budding cells was identified. We found that miR-320a was dramatically decreased in tumor budding cells. Knockdown of miR-320a significantly enhanced migration and invasion of TSCC cell lines. Suz12 was shown to be a direct target of miR-320a. Similar results were also observed in nude mouse models. Multivariate analysis indicated that miR-320a was an independent prognostic factor. Kaplan-Meier analysis demonstrated that decreased miR-320a and high intensity of tumor budding were correlated with poor survival rate, especially in the subgroup with high-intensity tumor budding and low expression of miR-320a. We concluded that decreased expression of miR-320a could promote invasion and metastasis of tumor budding cells by targeting Suz12 in TSCC. A combination of tumor budding and miR-320a may serve as an index to identify an aggressive sub-population of TSCC cells with high metastatic potential.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/secondary , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Polycomb Repressive Complex 2/metabolism , Tongue Neoplasms/pathology , Animals , Apoptosis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Proliferation , Female , Follow-Up Studies , Humans , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins , Prognosis , Survival Rate , Tongue Neoplasms/genetics , Tongue Neoplasms/metabolism , Transcription Factors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...