Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.351
Filter
1.
J Immunother Cancer ; 12(7)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39038919

ABSTRACT

BACKGROUND: Addition of neoadjuvant immune checkpoint inhibition to standard-of-care interventions for locally advanced oral cancer could improve clinical outcome. METHODS: In this study, 16 evaluable patients with stage III/IV oral cancer were treated with one dose of 480 mg nivolumab 3 weeks prior to surgery. Primary objectives were safety, feasibility, and suitability of programmed death receptor ligand-1 positron emission tomography (PD-L1 PET) as a biomarker for response. Imaging included 18F-BMS-986192 (PD-L1) PET and 18F-fluorodeoxyglucose (FDG) PET before and after nivolumab treatment. Secondary objectives included clinical and pathological response, and immune profiling of peripheral blood mononuclear cells (PBMCs) for response prediction. Baseline tumor biopsies and postnivolumab resection specimens were evaluated by histopathology. RESULTS: Grade III or higher adverse events were not observed and treatment was not delayed in relation to nivolumab administration and other study procedures. Six patients (38%) had a pathological response, of whom three (19%) had a major (≥90%) pathological response (MPR). Tumor PD-L1 PET uptake (quantified using standard uptake value) was not statistically different in patients with or without MPR (median 5.3 vs 3.4). All major responders showed a significantly postnivolumab decreased signal on FDG PET. PBMC immune phenotyping showed higher levels of CD8+ T cell activation in MPR patients, evidenced by higher baseline expression levels of PD-1, TIGIT, IFNγ and lower levels of PD-L1. CONCLUSION: Together these data support that neoadjuvant treatment of advanced-stage oral cancers with nivolumab was safe and induced an MPR in a promising 19% of patients. Response was associated with decreased FDG PET uptake as well as activation status of peripheral T cell populations.


Subject(s)
Mouth Neoplasms , Neoadjuvant Therapy , Humans , Male , Female , Mouth Neoplasms/drug therapy , Mouth Neoplasms/diagnostic imaging , Mouth Neoplasms/pathology , Middle Aged , Neoadjuvant Therapy/methods , Aged , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Molecular Imaging/methods , Nivolumab/therapeutic use , Nivolumab/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Positron-Emission Tomography/methods , Adult
2.
J Cancer Res Ther ; 20(3): 745-749, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-39023577

ABSTRACT

ABSTRACT: The current scoping review's objective was to outline existing applications, recent breakthroughs, and quantum dots' applicability in imaging of oral squamous cell cancer. Quantum dots are nanometric semiconductor crystals with customizable optical characteristics and intense, stable fluorescence suited for bioimaging and labeling. We used the Preferred reporting items for systematic reviews and meta-analyses (PRISMA) recommendations for conducting our systematic search. An analysis of the properties and applications of quantum dots in noninvasive detection of oral squamous cell cancer is presented in this study, which comprehensively explores the available evidence. Following searches in the databases PubMed, Ovid SP, and Cochrane using the search terms quantum dots AND oral squamous cell cancer, 55 published publications were chosen for this review. The review identified a total of eight papers that met the criteria. In noninvasive detection of oral squamous cell carcinoma, quantum dots have the potential to offer an array of therapeutic and diagnostic applications. Furthermore, quantum dots emit near-infrared and visible light, which is advantageous in biological imaging since it reduces light dispersion and absorption of tissue. The future may see quantum dots become a popular noninvasive imaging technique for oral squamous cell cancer. The number of studies accessible is quite limited, and further research is required.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Quantum Dots , Quantum Dots/chemistry , Humans , Mouth Neoplasms/diagnostic imaging , Mouth Neoplasms/diagnosis , Mouth Neoplasms/pathology , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/pathology , Optical Imaging/methods
3.
J Cancer Res Ther ; 20(3): 770-775, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-39023581

ABSTRACT

INTRODUCTION: CX3CL1 exhibits chemoattraction for T-cells, monocytes, and CD57+ natural killer cells mediating antitumor immunity. The role of CX3CL1 has been studied in tumors of the breast, lung, colon, pancreas, prostate, etc. The current study was undertaken to understand the importance of CX3CL1 and its correlation with CD57+ cells in oral squamous cell carcinoma (OSCC). MATERIAL AND METHODS: Seventy-five primary OSCC were staged and histopathologically graded, followed by immunohistochemistry for CX3CL1 and CD57. Mann-Whitney U-test, Kruskal-Wallis test, Post hoc Bonferroni test, and Pearson's correlation coefficient were applied. RESULTS: CX3CL1 assessment within the tumor cells was high in 62.66% of cases, and the CD57 Labeling Index (LI) varied over a wide range of 8.2-111.6. A statistically significant reduction in expression of both CX3CL1 and CD57 was observed with an increase in histologic grade (p = 0.021 and 0.038, respectively). DISCUSSION: It is concluded that CX3CL1 and CD57 may be important players in the immune surveillance of OSCC. Further studies with detailed follow-up for the overall survival of patients will help in studying the diagnostic, prognostic, and therapeutic roles of CX3CL1 in OSCC.


Subject(s)
CD57 Antigens , Carcinoma, Squamous Cell , Chemokine CX3CL1 , Mouth Neoplasms , Humans , Chemokine CX3CL1/metabolism , Mouth Neoplasms/pathology , Mouth Neoplasms/immunology , Mouth Neoplasms/metabolism , CD57 Antigens/metabolism , Male , Female , Middle Aged , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/immunology , Aged , Adult , Prognosis , Immunohistochemistry , Biomarkers, Tumor/metabolism , Neoplasm Grading , Neoplasm Staging , Lymphocytes/metabolism , Lymphocytes/immunology , Lymphocytes/pathology
4.
J Cancer Res Ther ; 20(3): 750-754, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-39023578

ABSTRACT

BACKGROUND: Moderately advanced and technically unresectable oral cavity cancers have a poor prognosis. Neoadjuvant chemotherapy might be beneficial in such patients by reducing tumour bulk and allowing definitive surgery. AIM: To evaluate the response of neoadjuvant chemotherapy in moderately advanced technically unresectable oral cavity cancers. METHODOLOGY: Prospective observational study - secondary data analysis of patients with moderately advanced oral cavity cancer, which were treated with neoadjuvant chemotherapy (NACT) during the period November 2014-April 2016. Data was analysed for information on patient characteristics, chemotherapy received, toxicity, clinical response rates, local treatment offered and pathological response rates. The statistical analysis was performed with SPSS version 20. RESULTS: 30 patients, with a median age of 52 years were analyzed. Buccal mucosa was the most common sub site (50%). Three drug regimen was utilized in all patients. Resectability was achieved in 14 patients (46.67%). Febrile neutropenia was seen in 3 patients (10%). The overall response rate was 31%. CONCLUSION: NACT was effective in converting moderately advanced technically unresectable oral cavity cancers to operable disease in approximately 47% of patients. Post NACT, there is significant association between clinical and pathological findings of response rates. There is no increase in surgical complication rates following NACT.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Mouth Neoplasms , Neoadjuvant Therapy , Humans , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Mouth Neoplasms/surgery , Neoadjuvant Therapy/methods , Middle Aged , Male , Female , Aged , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Treatment Outcome , Prospective Studies , Neoplasm Staging , Prognosis
5.
J Cancer Res Ther ; 20(3): 788-792, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-39023584

ABSTRACT

BACKGROUND AND AIM: Precancer biomarkers help in early detection and management of oral potentially malignant disorders (OPMDs). Interleukin-1ß (IL-1ß), a biomarker, is known to be altered in oral submucous fibrosis (OSMF) and oral leukoplakia (OL). Therefore, we evaluated and compared the serum and salivary IL-1ß levels in patients with OSMF/oral leukoplakia and in gender- and age-matched healthy individuals. MATERIALS AND METHODS: An in vivo, prospective, observational study was conducted on 40 subjects. Subjects were divided into two groups with 20 individuals in each group, that is, Group I: OSMF/oral leukoplakia and Group II: control group. Salivary and serum IL-1ß levels were quantitatively estimated using enzyme-linked immunosorbent assay (ELISA). The statistical tests used were unpaired t-test and Chi-square test. RESULTS: The serum IL-1ß levels were significantly (P 0.001) lesser in Group I in comparison to Group II. The salivary IL-1ß levels remained insignificant between both the groups. However, in both the groups, the salivary IL-1ß levels were significantly higher compared to the serum IL-1ß levels. CONCLUSION: We found that the serum IL-1ß level can be considered as a prospective biomarker for dysplasia, whereas salivary IL-1ß alone needs more elaborated studies to account for its application as a potential biomarker in OPMD.


Subject(s)
Interleukin-1beta , Leukoplakia, Oral , Mouth Neoplasms , Oral Submucous Fibrosis , Precancerous Conditions , Saliva , Humans , Interleukin-1beta/blood , Interleukin-1beta/analysis , Interleukin-1beta/metabolism , Male , Female , Saliva/metabolism , Saliva/chemistry , Leukoplakia, Oral/blood , Leukoplakia, Oral/diagnosis , Leukoplakia, Oral/metabolism , Leukoplakia, Oral/pathology , Prospective Studies , Adult , Middle Aged , Precancerous Conditions/blood , Precancerous Conditions/diagnosis , Precancerous Conditions/pathology , Precancerous Conditions/metabolism , Oral Submucous Fibrosis/blood , Oral Submucous Fibrosis/metabolism , Oral Submucous Fibrosis/diagnosis , Oral Submucous Fibrosis/pathology , Mouth Neoplasms/blood , Mouth Neoplasms/diagnosis , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Case-Control Studies , Biomarkers/blood , Biomarkers/analysis
6.
Sci Rep ; 14(1): 17591, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080384

ABSTRACT

The uncertainty of true labels in medical images hinders diagnosis owing to the variability across professionals when applying deep learning models. We used deep learning to obtain an optimal convolutional neural network (CNN) by adequately annotating data for oral exfoliative cytology considering labels from multiple oral pathologists. Six whole-slide images were processed using QuPath for segmenting them into tiles. The images were labeled by three oral pathologists, resulting in 14,535 images with the corresponding pathologists' annotations. Data from three pathologists who provided the same diagnosis were labeled as ground truth (GT) and used for testing. We investigated six models trained using the annotations of (1) pathologist A, (2) pathologist B, (3) pathologist C, (4) GT, (5) majority voting, and (6) a probabilistic model. We divided the test by cross-validation per slide dataset and examined the classification performance of the CNN with a ResNet50 baseline. Statistical evaluation was performed repeatedly and independently using every slide 10 times as test data. For the area under the curve, three cases showed the highest values (0.861, 0.955, and 0.991) for the probabilistic model. Regarding accuracy, two cases showed the highest values (0.988 and 0.967). For the models using the pathologists and GT annotations, many slides showed very low accuracy and large variations across tests. Hence, the classifier trained with probabilistic labels provided the optimal CNN for oral exfoliative cytology considering diagnoses from multiple pathologists. These results may lead to trusted medical artificial intelligence solutions that reflect diverse diagnoses of various professionals.


Subject(s)
Deep Learning , Neural Networks, Computer , Humans , Cytodiagnosis/methods , Image Processing, Computer-Assisted/methods , Mouth Neoplasms/diagnosis , Mouth Neoplasms/pathology , Pathologists
7.
BMC Oral Health ; 24(1): 773, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987730

ABSTRACT

OBJECTIVE: Resveratrol (Res) is a natural phytoestrogen with antitumor activity. This study sought to investigate the role of Res in ferroptosis in oral squamous cell carcinoma (OSCC). METHODS: Normal human oral keratinocyte (HOK)/oral OSCC (CAL-27/SCC-9) cell lines were treated with different doses of Res. Res toxicity was determined by MTT assay, with half maximal inhibitory concentration values of Res on CAL-27 and SCC-9 cells calculated. Cell viability/colony formation efficiency/migration/invasion/cycle were assessed by CCK-8/colony formation assay/transwell assay/flow cytometry. The expression of p53 protein in the nucleus and cytoplasm, glutathione peroxidase 4 (GPX4) expression, and SLC7A11 messenger RNA (mRNA) and protein expression levels were determined by Western blot and RT-qPCR. Fe2+ content, reactive oxygen species (ROS) level, reduced glutathione (GSH), and lactate dehydrogenase (LDH) release were assessed. RESULTS: Medium- to low-dose Res had no toxic effect on HOK cells, while high-dose Res markedly reduced HOK cell viability. Res significantly suppressed the viability of OSCC cells (CAL-27 and SCC-9). Res inhibited OSCC cell colony formation/migration/invasion, and induced G1 phase arrest. Res caused the translocation of p53 protein to the nucleus, obviously increased Fe2+ content, ROS level and LDH release, decreased GSH content and GPX4 protein expression, and induced ferroptosis. Down-regulation of p53 partially reversed the inhibitory effects of Res on CAL-27 cell malignant behaviors. Res inhibited SLC7A11 transcription by promoting p53 entry into the nucleus. SLC7A11 overexpression negated the the regulatory effects of p53 knockout on the role of Res in OSCC cell malignant behaviors and ferroptosis. CONCLUSION: Res accelerated ferroptosis and inhibited malignant behaviors in OSCC cells by regulating p53/SLC7A11.


Subject(s)
Amino Acid Transport System y+ , Carcinoma, Squamous Cell , Ferroptosis , Mouth Neoplasms , Resveratrol , Tumor Suppressor Protein p53 , Humans , Resveratrol/pharmacology , Resveratrol/therapeutic use , Ferroptosis/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/pathology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Amino Acid Transport System y+/metabolism , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
8.
Biomed Res Int ; 2024: 9543897, 2024.
Article in English | MEDLINE | ID: mdl-39026517

ABSTRACT

Selective neck dissection (SND) is the treatment of choice in patients with oral squamous cell carcinomas (OSCCs) and clinically node-negative necks (cN0). The treatment of patients with positive-staged necks (cN+) includes SND as well as comprehensive neck dissection (CND). The clear benefit of one or the other remains under debate. We aim to address this lack of clarity by analysing patients with OSCC staged with clinically node-positive necks, treated with either CND or SND using a level-by-level approach. This retrospective study included patients diagnosed with OSCC with clinically (cN+) and pathologically (pN+) positive cervical lymph nodes (LNs) with clear neck level categorization during the years 2010-2019. In total, 74 patients were analysed. Cox regression analysis found no significance for the type of ND being an independent risk factor, neither for overall survival (OS) nor for disease-free survival (DFS). Regional recurrence of CND cases (5.77%) was comparable to SND cases (9.09%). For OS, extracapsular spread (ECS) and male sex were identified as independent risk factors with poorer outcome. pT-stage and ECS were found to be independent risk factors for DFS. The results of this study suggest that both CND and SND may be viable treatment options for certain patients with OSCC pN+.


Subject(s)
Carcinoma, Squamous Cell , Lymph Nodes , Lymphatic Metastasis , Mouth Neoplasms , Neck Dissection , Humans , Male , Female , Mouth Neoplasms/surgery , Mouth Neoplasms/pathology , Mouth Neoplasms/mortality , Middle Aged , Neck Dissection/methods , Aged , Retrospective Studies , Adult , Disease-Free Survival , Lymph Nodes/pathology , Lymph Nodes/surgery , Carcinoma, Squamous Cell/surgery , Carcinoma, Squamous Cell/pathology , Treatment Outcome , Neoplasm Recurrence, Local/pathology , Aged, 80 and over , Risk Factors
9.
Can J Dent Hyg ; 58(2): 98-105, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38974821

ABSTRACT

Objective: Oral lichen planus (OLP) is an immune-mediated condition featuring chronic inflammation. The World Health Organization classifies OLP as potentially malignant, but it is believed that the malignant transformation of OLP occurs in lesions with both lichenoid and dysplastic features (LD). This review discusses the issues surrounding OLP and LD, including their malignancy, classification, and categorization, and whether lichenoid inflammation causes dysplastic changes in LD or vice versa. Methods: English full-text literature on OLP, LD and/or dysplasia was retrieved from PubMed, CINAHL, and Google Scholar. Results: Thirty-six publications including original research articles, reviews, meta-analyses, books, reports, letters, and editorials were selected for review. Discussion: Research suggests that OLP has malignant potential, although small, and that LD should not be disregarded, as dysplasia presenting with or without lichenoid features may develop into cancer. There is also disagreement over the classification and categorization of LD. Different terms have been used to classify these lesions, including lichenoid dysplasia, OLP with dysplasia, and dysplasia with lichenoid features. Moreover, in LD, it is not clear if dysplasia or lichenoid infiltration appears first, and if inflammation is a response to dysplasia or if dysplasia is a response to the persistent inflammation. The main limitation in the literature is the inconsistency and subjective nature of histological diagnoses, which can lead to interobserver and intraobserver variation, ultimately resulting in the inaccurate diagnosis of OLP and LD. Conclusion: Although further research is required to understand OLP and LD, both lesions should be considered potentially malignant and should not be disregarded.


Objectif: Le lichen plan buccal (LPB) est une pathologie auto-immune qui se présente sous la forme d'une inflammation chronique. Selon la classification de l'Organisation mondiale de la santé, le LPB est une pathologie potentiellement maligne. Toutefois, on soupçonne que la transformation maligne du LPB se produit dans des lésions présentant à la fois des caractéristiques lichénoïdes et dysplasiques (LD). Cet examen porte sur les questions relatives au LPB et aux LD, notamment leur malignité, leur classification et leur catégorisation, et pour savoir si l'inflammation du lichénoïde entraîne des changements dysplasiques des LD ou vice versa. Méthodes: On a utilisé le texte intégral de documents rédigés en anglais sur le LPB, les LD et la dysplasie issus de PubMed, de CINAHL et de Google Scholar. Résultats: Trente-six publications, notamment des articles sur des études originales, des revues, des méta-analyses, des livres, des rapports, des lettres et des éditoriaux, ont été sélectionnées aux fins d'examen. Discussion: Des études suggèrent que le LPB est potentiellement malin, bien que ce potentiel soit faible, et que les LD ne doivent pas être ignorés : en effet, une dysplasie peut évoluer en cancer, qu'elle présente des caractéristiques lichénoïdes ou non. On constate également un désaccord quant à la classification et à la catégorisation des LD. Différents termes ont été utilisés pour la classification de ces lésions, notamment « dysplasie lichénoïde ¼, « LPB dysplasique ¼ et « dysplasie à caractéristiques lichénoïdes ¼. De plus, dans le cas des LD, on ne sait pas avec certitude si la dysplasie ou l'infiltration lichénoïde apparaît en premier, ni si l'inflammation découle de la dysplasie ou si la dysplasie est une conséquence de l'inflammation persistante. La principale limite de la littérature est due aux incohérences et à la nature subjective des diagnostics histologiques, qui peut entraîner des variations d'un observateur à l'autre ou même avec un même observateur, ce qui entraîne à terme des diagnostics erronés de LPB et de LD. Conclusion: Bien que d'autres études soient nécessaires pour comprendre le LPB et les LD, les lésions de ces 2 catégories doivent être considérées comme potentiellement malignes et ne doivent pas être ignorées.


Subject(s)
Lichen Planus, Oral , Precancerous Conditions , Lichen Planus, Oral/pathology , Lichen Planus, Oral/diagnosis , Lichen Planus, Oral/immunology , Humans , Precancerous Conditions/pathology , Mouth Neoplasms/pathology , Mouth Neoplasms/diagnosis , Cell Transformation, Neoplastic/pathology , Lichenoid Eruptions/pathology , Lichenoid Eruptions/diagnosis
10.
BMC Oral Health ; 24(1): 780, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992585

ABSTRACT

BACKGROUND: This study delves into the intricate landscape of oral cancer, a global concern with a high incidence in Asian countries. We focus on oral squamous cell carcinoma (OSCC), primarily driven by the consumption of betel nut and its derivatives. OSCC often arises from premalignant lesions like oral submucous fibrosis (OSF). In Pakistan, OSCC is prevalent among men due to various addictive substances, including smokeless tobacco and chewing materials. Mutations in tumor suppressor genes, such as TP53 and p21, play crucial roles in this malignancy's development. We also explore the involvement of TUSC3 gene deletion in OSCC and OSF. METHODS: In this study we investigated demographics, TUSC3 gene expression, deletion analysis, and TP53 and p21 genetic alterations in OSCC and OSF patients (blood and tissue of 50 samples in each condition) who had tobacco derivates usage history. The association analysis was carried out mainly through PCR based genotyping. RESULTS: The study's patient cohort (OSCC and OSF) displayed a wide age range from 13 to 65 years (Mean = 32.96 years). Both conditions were more prevalent in males, with a male-female ratio of approximately 2.5:1. Chewing habits analysis revealed high frequencies of gutka use in both OSF and OSCC patients. TUSC3 expression analysis in OSCC cell lines indicated significant downregulation. Genotyping showed no TUSC3 deletion in OSF cases, but a deletion rate of over 22% in OSCC tissue samples. Analysis supported a significant association of TUSC3 deletion with OSCC development but not with OSF. Polymorphism in p53 exon 4 and p21 (rs1801270) were significantly associated with both OSCC and OSF, adding to their pathogenesis. Our findings further revealed a strong correlation between TUSC3 deletion and the excessive use of tobacco and related products, shedding light on the genetic underpinnings of OSCC development. CONCLUSIONS: Notably, our study provides a crucial insight into genetic aspects underlying OSCC and OSF in response of addictive consumption of areca nut, betel quid, and tobacco derivatives. A significant association between TUSC3 deletion and OSCC development, along with polymorphisms in TP53 and p21, underscores the importance of further research into the molecular mechanisms driving oral cancer progression for improved diagnosis and treatment outcomes.


Subject(s)
Carcinoma, Squamous Cell , Cyclin-Dependent Kinase Inhibitor p21 , Membrane Proteins , Mouth Neoplasms , Oral Submucous Fibrosis , Tobacco, Smokeless , Tumor Suppressor Protein p53 , Humans , Male , Oral Submucous Fibrosis/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Female , Adult , Middle Aged , Carcinoma, Squamous Cell/genetics , Pakistan , Aged , Tobacco, Smokeless/adverse effects , Young Adult , Cyclin-Dependent Kinase Inhibitor p21/genetics , Adolescent , Membrane Proteins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Areca/adverse effects , Gene Deletion , Sex Factors
11.
Int J Biol Sci ; 20(9): 3372-3392, 2024.
Article in English | MEDLINE | ID: mdl-38993570

ABSTRACT

Oral squamous cell carcinoma (OSCC) is an aggressive cancer that poses a substantial threat to human life and quality of life globally. Lipid metabolism reprogramming significantly influences tumor development, affecting not only tumor cells but also tumor-associated macrophages (TAMs) infiltration. SOAT1, a critical enzyme in lipid metabolism, holds high prognostic value in various cancers. This study revealed that SOAT1 is highly expressed in OSCC tissues and positively correlated with M2 TAMs infiltration. Increased SOAT1 expression enhanced the capabilities of cell proliferation, tumor sphere formation, migration, and invasion in OSCC cells, upregulated the SREBP1-regulated adipogenic pathway, activated the PI3K/AKT/mTOR pathway and promoted M2-like polarization of TAMs, thereby contributing to OSCC growth both in vitro and in vivo. Additionally, we explored the upstream transcription factors that regulate SOAT1 and discovered that ETS1 positively regulates SOAT1 expression levels. Knockdown of ETS1 effectively inhibited the malignant phenotype of OSCC cells, whereas restoring SOAT1 expression significantly mitigated this suppression. Based on these findings, we suggest that SOAT1 is regulated by ETS1 and plays a pivotal role in the development of OSCC by facilitating lipid metabolism and M2-like polarization of TAMs. We propose that SOAT1 is a promising target for OSCC therapy with tremendous potential.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Proto-Oncogene Protein c-ets-1 , Tumor-Associated Macrophages , Humans , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Tumor-Associated Macrophages/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Protein c-ets-1/genetics , Cell Line, Tumor , Animals , Mice , Cell Proliferation , Gene Expression Regulation, Neoplastic , Male , Cell Movement
12.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000035

ABSTRACT

Alternative splicing dysregulation is an emerging cancer hallmark, potentially serving as a source of novel diagnostic, prognostic, or therapeutic tools. Inhibitors of the activity of the splicing machinery can exert antitumoral effects in cancer cells. We aimed to characterize the splicing machinery (SM) components in oral squamous cell carcinoma (OSCC) and to evaluate the direct impact of the inhibition of SM-activity on OSCC-cells. The expression of 59 SM-components was assessed using a prospective case-control study of tumor and healthy samples from 37 OSCC patients, and the relationship with clinical and histopathological features was assessed. The direct effect of pladienolide-B (SM-inhibitor) on the proliferation rate of primary OSCC cell cultures was evaluated. A significant dysregulation in several SM components was found in OSCC vs. adjacent-healthy tissues [i.e., 12 out of 59 (20%)], and their expression was associated with clinical and histopathological features of less aggressiveness and overall survival. Pladienolide-B treatment significantly decreased OSCC-cell proliferation. Our data reveal a significantly altered expression of several SM-components and link it to pathophysiological features, reinforcing a potential clinical and pathophysiological relevance of the SM dysregulation in OSCC. The inhibition of SM-activity might be a therapeutic avenue in OSCC, offering a clinically relevant opportunity to be explored.


Subject(s)
Carcinoma, Squamous Cell , Cell Proliferation , Mouth Neoplasms , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Male , Female , Middle Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Aged , Gene Expression Regulation, Neoplastic , Macrolides/pharmacology , Alternative Splicing , Epoxy Compounds/pharmacology , Case-Control Studies , Cell Line, Tumor , RNA Splicing , Adult , Prospective Studies
13.
14.
Oral Oncol ; 156: 106946, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002299

ABSTRACT

OBJECTIVES: This study aims to address the critical gap of unavailability of publicly accessible oral cavity image datasets for developing machine learning (ML) and artificial intelligence (AI) technologies for the diagnosis and prognosis of oral cancer (OCA) and oral potentially malignant disorders (OPMD), with a particular focus on the high prevalence and delayed diagnosis in Asia. MATERIALS AND METHODS: Following ethical approval and informed written consent, images of the oral cavity were obtained from mobile phone cameras and clinical data was extracted from hospital records from patients attending to the Dental Teaching Hospital, Peradeniya, Sri Lanka. After data management and hosting, image categorization and annotations were done by clinicians using a custom-made software tool developed by the research team. RESULTS: A dataset comprising 3000 high-quality, anonymized images obtained from 714 patients were classified into four distinct categories: healthy, benign, OPMD, and OCA. Images were annotated with polygonal shaped oral cavity and lesion boundaries. Each image is accompanied by patient metadata, including age, sex, diagnosis, and risk factor profiles such as smoking, alcohol, and betel chewing habits. CONCLUSION: Researchers can utilize the annotated images in the COCO format, along with the patients' metadata, to enhance ML and AI algorithm development.


Subject(s)
Mouth Neoplasms , Humans , Mouth Neoplasms/diagnostic imaging , Mouth Neoplasms/diagnosis , Mouth Neoplasms/pathology , Male , Female , Middle Aged , Adult , Aged , Mouth/pathology , Mouth/diagnostic imaging , Aged, 80 and over , Young Adult , Machine Learning , Adolescent , Artificial Intelligence , Precancerous Conditions/diagnostic imaging , Precancerous Conditions/pathology , Precancerous Conditions/diagnosis
15.
Head Neck Pathol ; 18(1): 62, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958825

ABSTRACT

In 1977, the American Joint Committee on Cancer (AJCC) introduced the inaugural Cancer Staging Manual, which implemented the T (tumor extent), N (regional lymph node status), and M (presence or absence of distant metastasis) staging system. This systematic approach aimed to convey the extent of disease across various cancer types, providing clinicians with a practical framework to plan treatment strategies, predict prognosis, and assess outcomes. The AJCC 8th edition, effective from January 1, 2018, continues this tradition. However, certain shortcomings persist in the AJCC 8th edition, as identified through clinical experience. Specifically, challenges arise in accurately assessing depth of invasion in unique histological variants of oral squamous cell carcinoma (e.g., Oral verrucous carcinoma, Carcinoma cuniculatum, and Papillary squamous cell carcinoma) and minor salivary gland tumors. Additionally, discrepancies exist in the perception of bone invasion patterns and in reporting practices. There is also a need for staging guidelines for malignant odontogenic tumors and multifocal tumors of the oral cavity, supplemented by diagrammatic representations. Lastly, there is a call for comprehensive staging criteria for carcinomas of the ear, external auditory canal, and temporal bone. We advocate for the inclusion of these considerations in future editions of the AJCC Cancer Staging Manual.


Subject(s)
Lip Neoplasms , Mouth Neoplasms , Neoplasm Staging , Humans , Mouth Neoplasms/pathology , Neoplasm Staging/standards , Neoplasm Staging/methods , Lip Neoplasms/pathology
16.
BMC Cancer ; 24(1): 793, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961353

ABSTRACT

BACKGROUND: Accurate regulation of gene expression is crucial for normal development and function of cells. The prognostic significance and potential carcinogenic mechanisms of the related gene JARID2 in OSCC are not yet clear, but existing research has indicated a significant association between the two. METHODS AND MATERIALS: The relationship between the expression of the JARID2 gene in tumor samples of OSCC patients and clinical pathological factors was analyzed using immunohistochemistry experiments and RT-qPCR analysis. Based on the clinical pathological data of patients, bioinformatics analysis was conducted using public databases to determine the function of JARID2 in OSCC. Knockdown OSCC cell lines were constructed, and the impact of JARID2 on the biological behavior of OSCC cell lines was assessed through CCK-8, wound healing assay, and transwell analysis. RESULTS: Immunohistochemistry experiments confirmed the correlation between JARID2 and the prognosis of OSCC patients, while RT-qPCR experiments demonstrated its expression levels in tissue and cells. CKK-8 experiments, wound healing assays, and Transwell experiments indicated that knocking down JARID2 had a negative impact on the proliferation, invasion, and migration of OSCC cells. Bioinformatics analysis results showed that the expression of JARID2 in OSCC is closely associated with patient gene co-expression, gene function enrichment, immune infiltration, and drug sensitivity. CONCLUSION: Our study indicates that JARID2 is a novel prognostic biomarker and potential therapeutic target for OSCC.


Subject(s)
Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Mouth Neoplasms , Neoplasm Invasiveness , Polycomb Repressive Complex 2 , Humans , Mouth Neoplasms/pathology , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Cell Movement/genetics , Prognosis , Cell Line, Tumor , Female , Male , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Middle Aged , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Gene Knockdown Techniques
17.
Shanghai Kou Qiang Yi Xue ; 33(2): 117-122, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-39005085

ABSTRACT

PURPOSE: To prepare single-atom iron nanocatalysts(SAF NCs) and explore its efficacy on oral squamous cell carcinoma Cal 27 cells in vitro, and provide a new strategy for the treatment of oral squamous cell carcinoma. METHODS: SAF NCs were prepared with combination of isolation and pyrolysis, the microscopic characterization was observed by transmission electron microscopy, the morphology and chemical composition were analysed by X-ray diffractograms and elemental distribution energy spectroscopy. The catalytic properties were detected by TMB assay and electron spin resonance test, and finally the changes in the activity of Cal27 cells were observed by CCK-8, flow cytometry and confocal microscopy for in vitro treatment of oral squamous carcinoma, to investigate the therapeutic effect against Cal27 cells. Statistical analysis was performed with GraphPad Prism 9 software package. RESULTS: SAF NCs were successfully synthesized and characterized, which showed excellent catalytic properties at the solution level and good biocompatibility in in vitro cellular level. The viability of Cal27 cell was reduce to 32.08% after in vitro catalytic treatment under conditions mimicking the characteristics of the tumor microenvironment. CONCLUSIONS: Preliminary experiments demonstrated that SAF NCs with good biological properties could effectively inhibit the proliferation of Cal 27 cells in vitro, providing a new strategy for the treatment of oral squamous carcinoma.


Subject(s)
Carcinoma, Squamous Cell , Iron , Mouth Neoplasms , Mouth Neoplasms/pathology , Mouth Neoplasms/drug therapy , Humans , Iron/chemistry , Cell Line, Tumor , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Catalysis , Cell Proliferation/drug effects , Cell Survival/drug effects , Tumor Microenvironment/drug effects
18.
Asian Pac J Cancer Prev ; 25(7): 2265-2269, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39068557

ABSTRACT

INTRODUCTION: Oral squamous cell carcinoma (OSCC) includes about 90% of all oral malignant tumors, and most of them are diagnosed in advanced stages. This study investigated the expression changes of miR-24, miR-200, and miR-34 in saliva samples of patients with oral squamous cell carcinoma, for early diagnosis. METHODS: In this study, 30 patients and 30 healthy individuals were selected. After RNA extraction and cDNA synthesis, the expression levels of miR-24, miR-200, and miR-34 in saliva samples were measured and evaluated using the Real-Time PCR technique. RESULTS: Folding change calculation using 2^(-∆∆ Ct) refers to the relative difference in the expression of the markers of the two groups. The expression level of two biomarkers, miR-200 and miR-34, is decreased in patients compared to healthy people; and the expression level of miR-24 is increased in patients compared to healthy people. CONCLUSION: In general, considering the availability and convenience of saliva sample collection for early detection of the disease, this research result can be considered a diagnostic screening test. To further prove the research results, conducting more extensive studies with more samples is recommended.


Subject(s)
Biomarkers, Tumor , Carcinoma, Squamous Cell , MicroRNAs , Mouth Neoplasms , Saliva , Humans , MicroRNAs/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/diagnosis , Mouth Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/pathology , Saliva/metabolism , Saliva/chemistry , Case-Control Studies , Male , Female , Middle Aged , Prognosis , Early Detection of Cancer/methods , Follow-Up Studies , Adult
19.
Front Biosci (Landmark Ed) ; 29(7): 257, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39082352

ABSTRACT

BACKGROUND: The importance of N6-methyladenosine (m6A) modification in tumorigenesis and progression have been highlighted. This study aimed to investigate the modification of insulin receptor substrate 1 (IRS1) by m6A and its role in oral squamous cell carcinoma (OSCC). METHODS: Bioinformatics was employed to predict differential genes related to epithelial-mesenchymal transition (EMT) in OSCC. Seventeen pairs of OSCC and paracancerous tissue samples were collected. The impact of IRS1 on OSCC cell growth and EMT was evaluated. The fluctuations in IRS1 enrichment and the involvement of p53/Line-1 were investigated. RESULTS: IRS1 was highly expressed in OSCC. IRS1 silencing decreased OSCC cell proliferation and increased apoptosis. IRS1 silencing hindered EMT by regulating related markers. IRS1 silencing upregulated p53 and downregulated Line-1 ORF1p. The p53 inhibition reversed the effects of IRS1 silencing and induced EMT in OSCC cells. Furthermore, the m6A modification of IRS1 was increased in OSCC cells. IRS1 were positively regulated by the m6A regulators methyltransferase-like 14 (METTL14) and YTH domain-containing protein 1 (YTHDC1). IRS1 bound to YTHDC1, and YTHDC1 knockdown inhibited the IRS1 nuclear export. The obesity-associated protein (FTO) negatively regulated IRS1, and FTO overexpression reversed the IRS1-induced OSCC tumor growth. CONCLUSIONS: m6A methylation-mediated IRS1 regulated EMT in OSCC through p53/Line-1. These findings provide potential therapeutic strategies for managing OSCC.


Subject(s)
Adenosine , Carcinoma, Squamous Cell , Cell Proliferation , Epithelial-Mesenchymal Transition , Insulin Receptor Substrate Proteins , Mouth Neoplasms , Signal Transduction , Tumor Suppressor Protein p53 , Insulin Receptor Substrate Proteins/metabolism , Insulin Receptor Substrate Proteins/genetics , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Adenosine/analogs & derivatives , Adenosine/metabolism , Cell Line, Tumor , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Apoptosis/genetics , Animals , Mice , Mice, Nude
20.
Anticancer Res ; 44(8): 3365-3374, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060037

ABSTRACT

BACKGROUND/AIM: Cancer-associated fibroblasts (CAFs) have recently been suggested as critical cellular components of bone invasion in oral squamous cell carcinoma (OSCC). However, the underlying molecular mechanisms and subtypes related to their bone-invasive function are unclear. This study investigated the implications of thymidine phosphorylase (TP)-positive CAFs (TP+CAFs) in OSCC bone invasion. MATERIALS AND METHODS: TP expression was determined in 116 patients with OSCC using immunohistochemistry. The influence of TP expression on the biological behavior of CAFs was investigated in vitro. The possible impact of TP+CAFs on bone invasion in OSCC was further evaluated using patient-derived xenograft (PDX) mouse models. RESULTS: In bone-invasive OSCC tissues, TP+CAFs were mainly distributed on the surface of resorbed bone tissue rather than on the tumor side. High levels of TP+CAFs were significantly associated with higher T-stage, bone invasion, and worse overall survival and recurrence-free survival in our study cohort. Recombinant human TP promoted the proliferative and invasive abilities of CAFs and increased matrix metalloproteinase-9 mRNA expression in vitro, related to bone resorption. In the PDX mouse models, TP+CAFs were found in early bone resorption on the surface of resorbed bony tissues. Bone resorption occurred more frequently in the PDX models with TP+CAFs than in those without. CONCLUSION: TP+CAFs were significantly associated with bone invasion and the prognosis of OSCC. This study provides insights into cellular and molecular targets for the early diagnosis and treatment of bone-invasive OSCC.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Squamous Cell , Mouth Neoplasms , Neoplasm Invasiveness , Thymidine Phosphorylase , Humans , Animals , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/genetics , Female , Male , Mice , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Thymidine Phosphorylase/metabolism , Thymidine Phosphorylase/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/genetics , Middle Aged , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Cell Line, Tumor , Aged , Cell Proliferation , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Prognosis , Xenograft Model Antitumor Assays , Bone Resorption/pathology , Bone Resorption/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL