Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.360
Filter
1.
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
2.
Klin Onkol ; 38(3): 189-201, 2024.
Article in English | MEDLINE | ID: mdl-38960675

ABSTRACT

BACKGROUND: Today, a number of methods and ways of prevention and treatment of radiation- -induced mucositis of the oral cavity and oropharynx have been developed, but the represented approaches are still not effective enough. Therefore, to increase the effectiveness of the prevention and treatment of radiation-induced mucositis, it is necessary to approach this problem comprehensively and individually, and to evaluate the factors affecting the development of mucositis. MATERIALS AND METHODS: In this single-center prospective controlled non-randomized clinical trial, the results of clinical observation of the development of complications of radiation and chemoradiation therapy in 105 patients with a newly diagnosed squamous cell cancer of the oral cavity and oropharynx were analyzed. Factors affecting the risk of the development of grade III radiation-induced mucositis including the age, gender of the patients, their general condition before the treatment according to World Health Organisation scales, type of the treatment and its doses, additional use of immunotherapy with alpha/beta defensins, characteristic signs of the tumor process and all indices of the immune status of the patients before the treatment have been analyzed. RESULTS: The method of construction and analysis of one-factor logistic regression models, where 24 indices were analyzed as factorial features, showed that the reduction of the risk of the development of grade III radiation-induced mucositis is predicted by several factors: immunotherapy, gender, serum concentrations of IgG and IgA. A decrease (P < 0.001) in the risk of the development of grade III radiation-induced mucositis was revealed if immunotherapy with alpha/beta defensins (with a total dose of 40 mg) was included into the treatment scheme (relative odds (RO) 0.05; 95% reference interval (RI) 0.02-0.18), in comparison with patients of the groups where it was not present or this immune agent was used in a total dose of 60 mg (P = 0.001, RO 0.06; 95% RI 0.01-0.30). The next factorial sign was gender, namely the risk of the development of grade III radiation-induced mucositis was lower for men (P = 0.003; RO 0.15; 95% RI 0.04-0.53) compared to women. An increase (P = 0.024) in the risk of the development of grade III radiation-induced mucositis with an increase in the initial level of IgG serum concentration was revealed, (RO 1.08; 95% RI 1.01-1.16) for each 1 mg/mL, as well as an increase (P = 0.044) in the possibility of the appearance of grade III radiation-induced mucositis with an increase in the serum concentration of IgA (RO 1.23; 95% RI 1.01-1.50) for every 1 mg/mL also before the beginning of the treatment. Multifactorial analysis has also confirmed that the risk of the development of grade III radiation-induced mucositis increases (P = 0.008) with a high serum IgG concentration before the treatment or with an increase in this index during therapy (RO 1.13; 95% RI 1.03-1.09) for every 1 mg/mL (when standardized by other risk factors). It was determined that when standardizing according to other factors (gender, IgG level), the risk of the development of grade III radiation-induced mucositis in the use of the immune agent alpha/beta defensins in a total dose of 40 mg per course decreases (P < 0.001; RO 0.08; 95% RI 0.02-0.27) compared to patients with oral cavity and oropharynx cancer who were not treated with immunotherapy. The risk of the development of grade III radiation-induced mucositis also decreases (P = 0.001) in the use of immunotherapy in a higher dose, i.e. 60 mg per course (RO 0.03; 95% RI 0.004-0.24 compared to patients whose treatment did not include immunotherapy (when standardized by other factors). CONCLUSION: As a result of this controlled clinical study, some factors were determined in addition to the radiation as those affecting the risk of the development of grade III radiation-induced mucositis in patients with oral cavity and oropharynx cancer during special treatment. These factors comprise the inclusion of immunotherapy with alpha/beta defensins into the specific treatment; gender, and baseline levels of serum IgG and IgA concentrations suggest a pattern in which the higher the serum IgG and IgA concentrations are before the start of the treatment, the greater is the likelihood of severe radiation-induced mucositis degree during special therapy. The results of the study of humoral state of the immune system in patients with oral cavity and oropharynx cancer before the beginning of chemoradiation therapy can be used as prognostic risk factors for the development of severe gamma-irradiation-induced mucositis of the oropharyngeal area, as well as an indication for the use of immunotherapeutic agents (in particular, alpha/beta defensins) that are able to polarize the immune response towards type 1 T-helpers through their immunomodulatory action.


Subject(s)
Chemoradiotherapy , Mouth Neoplasms , Oropharyngeal Neoplasms , Humans , Oropharyngeal Neoplasms/radiotherapy , Oropharyngeal Neoplasms/therapy , Male , Female , Chemoradiotherapy/adverse effects , Mouth Neoplasms/radiotherapy , Mouth Neoplasms/drug therapy , Risk Factors , Radiation Injuries/etiology , Prospective Studies , Middle Aged , Mucositis/etiology , Carcinoma, Squamous Cell/drug therapy , Aged , Stomatitis/etiology
3.
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
5.
BMC Complement Med Ther ; 24(1): 276, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033112

ABSTRACT

BACKGROUND: Plant-derived compounds have chemopreventive properties to be used as alternative medicine. Pericarp of Mangosteen (Garcinia mangostana Linn.), a tropical fruit in Southeast Asia contains a phytochemical α-mangostin (α-MG) that demonstrates potent anticancer effects against various types of cancer. α-MG has been reported to be the most effective agent in human cancer cell lines. The objectives of this study were to develop oral gel formulations containing α-MG and determine their (1) anticancer activity, (2) anti-HPV-16 and antimicrobial activities, (3) nitric oxide (NO) inhibitory activity, and (4) wound healing effect. METHODS: Formulations of oral gel containing α-MG were developed. Anticancer activity on SCC-25 was assessed. Apoptotic induction was determined using flow cytometry technique. Antiviral activity against HPV-16 pseudovirus and antimicrobial activity against S. mutans, P. gingivalis and C. albicans were investigated. NO inhibition was carried out. Fibroblast cell migration was determined by in vitro scratch assay. RESULTS: The formulation of 1% α-MG in orabase gel demonstrated anticancer activity by promoting apoptosis in SCC-25. The induction of apoptotic activity was dose dependent with pronounced effect in late apoptosis. The formulation appeared to reduce cell viability of oral keratinocytes (OKC). At CC50 it showed an inhibition against HPV-16 pseudovirus infection. The formulation had no antimicrobial activity against S. mutans, P. gingivalis and C. albicans. No significant NO inhibitory activity and wound healing effects were found. CONCLUSIONS: 1% α-MG in orabase gel exhibited anticancer activity by inducing apoptosis although low level of cytotoxicity observed in OKC was present. The appropriate carrier for novel nano-particles targeting cancer cells should be further investigated.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Garcinia mangostana , Gels , Mouth Neoplasms , Xanthones , Xanthones/pharmacology , Humans , Apoptosis/drug effects , Mouth Neoplasms/drug therapy , Garcinia mangostana/chemistry , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Plant Extracts/pharmacology , Plant Extracts/chemistry , Human papillomavirus 16/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
6.
BMC Complement Med Ther ; 24(1): 287, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068492

ABSTRACT

BACKGROUND: Oral squamous cell carcinoma (OSCC) is the most common malignancy of the head and neck. Zeng-Sheng-Ping, composed of Sophora tonkinensis Gagnep., Bistorta officinalis Delarbre, Sonchus arvensis L., Prunella vulgaris L., Dioscorea bulbifera L., and Dictamnus dasycarpus Turcz., was regarded as an anti-cancer drug with significant clinical efficacy, but was discontinued due to liver toxicity. Our research group developed a modified Zeng-Sheng-Ping (ZSP-M) based on original Zeng-Sheng-Ping that exhibited high efficiency and low toxicity in preliminary investigations, although its pharmacodynamic mechanism is still unclear. Here, we aimed to elucidate the pharmacodynamic material basis of ZSP-M and investigate its chemopreventive effect on OSCC by modulating tumor associated macrophages (TAMs). METHODS: Components of ZSP-M were characterized using ultra-performance liquid chromatography-mass spectrometry. Chemopreventive effect induced by ZSP-M against experimental oral cancer was investigated using the 4-nitroquinoline N-oxide precancerous lesion mouse model. RNA sequencing analysis was used to gain a global transcriptional view of the effect of ZSP-M treatment. A cell co-culture model was used to study the targeted effect of ZSP-M on TAMs and the biological properties of OSCC cells and to detect changes in TAM phenotypes. The binding of ZSP-M active compounds to TNF alpha induced protein 6 (TNFAIP6) protein was analyzed by molecular docking and dynamic simulation. RESULTS: Forty main components of ZSP-M were identified, the most abundant of which were flavonoids. ZSP-M inhibited the degree of epithelial dysplasia in precancerous lesions by inhibiting the expression of the TNFAIP6 and CD163 proteins in the precancerous lesions of the tongue. ZSP-M inhibited proliferation, colony formation, migration and invasion of SCC7 cells by targeting TAMs. ZSP-M reduced the expression of CD163+ cells, inhibited the expression of TNFAIP6 protein, Arg1 mRNA and Il10 mRNA in TAMs, and reduced IL-10 cytokine release in the co-culture environment. This effect was maintained after the addition of recombinant TNFAIP6 protein. Computer simulations showed that trifolirhizin and maackiain are well-connected to TNFAIP6. CONCLUSIONS: ZSP-M counteracts the immunosuppressive action of TAMs by specific targeting of TNFAIP6, thereby exerting chemopreventive activity of OSCC.


Subject(s)
Mouth Neoplasms , Tumor-Associated Macrophages , Animals , Mice , Mouth Neoplasms/drug therapy , Tumor-Associated Macrophages/drug effects , Carcinoma, Squamous Cell/drug therapy , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Humans , Cell Line, Tumor , Male , Squamous Cell Carcinoma of Head and Neck/drug therapy
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.
BMC Med ; 22(1): 314, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075531

ABSTRACT

BACKGROUND: Oral squamous cell carcinoma (OSCC) causes significant mortality and morbidity worldwide. Surgical resection with adjuvant radiotherapy remains the standard treatment for locally advanced resectable OSCC. Results from landmark trials have established postoperative concurrent cisplatin-radiotherapy (Cis-RT) as the standard treatment for OSCC patients with high-risk pathologic features. However, cisplatin-related toxicity limits usage in clinical practice. Given the need for effective but less toxic alternatives, we previously conducted a single-arm trial showing favorable safety profiles and promising efficacy of concurrent docetaxel-radiotherapy (Doc-RT). METHODS: In this randomized phase 2 trial, we aimed to compare Doc-RT with the standard Cis-RT in postoperative OSCC patients. Eligible patients had AJCC stage III-IV resectable OSCC with high-risk pathologic features. Two hundred twenty-four patients were enrolled and randomly assigned to receive concurrent Doc-RT or Cis-RT. The primary endpoint was 2-year disease-free survival (DFS). Secondary endpoints included overall survival (OS), locoregional-free survival (LRFS), distant metastasis-free survival (DMFS), and adverse events (AEs). Integrin ß1 (ITGB1) expression was analyzed as a biomarker for efficacy. RESULTS: After a median 28.8-month follow-up, 2-year DFS rates were 63.7% for Doc-RT arm and 56.1% for Cis-RT arm (p = 0.55). Meanwhile, Doc-RT demonstrated comparable efficacy to Cis-RT in OS, LRFS, and DMFS. Doc-RT resulted in fewer grade 3 or 4 hematological AEs. Low ITGB1 was associated with improved Doc-RT efficacy versus Cis-RT. CONCLUSIONS: This randomized trial directly compared Doc-RT with Cis-RT for high-risk postoperative OSCC patients, with comparable efficacy and less toxicity. ITGB1 merits further validation as a predictive biomarker to identify OSCC patients most likely to benefit from Doc-RT. Findings indicate docetaxel may be considered as a concurrent chemoradiation option in this setting. TRIAL REGISTRATION: www. CLINICALTRIALS: gov . NCT02923258 (date of registration: October 4, 2016).


Subject(s)
Cisplatin , Docetaxel , Integrin beta1 , Mouth Neoplasms , Humans , Docetaxel/therapeutic use , Docetaxel/administration & dosage , Female , Male , Middle Aged , Cisplatin/therapeutic use , Cisplatin/administration & dosage , Mouth Neoplasms/drug therapy , Mouth Neoplasms/therapy , Aged , Adult , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/therapy , Biomarkers, Tumor , Antineoplastic Agents/therapeutic use , Treatment Outcome
9.
Chin J Nat Med ; 22(7): 632-642, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39059832

ABSTRACT

Gambogenic acid (GNA), a bioactive compound derived from the resin of Garcinia hanburyi, has demonstrated significant antitumor properties. However, its mechanisms of action in oral squamous cell carcinoma (OSCC) remain largely unclear. This study aimed to elucidate the apoptotic effects of GNA on OSCC cell lines CAL-27 and SCC-15. Our results indicated that GNA induced apoptosis by upregulating the pro-apoptotic protein Noxa. Mechanistic investigations revealed that GNA treatment led to the generation of reactive oxygen species (ROS), which activated endoplasmic reticulum (ER) stress, culminating in cell apoptosis. Inhibition of ROS production and ER stress pathways significantly mitigated GNA-induced Noxa upregulation and subsequent apoptosis. Furthermore, in vivo studies using a murine xenograft model demonstrated that GNA administration effectively inhibited the growth of CAL-27 tumors. Collectively, these findings underscore GNA's potential as a therapeutic agent for the treatment of OSCC.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Endoplasmic Reticulum Stress , Garcinia , Mouth Neoplasms , Proto-Oncogene Proteins c-bcl-2 , Reactive Oxygen Species , Up-Regulation , Xanthenes , Humans , Apoptosis/drug effects , Animals , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Garcinia/chemistry , Cell Line, Tumor , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Xanthenes/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Mice , Up-Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Reactive Oxygen Species/metabolism , Mice, Nude , Mice, Inbred BALB C , Male
10.
Anticancer Res ; 44(8): 3269-3276, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060075

ABSTRACT

BACKGROUND/AIM: How tumors regulate the genes of the coagulome is crucial for cancer-associated thrombosis and the occurrence of venous thromboembolic complications in patients with cancer. We have previously reported potent yet complex effects of glucocorticoids (GC) on the expression of three genes that play a key role in the regulation of thrombin/plasmin activation (F3, PLAU, and SERPINE1). This study aimed to extend the investigation of GC effects to the whole tumor coagulome and assess the resulting impact on the ability of cancer cells to activate thrombin and plasmin. MATERIALS AND METHODS: Cancer RNA expression data were retrieved from various sources. Additionally, oral squamous cell carcinoma (OSCC) cells exposed to GC in vitro were analyzed using QPCR, enzymatic assays measuring thrombin and urokinase-type Plasminogen Activator (uPA) activity, and D-dimer concentrations. RESULTS: Our findings highlight the potent and specific stimulatory effect of GC on SERPINE1 expression across different types of cancer. Consistently, GC were found to inhibit uPA proteolytic activity and reduce the concentrations of D-dimers in OSCC in vitro. CONCLUSION: Fibrinolysis inhibition is a key consequence of cancer cell exposure to GC, possibly leading to the stabilization of the fibrin clot in cancer.


Subject(s)
Fibrinolysis , Glucocorticoids , Plasminogen Activator Inhibitor 1 , Humans , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/genetics , Fibrinolysis/drug effects , Glucocorticoids/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Urokinase-Type Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/genetics , Thrombin/metabolism , Thrombin/pharmacology , Fibrin Fibrinogen Degradation Products/metabolism , Transcriptional Activation/drug effects , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Mouth Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Blood Coagulation/drug effects
11.
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
12.
Molecules ; 29(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38893579

ABSTRACT

The fabrication of zinc oxide-based nanomaterials (including natural and synthetic polymers like sulfated polysaccharide, chitosan, and polymethyl methacrylate) has potential to improve oral cancer treatment strategies. This comprehensive review explores the diverse synthesis methods employed to fabricate zinc oxide nanomaterials tailored for oral cancer applications. Several synthesis processes, particularly sol-gel, hydrothermal, and chemical vapor deposition approaches, are thoroughly studied, highlighting their advantages and limitations. The review also examines how synthesis parameters, such as precursor selection, the reaction temperature, and growth conditions, influence both the physicochemical attributes and biological efficacy of the resulting nanomaterials. Furthermore, recent advancements in surface functionalization and modification strategies targeted at improving the targeting specificity and pharmaceutical effectiveness of zinc oxide-based nanomaterials in oral cancer therapy are elucidated. Additionally, the review provides insights into the existing issues and prospective views in the field, emphasizing the need for further research to optimize synthesis methodologies and elucidate the mechanisms underlying the efficacy of zinc oxide-based nanoparticles in oral cancer therapy.


Subject(s)
Mouth Neoplasms , Nanostructures , Zinc Oxide , Humans , Zinc Oxide/chemistry , Zinc Oxide/chemical synthesis , Mouth Neoplasms/drug therapy , Nanostructures/chemistry , Nanostructures/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Animals
13.
Cell Biochem Funct ; 42(4): e4074, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38874340

ABSTRACT

Usnic acid (UA) is a unique bioactive substance in lichen with potential anticancer properties. Recently, we have reported that UA can reduce 7,12-dimethylbenz[a] anthracene-induced oral carcinogenesis by inhibiting oxidative stress, inflammation, and cell proliferation in a male golden Syrian hamster in vivo model. The present study aims to explore the relevant mechanism of cell death induced by UA on human oral carcinoma (KB) cell line in an in vitro model. We found that UA can induce apoptosis (cell death) in KB cells by decreasing cell viability, increasing the production of reactive oxygen species (ROS), depolarizing mitochondrial membrane potential (MMP) levels, causing nuclear fragmentation, altering apoptotic morphology, and causing excessive DNA damage. Additionally, UA inhibits the expression of Bcl-2, a protein that promotes cell survival, while increasing the expression of p53, Bax, Cytochrome-c, Caspase-9, and 3 proteins in KB cells. UA also inhibits the expression of nuclear factor-κB (NF-κB), a protein that mediates the activation of pro-inflammatory cytokines such as TNF-α and IL-6, in KB cells. Furthermore, UA promotes apoptosis by enhancing the mitochondrial-mediated apoptotic mechanism through oxidative stress, depletion of cellular antioxidants, and an inflammatory response. Ultimately, the findings of this study suggest that UA may have potential as an anticancer therapeutic agent for oral cancer treatments.


Subject(s)
Apoptosis , Benzofurans , Inflammation , Mouth Neoplasms , NF-kappa B , Signal Transduction , Humans , Apoptosis/drug effects , NF-kappa B/metabolism , Benzofurans/pharmacology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Mouth Neoplasms/drug therapy , Signal Transduction/drug effects , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Cell Survival/drug effects , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects
14.
Int Immunopharmacol ; 136: 112377, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38838554

ABSTRACT

The tumor microenvironment (TME) concept has been proposed and is currently being actively studied. The development of extracellular matrix (ECM) in the TME is known as desmoplasia and is observed in many solid tumors. It has also been strongly associated with poor prognosis and resistance to drug therapy. Recently, cellular senescence has gained attention as an effect of drug therapy on cancer cells. Cellular senescence is a phenomenon wherein proliferating cells become resistant to growth-promoting stimuli, secrete the SASP (senescence-associated phenotypic) factors, and stably arrest the cell cycle. These proteins are rich in pro-inflammatory factors, such as interleukin (IL)-6, IL-8, C-X-C motif chemokine ligand 1, C-C motif chemokine ligand (CCL)2, CCL5, and matrix metalloproteinase 3. This study aimed to investigate the desmoplasia-like changes in the TME before and after cancer drug therapy in oral squamous cell carcinomas, evaluate the effect of anticancer drugs on the TME, and the potential involvement of cancer cell senescence. Using a syngeneic oral cancer transplant mouse model, we confirmed that cis-diamminedichloroplatinum (II) (CDDP) administration caused desmoplasia-like changes in cancer tissues. Furthermore, CDDP treatment-induced senescence in tumor-bearing mouse tumor tissues and cultured cancer cells. These results suggest CDDP administration-induced desmoplasia-like structural changes in the TME are related to cellular senescence. Our findings suggest that the administration of anticancer drugs alters the TME of oral cancer cells. Additionally, oral cancer cells undergo senescence, which may influence the TME through the production of SASP factors.


Subject(s)
Antineoplastic Agents , Cellular Senescence , Cisplatin , Mouth Neoplasms , Senescence-Associated Secretory Phenotype , Tumor Microenvironment , Animals , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Cisplatin/pharmacology , Humans , Cellular Senescence/drug effects , Tumor Microenvironment/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Mice , Cell Line, Tumor , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cytokines/metabolism , Male , Female
15.
Med Oncol ; 41(7): 167, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38831079

ABSTRACT

Cancer stem cells (CSCs) are mainly responsible for tumorigenesis, chemoresistance, and cancer recurrence. CSCs growth and progression are regulated by multiple signaling cascades including Wnt/ß-catenin and Hh/GLI-1, which acts independently or via crosstalk. Targeting the crosstalk of signaling pathways would be an effective approach to control the CSC population. Both Wnt/ß-catenin and Hh/GLI-1 signaling cascades are known to be regulated by p53/p21-dependent mechanism. However, it is interesting to delineate whether p21 can induce apoptosis in a p53-independent manner. Therefore, utilizing various subtypes of oral CSCs (SCC9-PEMT p53+/+p21+/+, SCC9-PEMT p53-/-p21+/+, SCC9-PEMT p53+/+p21-/- and SCC9-PEMT p53-/-p21-/-), we have examined the distinct roles of p53 and p21 in Resveratrol nanoparticle (Res-Nano)-mediated apoptosis. It is interesting to see that, besides the p53/p21-mediated mechanism, Res-Nano exposure also significantly induced apoptosis in oral CSCs through a p53-independent activation of p21. Additionally, Res-Nano-induced p21-activation deregulated the ß-catenin-GLI-1 complex and consequently reduced the TCF/LEF and GLI-1 reporter activities. In agreement with in vitro data, similar experimental results were obtained in in vivo mice xenograft model.


Subject(s)
Apoptosis , Cyclin-Dependent Kinase Inhibitor p21 , Mouth Neoplasms , Nanoparticles , Neoplastic Stem Cells , Resveratrol , Tumor Suppressor Protein p53 , Zinc Finger Protein GLI1 , beta Catenin , Apoptosis/drug effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Resveratrol/pharmacology , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , beta Catenin/metabolism , Tumor Suppressor Protein p53/metabolism , Humans , Mouth Neoplasms/pathology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Mice , Cell Line, Tumor , Xenograft Model Antitumor Assays
16.
Int J Pharm ; 660: 124311, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38848798

ABSTRACT

The challenges in treating oral cancer include the limited effectiveness and systemic side effects of conventional chemotherapy and radiation therapy. Hyaluronic acid (HA) based Glycyrrhizin (GL) and Methotrexate (MT) loaded localized delivery systems, specifically nanofiber (NF) based platforms, were developed to address these challenges. The electrospinning method was used for the successful fabrication of a homogenous NF membrane and characterized for morphology, drug entrapment efficiency, tensile strength, and ex-vivo mucoadhesive study. Also, it was evaluated for in-vitro drug release profile, ex-vivo drug permeability, in-vitro anti-inflammatory, apoptosis assay by MTT and flow, and against specific cell lines in order to determine their potential for therapeutic use. Superior tensile breaking force (50 g), mucoadhesive strength of 153 gm/cm2, drug permeability, and releasing properties of designed NF, making them perfect requirements for oral cavity delivery. The anticancer potential of MT in the MTT assay and flow cytometry analysis was significantly increased in oral epidermal carcinoma cell (KB cell) for drug-loaded NF with 63.97 ± 1.99 % apoptosis, at 24 h. With these incorporated, GL with MT in NF had an anti-inflammatory potential, also demonstrated in-vitro and in-vivo. In the Ehrlich Ascites Carcinoma (EAC) induced mice model, the optimal formulation's shows better potential for tumor regression when comparing the developed NF formulation to the drugs. Experimental results show that by lowering mucositis-related inflammation and enhancing the effectiveness of oral cancer treatment, a developed nanofiber-based local drug delivery system offers a feasible strategy for managing oral cancer.


Subject(s)
Apoptosis , Drug Liberation , Glycyrrhizic Acid , Hyaluronic Acid , Methotrexate , Mouth Neoplasms , Nanofibers , Hyaluronic Acid/chemistry , Nanofibers/chemistry , Animals , Methotrexate/administration & dosage , Methotrexate/chemistry , Methotrexate/pharmacology , Mouth Neoplasms/drug therapy , Humans , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/administration & dosage , Cell Line, Tumor , Apoptosis/drug effects , Drug Delivery Systems/methods , Mice , Male , Drug Carriers/chemistry , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology
17.
BMC Oral Health ; 24(1): 715, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907185

ABSTRACT

BACKGROUND: Dental pathogens play a crucial role in oral health issues, including tooth decay, gum disease, and oral infections, and recent research suggests a link between these pathogens and oral cancer initiation and progression. Innovative therapeutic approaches are needed due to antibiotic resistance concerns and treatment limitations. METHODS: We synthesized and analyzed piperine-coated zinc oxide nanoparticles (ZnO-PIP NPs) using UV spectroscopy, SEM, XRD, FTIR, and EDAX. Antioxidant and antimicrobial effectiveness were evaluated through DPPH, ABTS, and MIC assays, while the anticancer properties were assessed on KB oral squamous carcinoma cells. RESULTS: ZnO-PIP NPs exhibited significant antioxidant activity and a MIC of 50 µg/mL against dental pathogens, indicating strong antimicrobial properties. Interaction analysis revealed high binding affinity with dental pathogens. ZnO-PIP NPs showed dose-dependent anticancer activity on KB cells, upregulating apoptotic genes BCL2, BAX, and P53. CONCLUSIONS: This approach offers a multifaceted solution to combatting both oral infections and cancer, showcasing their potential for significant advancement in oral healthcare. It is essential to acknowledge potential limitations and challenges associated with the use of ZnO NPs in clinical applications. These may include concerns regarding nanoparticle toxicity, biocompatibility, and long-term safety. Further research and rigorous testing are warranted to address these issues and ensure the safe and effective translation of ZnO-PIP NPs into clinical practice.


Subject(s)
Alkaloids , Apoptosis , Benzodioxoles , Biofilms , Mouth Neoplasms , Piperidines , Polyunsaturated Alkamides , Zinc Oxide , bcl-2-Associated X Protein , Humans , Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/drug effects , Benzodioxoles/pharmacology , Biofilms/drug effects , Cell Line, Tumor , KB Cells , Metal Nanoparticles/therapeutic use , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Nanoparticles , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/drug effects , X-Ray Diffraction , Zinc Oxide/pharmacology
18.
Discov Med ; 36(185): 1298-1305, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38926116

ABSTRACT

BACKGROUND: Oral squamous cell carcinoma (OSCC) is associated with high recurrence and poor prognosis. Baicalin has multiple pharmacological effects, including anti-inflammatory and anti-proliferative activities. Here, we examine the effect of baicalein on OSCC metastasis and its potential mechanism of action. METHODS: SCC-4 and CAL-27 cells were treated with different concentrations of baicalein. The proliferation of OSCC cells was evaluated by Methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay. As for migration and metastasis, baicalein-treated OSCC cells were used for wound healing assay and Transwell assay. The levels of epithelial-mesenchymal transition-related proteins (E-cadherin, N-cadherin, vimentin) and extracellular regulated protein kinases (ERK)/ETS Transcription Factor ELK1 (ELK-1)/Snail signaling pathway-related proteins in baicalein-treated OSCC cells were evaluated by western blotting. RESULTS: The rates of cell proliferation and migration, along with the metastatic potential, of baicalein-treated cells were significantly lower than those of the control (p < 0.05), and the effects were concentration-dependent. Furthermore, compared to the control, baicalein significantly decreased the levels of N-cadherin and vimentin in SCC-4 and CAL-27 cells, and increased the E-cadherin level (p < 0.05). Mechanistically, baicalein downregulated the levels of p-ERK1/2, phospho-ETS Transcription Factor ELK1 (p-ELK-1), and Snail (p < 0.05). Finally, the ERK/ELK-1/Snail pathway inhibitor (U0126) promoted the effect of baicalein in inhibiting the migration and invasion of OSCC cells (p < 0.05). CONCLUSION: Baicalein abates the migration, invasion, and metastasis of OSCC cells through the ERK/ELK-1/Snail signaling pathway. This study provides a basis for the development of baicalein as a compound for the treatment of OSCC.


Subject(s)
Carcinoma, Squamous Cell , Cell Movement , Cell Proliferation , Flavanones , Mouth Neoplasms , Signal Transduction , Snail Family Transcription Factors , ets-Domain Protein Elk-1 , Flavanones/pharmacology , Flavanones/therapeutic use , Humans , ets-Domain Protein Elk-1/metabolism , Snail Family Transcription Factors/metabolism , Mouth Neoplasms/pathology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Proliferation/drug effects , Signal Transduction/drug effects , Epithelial-Mesenchymal Transition/drug effects , MAP Kinase Signaling System/drug effects , Neoplasm Metastasis , Extracellular Signal-Regulated MAP Kinases/metabolism
19.
Int J Pharm ; 661: 124385, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38925237

ABSTRACT

Curcumin, a polyphenol extracted from turmeric, is a potential alternative for the treatment of oral squamous cell carcinoma (OSCC) due to its remarkable anticancer activity and low systemic toxicity. To further enhance the anticancer activity and bioavailability of curcumin, we synthesized a curcumin analogue, AC17, by modifying the benzene ring and methylene group of curcumin. A soluble hyaluronic acid microneedle patch (AC17@HAMN) was developed to ensure accurate and safe delivery of AC17 to tumor tissues. The inhibitory effect of AC17 on OSCC cells was stronger than that of curcumin and some common analogues. Transcriptome sequencing showed that the target genes of AC17 were mainly concentrated in apoptosis, cell cycle and cell senescence pathways. Among them, AC17 induces cell cycle arrest and inhibits cell proliferation mainly by activating FOXO3 signaling. With good penetration and dissolution properties, microneedles can deliver AC17 directly to the tumor site and show good anti-tumor effect. Moreover, AC17@HAMN showed good biosafety. In summary, AC17@HAMN offers high efficiency, minimal invasiveness, and few adverse reactions. This microneedle patch holds great promise for potential clinical applications, especially for the treatment of OSCC.


Subject(s)
Carcinoma, Squamous Cell , Curcumin , Drug Delivery Systems , Forkhead Box Protein O3 , Mouth Neoplasms , Needles , Curcumin/administration & dosage , Curcumin/pharmacology , Curcumin/pharmacokinetics , Curcumin/chemistry , Mouth Neoplasms/drug therapy , Humans , Animals , Forkhead Box Protein O3/metabolism , Cell Line, Tumor , Carcinoma, Squamous Cell/drug therapy , Cell Proliferation/drug effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Mice, Inbred BALB C , Mice , Mice, Nude , Male
20.
Int Immunopharmacol ; 137: 112480, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38885603

ABSTRACT

OBJECTIVES: We aimed to explore the effects and mechanisms of action of dehydroepiandrosterone (DHEA) on immune evasion of oral squamous cell carcinoma (OSCC) to provide evidence for enhancing the effect of immunotherapy. MATERIALS AND METHODS: A xenograft mouse model and immunohistochemistry were used to reveal the patterns of tumor-infiltrating lymphocytes (TILs). The CAL27 and SCC VII cell lines were used for the in vitro study. Western blotting, qPCR, immunofluorescence, and flow cytometry were used to evaluate the expression of B7-H4. Recombinant mouse B7-H4 protein (rmB7-H4) and PG490, an inhibitor of NF-κB p65 were used for the "rescue study." Gain- and loss-of-function, luciferase reporter, and chromatin immunoprecipitation assays were performed to verify this mechanism. RESULTS: DHEA inhibited tumor growth in an OSCC xenograft mouse model, increased CD8 + cells, and decreased FOXP3 + cells in TILs. DHEA reduced the expression of B7-H4 in CAL27 and SCC VII cells RmB7-H4 reverses the effect of DHEA on tumor growth and TIL patterns. DHEA increased the expression of miR-15b-5p and activated its transcriptional factor NF-κB p65. Further experiments demonstrated that miR-15b-5p inhibited B7-H4 expression by binding to its 3'-UTR regions, and NF-κB p65 activated miR-15b transcription. PG490 reversed the effects of DHEA on tumor growth, antitumor immunity in the OSCC xenograft model, and the expression/phosphorylation of NF-κB p65, miR-15b-5p, and B7-H4. CONCLUSIONS: This study indicates that DHEA attenuates the immune escape of OSCC cells by inhibiting B7-H4 expression, providing new insights for cancer immunotherapy.


Subject(s)
Carcinoma, Squamous Cell , Dehydroepiandrosterone , MicroRNAs , Mouth Neoplasms , Transcription Factor RelA , Tumor Escape , V-Set Domain-Containing T-Cell Activation Inhibitor 1 , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Mouth Neoplasms/immunology , Mouth Neoplasms/drug therapy , Humans , Transcription Factor RelA/metabolism , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Tumor Escape/drug effects , Cell Line, Tumor , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism , Mice , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/drug therapy , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic/drug effects , Signal Transduction/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Mice, Inbred BALB C , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL