Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.326
Filter
1.
J Evid Based Dent Pract ; 24(2): 101899, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821659

ABSTRACT

OBJECTIVES: Despite phototherapy (in the form of photodynamic therapy (PDT)-mediated oxidative stress) being utilized in the management of oral potentially malignant disorders (OPMDs), the evidence of certainty remains unclear. Hence, this systematic review and meta-analysis (PROSPERO # CRD42021218748) is aimed to evaluate the clinical efficacy of PDT-induced oxidative stress in OPMDs METHODS: PubMed, Embase, Web of Science, Scopus, and Cochrane Library databases were searched without restriction of language or year of publication. In addition, gray literature was searched and a manual search was performed. Two independent reviewers screened all the studies, assessing data extraction, risk of bias and certainty of evidence. A narrative synthesis was carried out. For the meta-analysis, random effects were considered to determine the prevalence of a total and a partial remission (PR) of oral potentially malignant disorders (OPMDs). The certainty of evidence was explored using the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) approach. RESULTS: Twenty-three studies were included in the qualitative and quantitative syntheses. A total of 880 patients were included (564 males; 218 females) with an age range between 24 and 89-years-old. The results showed the prevalence of the total and partial remissions respectively for the following OPMLs: actinic cheilitis (AC): 69.9% and 2.4%; oral leukoplakia (OL): 44% and 36.9%; oral verrucous hyperplasia (OVH): 98.5%; oral erythroleukoplakia (OEL): 92.1% and 7.9%. The prevalence of no remission of OL was 18.8%. CONCLUSIONS: PDT demonstrated significant results in clinical remission of OPMDs and most of the eligible studies have shown a total or a partial remission of the included lesions, but at a low or a very low certainty of evidence. Hence, further clinical studies with robust methodology are warranted to offer further validated data. Also, further evidence is required to understand further the mechanism of PDT-induced oxidative stress.


Subject(s)
Mouth Neoplasms , Photochemotherapy , Humans , Photochemotherapy/methods , Mouth Neoplasms/drug therapy , Precancerous Conditions/drug therapy , Oxidative Stress , Female , Treatment Outcome , Male , Photosensitizing Agents/therapeutic use , Cheilitis/drug therapy , Aged , Aged, 80 and over
2.
Anticancer Res ; 44(6): 2377-2392, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821582

ABSTRACT

BACKGROUND/AIM: Oral squamous cell carcinoma (OSCC) is characterized by early metastasis, clinical resistance and poor prognosis. Recently, we showed that aggressive OSCC cells co-express endothelial cell markers and can form tube-like structures, known as vasculogenic mimicry (VM), a process associated with poor prognosis in head and neck cancers. Given the limited success of current antiangiogenic therapy in treating OSCC, this study sought to explore the efficiency of these drugs in targeting an ex vivo model of VM. MATERIALS AND METHODS: OSCC cell lines from the tongue and floor of the mouth in addition to human endothelial cells were used. The treatments comprised a set of clinically relevant antiangiogenic drugs: sorafenib, sunitinib, and axitinib, which were administered in different doses. Multiple ex vivo approaches including cell tubulogenesis, proliferation, apoptosis, and migration assays were used. RESULTS: Although these drugs inhibited the formation of endothelial cell capillaries, they showed clear differential effects on OSCC cell-derived VM and cell morphology. Sorafenib inhibited the tubulogenesis of aggressive OSCC cells compared with the limited effect of sunitinib and axitinib. Furthermore, our data consistently demonstrated a preferential efficacy of certain drugs over others. Sorafenib and sunitinib exhibited anti-cancer effects on tumor cell proliferation, apoptosis, and cell migration, compared with the limited effect of axitinib. CONCLUSION: The antiangiogenic drugs, except sorafenib, had limited effect on VM formation in vitro and exhibited varying anti-cancer effects on OSCC cells. These data support the notion that VM formation may in part explain the development of drug resistance in OSCC cells.


Subject(s)
Angiogenesis Inhibitors , Axitinib , Cell Movement , Cell Proliferation , Mouth Neoplasms , Neovascularization, Pathologic , Sorafenib , Sunitinib , Humans , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Mouth Neoplasms/blood supply , Mouth Neoplasms/metabolism , Cell Line, Tumor , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Sorafenib/pharmacology , Sorafenib/therapeutic use , Sunitinib/pharmacology , Sunitinib/therapeutic use , Cell Proliferation/drug effects , Cell Movement/drug effects , Axitinib/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/metabolism , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Niacinamide/therapeutic use
3.
Int J Med Sci ; 21(6): 1165-1175, 2024.
Article in English | MEDLINE | ID: mdl-38774756

ABSTRACT

Oral cancer is the most heterogeneous cancer at clinical and histological levels. PI3K/AKT/mTOR pathway was identified as one of the most commonly modulated signals in oral cancer, which regulates major cellular and metabolic activity of the cell. Thus, various proteins of PI3K/AKT/mTOR pathway were used as therapeutic targets for oral cancer, to design more specific drugs with less off-target toxicity. This review sheds light on the regulation of PI3K/AKT/mTOR, and its role in controlling autophagy and associated apoptosis during the progression and metastasis of oral squamous type of malignancy (OSCC). In addition, we reviewed in detail the upstream activators and the downstream effectors of PI3K/AKT/mTOR signaling as potential therapeutic targets for oral cancer treatment.


Subject(s)
Autophagy , Mouth Neoplasms , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Humans , Autophagy/physiology , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics
4.
J Evid Based Integr Med ; 29: 2515690X241258369, 2024.
Article in English | MEDLINE | ID: mdl-38778767

ABSTRACT

The aim of this study was to evaluate the effect of curcumin combined with Lactobacillus rhamnosus GG cell-free supernatant (LGG CFS) on the proliferation and induction of apoptosis in SCC-9 oral squamous cell carcinoma (OSCC) cells. Curcumin (40 µg/ml) and 25% v/v LGG CFS (108 CFU/ml), both alone and in a combination regimen, significantly decreased the viability of SCC-9 cells and normal human gingival fibroblast (HGF) cells. Interestingly, the combination of low doses of curcumin (5 µg/ml) and 25% v/v LGG CFS (106 CFU/ml) had no effect on the HGF cells but significantly inhibited the viability of SCC-9 cells (p < 0.05). Flow cytometric analysis revealed that SCC-9 cells treated with the combination of low-dose curcumin and low-dose LGG CFS had a higher apoptotic rate than the cells in the control group and the single treatment groups (p < 0.05). The combined treatment also significantly increased the Bax/Bcl2 mRNA and protein expression in SCC-9 cells (p < 0.05) but not in HGF cells, indicating the underlying mechanism of the combination regimen. There was no significant difference in caspase-3 protein expression or the Bcl-xL/Bak and Mcl-1/Bak ratios between the treatment and control groups in both cell lines. These findings suggested that the coadministration of curcumin and LGG could exhibit anticancer effects in SCC-9 cells without causing toxicity to normal fibroblast cells.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Cell Survival , Curcumin , Lacticaseibacillus rhamnosus , Mouth Neoplasms , Humans , Curcumin/pharmacology , Apoptosis/drug effects , Mouth Neoplasms/drug therapy , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Fibroblasts/drug effects , Cell Proliferation/drug effects , Gingiva/cytology , Probiotics/pharmacology , Antineoplastic Agents/pharmacology
5.
Biomolecules ; 14(5)2024 May 18.
Article in English | MEDLINE | ID: mdl-38786003

ABSTRACT

Oral squamous cell carcinoma (OSCC) stands as a prevalent subtype of head and neck squamous cell carcinoma, leading to disease recurrence and low survival rates. PPARγ, a ligand-dependent nuclear transcription factor, holds significance in tumor development. However, the role of PPARγ in the development of OSCC has not been fully elucidated. Through transcriptome sequencing analysis, we discovered a notable enrichment of ferroptosis-related molecules upon treatment with PPARγ antagonist. We subsequently confirmed the occurrence of ferroptosis through transmission electron microscopy, iron detection, etc. Notably, ferroptosis inhibitors could not completely rescue the cell death caused by PPARγ inhibitors, and the rescue effect was the greatest when disulfidptosis and ferroptosis inhibitors coexisted. We confirmed that the disulfidptosis phenotype indeed existed. Mechanistically, through qPCR and Western blotting, we observed that the inhibition of PPARγ resulted in the upregulation of heme oxygenase 1 (HMOX1), thereby promoting ferroptosis, while solute carrier family 7 member 11 (SLC7A11) was also upregulated to promote disulfidptosis in OSCC. Finally, a flow cytometry analysis of flight and multiplex immunohistochemical staining was used to characterize the immune status of PPARγ antagonist-treated OSCC tissues in a mouse tongue orthotopic transplantation tumor model, and the results showed that the inhibition of PPARγ led to ferroptosis and disulfidptosis, promoted the aggregation of cDCs and CD8+ T cells, and inhibited the progression of OSCC. Overall, our findings reveal that PPARγ plays a key role in regulating cell death in OSCC and that targeting PPARγ may be a potential therapeutic approach for OSCC.


Subject(s)
Ferroptosis , PPAR gamma , Ferroptosis/drug effects , Animals , PPAR gamma/metabolism , PPAR gamma/antagonists & inhibitors , Humans , Mice , Cell Line, Tumor , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/genetics , Heme Oxygenase-1/metabolism , Antineoplastic Agents/pharmacology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/genetics , Gene Expression Regulation, Neoplastic/drug effects
6.
Biomed Pharmacother ; 175: 116654, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692066

ABSTRACT

This paper explores the therapeutic perspectives of polyphenols and chitosan as potential anticancer agents in the mouthwash formulations. Taking into account the high incidence of squamous cell carcinoma (SCC) among oral cancers, this discussion will concentrate on the potential advantages of these compounds in oral care, focusing on their impact on improving oral health and cancer prevention. According to the data, it appears that the mixture of BACs extract and chitosan may increase the efficiency of the apoptosis of cancer cells while reducing the undesired side effects. The cytotoxicity assays demonstrate a significant reduction in squamous carcinoma cell viability after incubation with BACs extract, with a marked decrease observed over 24-72 hours up to 76%. The anti-cancer properties of the BAC extract are related to luteolin, which is a predominant compound. The addition of 0.025% chitosan reduced the metabolic activity of cancer cells by 37.5%, suggesting a synergistic interaction between the compounds. This research highlights the potential of BACs and chitosan in modulating important molecular targets associated with cancer cell.


Subject(s)
Chitosan , Mouth Neoplasms , Mouthwashes , Oral Health , Polyphenols , Chitosan/chemistry , Chitosan/pharmacology , Humans , Polyphenols/pharmacology , Mouthwashes/pharmacology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/prevention & control , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/prevention & control , Carcinoma, Squamous Cell/pathology , Drug Compounding
8.
Int J Oral Sci ; 16(1): 35, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719825

ABSTRACT

The efficient clinical treatment of oral squamous cell carcinoma (OSCC) is still a challenge that demands the development of effective new drugs. Phenformin has been shown to produce more potent anti-tumor activities than metformin on different tumors, however, not much is known about the influence of phenformin on OSCC cells. We found that phenformin suppresses OSCC cell proliferation, and promotes OSCC cell autophagy and apoptosis to significantly inhibit OSCC cell growth both in vivo and in vitro. RNA-seq analysis revealed that autophagy pathways were the main targets of phenformin and identified two new targets DDIT4 (DNA damage inducible transcript 4) and NIBAN1 (niban apoptosis regulator 1). We found that phenformin significantly induces the expression of both DDIT4 and NIBAN1 to promote OSCC autophagy. Further, the enhanced expression of DDIT4 and NIBAN1 elicited by phenformin was not blocked by the knockdown of AMPK but was suppressed by the knockdown of transcription factor ATF4 (activation transcription factor 4), which was induced by phenformin treatment in OSCC cells. Mechanistically, these results revealed that phenformin triggers endoplasmic reticulum (ER) stress to activate PERK (protein kinase R-like ER kinase), which phosphorylates the transitional initial factor eIF2, and the increased phosphorylation of eIF2 leads to the increased translation of ATF4. In summary, we discovered that phenformin induces its new targets DDIT4 and especially NIBAN1 to promote autophagic and apoptotic cell death to suppress OSCC cell growth. Our study supports the potential clinical utility of phenformin for OSCC treatment in the future.


Subject(s)
Autophagy , Carcinoma, Squamous Cell , Cell Proliferation , Endoplasmic Reticulum Stress , Mouth Neoplasms , Phenformin , Transcription Factors , Phenformin/pharmacology , Endoplasmic Reticulum Stress/drug effects , Humans , Mouth Neoplasms/drug therapy , Autophagy/drug effects , Carcinoma, Squamous Cell/drug therapy , Cell Proliferation/drug effects , Cell Line, Tumor , Transcription Factors/metabolism , Transcription Factors/drug effects , Mice , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Blotting, Western
9.
Sci Rep ; 14(1): 10616, 2024 05 09.
Article in English | MEDLINE | ID: mdl-38720012

ABSTRACT

Oral cancer stands as a prevalent maligancy worldwide; however, its therapeutic potential is limited by undesired effects and complications. As a medicinal edible fungus, Chaga mushroom (Inonotus obliquus) exhibits anticancer effects across diverse cancers. Yet, the precise mechanisms underlying its efficacy remain unclear. We explored the detailed mechanisms underlying the anticancer action of Chaga mushroom extract in oral cancer cells (HSC-4). Following treatment with Chaga mushroom extracts, we analyzed cell viability, proliferation capacity, glycolysis, mitochondrial respiration, and apoptosis. Our findings revealed that the extract reduced cell viability and proliferation of HSC-4 cells while arresting their cell cycle via suppression of STAT3 activity. Regarding energy metabolism, Chaga mushroom extract inhibited glycolysis and mitochondrial membrane potential in HSC-4 cells, thereby triggering autophagy-mediated apoptotic cell death through activation of the p38 MAPK and NF-κB signaling pathways. Our results indicate that Chaga mushroom extract impedes oral cancer cell progression, by inhibiting cell cycle and proliferation, suppressing cancer cell energy metabolism, and promoting autophagy-mediated apoptotic cell death. These findings suggest that this extract is a promising supplementary medicine for the treatment of patients with oral cancer.


Subject(s)
Apoptosis , Autophagy , Cell Proliferation , Energy Metabolism , Mouth Neoplasms , Humans , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Energy Metabolism/drug effects , Cell Proliferation/drug effects , Cell Line, Tumor , Apoptosis/drug effects , Autophagy/drug effects , Inonotus/chemistry , Cell Survival/drug effects , Membrane Potential, Mitochondrial/drug effects , Glycolysis/drug effects , Signal Transduction/drug effects , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Agaricales/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Cell Cycle/drug effects
10.
Eur J Med Chem ; 272: 116508, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38761583

ABSTRACT

The traditional clinical approaches for oral cancer consist of surgery, chemotherapy, radiotherapy, immunotherapy, and so on. However, these treatments often induce side effects and exhibit limited efficacy. Photothermal therapy (PTT) emerges as a promising adjuvant treatment, utilizing photothermal agents (PTAs) to convert light energy into heat for tumor ablation. Another innovative approach, photodynamic therapy (PDT), leverages photosensitizers (PSs) and specific wavelength laser irradiation to generate reactive oxygen species (ROS), offering an effective and non-toxic alternative. The relevant combination therapies have been reported in the field of oral cancer. Simultaneously, the advancement of nanomaterials has propelled the clinical application of PTT and PDT. Therefore, a comprehensive understanding of PTT and PDT is required for better application in oral cancer treatment. Here, we review the use of PTT and PDT in oral cancer, including noble metal materials (e.g., Au nanoparticles), carbon materials (e.g., graphene oxide), organic dye molecules (e.g., indocyanine green), organic molecule-based agents (e.g., porphyrin-analog phthalocyanine) and other inorganic materials (e.g., MXenes), exemplify the advantages and disadvantages of common PTAs and PSs, and summarize the combination therapies of PTT with PDT, PTT/PDT with chemotherapy, PTT with radiotherapy, PTT/PDT with immunotherapy, and PTT/PDT with gene therapy in the treatment of oral cancer. The challenges related to the PTT/PDT combination therapy and potential solutions are also discussed.


Subject(s)
Mouth Neoplasms , Nanostructures , Photochemotherapy , Photosensitizing Agents , Humans , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Nanostructures/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Photothermal Therapy , Animals
11.
Phytomedicine ; 129: 155511, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38723523

ABSTRACT

BACKGROUND: Mitochondrial dysfunction associated with mitochondrial DNA mutations, enzyme defects, generation of ROS, and altered oxidative homeostasis is known to induce oral carcinogenesis during exposure to arecoline. Butein, a natural small molecule from Butea monosperma, possesses anti-inflammatory, anti-diabetic, and anti-cancer effects. However, the role of butein in the mitochondrial quality control mechanism has not been illuminated clearly. PURPOSE: This study aimed to explore the role of butein in preserving mitochondrial quality control during arecoline-induced mitochondrial dysfunction in oral cancer to curtail the early onset of carcinogenesis. METHODS: Cell viability was evaluated by MTT assay. The relative protein expressions were determined by western blotting. Immunofluorescence and confocal imaging were used to analyze the relative fluorescence and co-localization of proteins. Respective siRNAs were used to examine the knockdown-based studies. RESULTS: Butein, in the presence of arecoline, significantly caused a decrease in mitochondrial hyperpolarization and ROS levels in oral cancer cells. Mechanistically, we found an increase in COXIV, TOM20, and PGC1α expression during butein treatment, and inhibition of PGC1α blunted mitochondrial biogenesis and decreased the mitochondrial pool. Moreover, the fission protein MTP18, and its molecular partners DRP1 and MFF were dose-dependently increased during butein treatment to maintain mitochondria mass. In addition, we also found increased expression of various mitophagy proteins, including PINK1, Parkin, and LC3 during butein treatment, suggesting the clearance of damaged mitochondria to maintain a healthy mitochondrial pool. Interestingly, butein increased the activity of SIRT1 to enhance the functional mitochondrial pool, and inhibition of SIRT1 found to reduce the mitochondrial levels, as evident from the decrease in the expression of PGC1α and MTP18 in oral cancer cells. CONCLUSION: Our study proved that SIRT1 maintains a functional mitochondrial pool through PGC1α and MTP18 for biogenesis and fission of mitochondria during arecoline exposure and could decrease the risk of mitochondria dysfunctionality associated with the onset of oral carcinogenesis.


Subject(s)
Arecoline , Chalcones , Mitochondria , Mouth Neoplasms , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Oxygen Species , Sirtuin 1 , Humans , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Mouth Neoplasms/chemically induced , Mouth Neoplasms/drug therapy , Mitochondria/drug effects , Mitochondria/metabolism , Arecoline/pharmacology , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Chalcones/pharmacology , Sirtuin 1/metabolism , Cell Survival/drug effects
12.
BMC Cancer ; 24(1): 564, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711026

ABSTRACT

BACKGROUND: 5-Fluorouracil (5FU) is a primary chemotherapeutic agent used to treat oral squamous cell carcinoma (OSCC). However, the development of drug resistance has significantly limited its clinical application. Therefore, there is an urgent need to determine the mechanisms underlying drug resistance and identify effective targets. In recent years, the Wingless and Int-1 (WNT) signaling pathway has been increasingly studied in cancer drug resistance; however, the role of WNT3, a ligand of the canonical WNT signaling pathway, in OSCC 5FU-resistance is not clear. This study delved into this potential connection. METHODS: 5FU-resistant cell lines were established by gradually elevating the drug concentration in the culture medium. Differential gene expressions between parental and resistant cells underwent RNA sequencing analysis, which was then substantiated via Real-time quantitative PCR (RT-qPCR) and western blot tests. The influence of the WNT signaling on OSCC chemoresistance was ascertained through WNT3 knockdown or overexpression. The WNT inhibitor methyl 3-benzoate (MSAB) was probed for its capacity to boost 5FU efficacy. RESULTS: In this study, the WNT/ß-catenin signaling pathway was notably activated in 5FU-resistant OSCC cell lines, which was confirmed through transcriptome sequencing analysis, RT-qPCR, and western blot verification. Additionally, the key ligand responsible for pathway activation, WNT3, was identified. By knocking down WNT3 in resistant cells or overexpressing WNT3 in parental cells, we found that WNT3 promoted 5FU-resistance in OSCC. In addition, the WNT inhibitor MSAB reversed 5FU-resistance in OSCC cells. CONCLUSIONS: These data underscored the activation of the WNT/ß-catenin signaling pathway in resistant cells and identified the promoting effect of WNT3 upregulation on 5FU-resistance in oral squamous carcinoma. This may provide a new therapeutic strategy for reversing 5FU-resistance in OSCC cells.


Subject(s)
Drug Resistance, Neoplasm , Fluorouracil , Mouth Neoplasms , Wnt Signaling Pathway , Wnt3 Protein , Humans , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Drug Resistance, Neoplasm/genetics , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Wnt Signaling Pathway/drug effects , Cell Line, Tumor , Wnt3 Protein/metabolism , Wnt3 Protein/genetics , beta Catenin/metabolism , beta Catenin/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic/drug effects , Antimetabolites, Antineoplastic/pharmacology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
13.
Can Vet J ; 65(4): 343-350, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38562982

ABSTRACT

Background: Effective treatment for canine oral malignant melanoma (e.g., curative-intent surgery) may not be feasible or radiation therapy may be unavailable. However, chemotherapy is usually an option, and more information is needed regarding its use without adequate local treatments. Objective: Our objective was to investigate the efficacy of chemotherapy in canine oral malignant melanoma without adequate local control, using carboplatin with dose reduction in small-breed dogs and metronomic chemotherapy. Animals and procedure: Client-owned dogs with histopathologically diagnosed oral malignant melanoma were retrospectively enrolled from 2016 to 2022. The chemotherapy protocol in each case was determined by the attending clinician. Results: Thirteen dogs were included. The median progression-free interval of all 13 dogs was 42 d (14 to 953 d). The median overall survival time of dogs with chemotherapy as their only systemic treatment was 181 d (50 to 960 d; n = 11). The median dosage of carboplatin was 250 mg/m2. Response to treatment and clinical stage were significant prognostic factors. Conclusion and clinical relevance: As chemotherapy provided a median survival of 6 mo, it could be considered when adequate local control is infeasible. Earlier clinical stages or achievement of at least stable disease during chemotherapy may indicate better survival in dogs.


Une étude rétrospective de l'effet chimiothérapeutique sur le mélanome malin buccal canin dépourvu de chirurgie et de radiothérapie á large marge : le stade clinique et la réponse au traitement prédisent les résultats du patient. Mise en contexte: Des traitements efficaces pour le mélanome malin oral canin, tels que la chirurgie á visée curative, ne sont parfois pas réalisables ou la radiothérapie n'est pas disponible dans certaines régions. La chimiothérapie reste une option de traitement et davantage d'informations devraient être fournies pour les cas qui n'ont pas eu accés á un traitement local adéquat. Objectif: Cette étude visait á étudier l'efficacité de la chimiothérapie dans le mélanome malin oral canin sans contrôle local adéquat, en utilisant le carboplatine avec réduction de dose chez les chiens de petite race et la chimiothérapie métronomique. Animaux et procédure: Treize chiens appartenant á des clients atteints d'un mélanome malin oral diagnostiqué par histopathologie ont été rétrospectivement inscrits de 2016 á 2022. Le protocole de chimiothérapie a été déterminé par le clinicien traitant. Résultats: L'intervalle médian sans progression des treize chiens était de 42 jours (14­953 jours). La durée médiane de survie globale des chiens ayant reçu une chimiothérapie comme seul traitement systémique était de 181 jours (50­960 jours; n = 11). La dose médiane de carboplatine était de 250 mg/m2. La réponse au traitement et le stade clinique étaient des facteurs pronostiques importants. Conclusion et pertinence clinique: La chimiothérapie pouvait encore être envisagée lorsqu'un contrôle local adéquat était impossible. Des stades cliniques plus précoces ou des patients atteignant au moins une maladie stable pendant la chimiothérapie peuvent indiquer une meilleure survie.(Traduit par les auteurs).


Subject(s)
Antineoplastic Agents , Dog Diseases , Melanoma , Mouth Neoplasms , Skin Neoplasms , Humans , Dogs , Animals , Melanoma/drug therapy , Melanoma/radiotherapy , Melanoma/veterinary , Carboplatin/therapeutic use , Retrospective Studies , Antineoplastic Agents/therapeutic use , Mouth Neoplasms/drug therapy , Mouth Neoplasms/radiotherapy , Mouth Neoplasms/surgery , Mouth Neoplasms/veterinary , Dog Diseases/drug therapy , Dog Diseases/radiotherapy , Dog Diseases/surgery , Skin Neoplasms/veterinary
14.
Clin Epigenetics ; 16(1): 54, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38600608

ABSTRACT

The polycomb group (PcG) comprises a set of proteins that exert epigenetic regulatory effects and play crucial roles in diverse biological processes, ranging from pluripotency and development to carcinogenesis. Among these proteins, enhancer of zeste homolog 2 (EZH2) stands out as a catalytic component of polycomb repressive complex 2 (PRC2), which plays a role in regulating the expression of homologous (Hox) genes and initial stages of x chromosome inactivation. In numerous human cancers, including head and neck squamous cell carcinoma (HNSCC), EZH2 is frequently overexpressed or activated and has been identified as a negative prognostic factor. Notably, EZH2 emerges as a significant gene involved in regulating the STAT3/HOTAIR axis, influencing HNSCC proliferation, differentiation, and promoting metastasis by modulating related oncogenes in oral cancer. Currently, various small molecule compounds have been developed as inhibitors specifically targeting EZH2 and have gained approval for treating refractory tumors. In this review, we delve into the epigenetic regulation mediated by EZH2/PRC2 in HNSCC, with a specific focus on exploring the potential roles and mechanisms of EZH2, its crucial contribution to targeted drug therapy, and its association with cancer markers and epithelial-mesenchymal transition. Furthermore, we aim to unravel its potential as a therapeutic strategy for oral squamous cell carcinoma.


Subject(s)
Enhancer of Zeste Homolog 2 Protein , Squamous Cell Carcinoma of Head and Neck , Humans , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , DNA Methylation , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Head and Neck Neoplasms/drug therapy , Mouth Neoplasms/drug therapy , Polycomb Repressive Complex 2/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy
15.
J Nanobiotechnology ; 22(1): 168, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38610015

ABSTRACT

BACKGROUND: Oral cancer is the most common malignant tumor of the head and neck, and 90% of cases are oral squamous cell carcinoma (OSCC). Chemotherapy is an important component of comprehensive treatment for OSCC. However, the clinical treatment effect of chemotherapy drugs, such as doxorubicin (DOX), is limited due to the lack of tumor targeting and rapid clearance by the immune system. Thus, based on the tumor-targeting and immune evasion abilities of macrophages, macrophage membrane-encapsulated poly(methyl vinyl ether alt maleic anhydride)-phenylboronic acid-doxorubicin nanoparticles (MM@PMVEMA-PBA-DOX NPs), briefly as MM@DOX NPs, were designed to target OSCC. The boronate ester bonds between PBA and DOX responded to the low pH value in the tumor microenvironment, selectively releasing the loaded DOX. RESULTS: The results showed that MM@DOX NPs exhibited uniform particle size and typical core-shell structure. As the pH decreased from 7.4 to 5.5, drug release increased from 14 to 21%. The in vitro targeting ability, immune evasion ability, and cytotoxicity of MM@DOX NPs were verified in HN6 and SCC15 cell lines. Compared to free DOX, flow cytometry and fluorescence images demonstrated higher uptake of MM@DOX NPs by tumor cells and lower uptake by macrophages. Cell toxicity and live/dead staining experiments showed that MM@DOX NPs exhibited stronger in vitro antitumor effects than free DOX. The targeting and therapeutic effects were further confirmed in vivo. Based on in vivo biodistribution of the nanoparticles, the accumulation of MM@DOX NPs at the tumor site was increased. The pharmacokinetic results demonstrated a longer half-life of 9.26 h for MM@DOX NPs compared to 1.94 h for free DOX. Moreover, MM@DOX NPs exhibited stronger tumor suppression effects in HN6 tumor-bearing mice and good biocompatibility. CONCLUSIONS: Therefore, MM@DOX NPs is a safe and efficient therapeutic platform for OSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Animals , Mice , Carcinoma, Squamous Cell/drug therapy , Squamous Cell Carcinoma of Head and Neck , Mouth Neoplasms/drug therapy , Tissue Distribution , Macrophages , Doxorubicin/pharmacology , Hydrogen-Ion Concentration , Tumor Microenvironment
16.
Elife ; 122024 Apr 03.
Article in English | MEDLINE | ID: mdl-38567911

ABSTRACT

The antibiotic heliomycin (resistomycin), which is generated from Streptomyces resistomycificus, has multiple activities, including anticancer effects. Heliomycin was first described in the 1960s, but its clinical applications have been hindered by extremely low solubility. A series of 4-aminomethyl derivatives of heliomycin were synthesized to increase water solubility; studies showed that they had anti-proliferative effects, but the drug targets remained unknown. In this study, we conducted cellular thermal shift assays (CETSA) and molecular docking simulations to identify and validate that heliomycin and its water-soluble derivative, 4-(dimethylaminomethyl)heliomycin (designated compound 4-dmH) engaged and targeted with sirtuin-1 (SIRT1) in p53-functional SAS and p53-mutated HSC-3 oral cancer cells. We further addressed the cellular outcome of SIRT1 inhibition by these compounds and found that, in addition to SIRT1, the water-soluble 4-dmH preferentially targeted a tumor-associated NADH oxidase (tNOX, ENOX2). The direct binding of 4-dmH to tNOX decreased the oxidation of NADH to NAD+ which diminished NAD+-dependent SIRT1 deacetylase activity, ultimately inducing apoptosis and significant cytotoxicity in both cell types, as opposed to the parental heliomycin-induced autophagy. We also observed that tNOX and SIRT1 were both upregulated in tumor tissues of oral cancer patients compared to adjacent normal tissues, suggesting their clinical relevance. Finally, the better therapeutic efficacy of 4-dmH was confirmed in tumor-bearing mice, which showed greater tNOX and SIRT1 downregulation and tumor volume reduction when treated with 4-dmH compared to heliomycin. Taken together, our in vitro and in vivo findings suggest that the multifaceted properties of water-soluble 4-dmH enable it to offer superior antitumor value compared to parental heliomycin, and indicated that it functions through targeting the tNOX-NAD+-SIRT1 axis to induce apoptosis in oral cancer cells.


Subject(s)
Mouth Neoplasms , Polycyclic Compounds , Sirtuin 1 , Humans , Animals , Mice , Sirtuin 1/metabolism , Cell Line, Tumor , NAD/metabolism , Tumor Suppressor Protein p53/metabolism , Molecular Docking Simulation , Apoptosis , Mouth Neoplasms/drug therapy
17.
BMC Oral Health ; 24(1): 412, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38575921

ABSTRACT

The present study aimed to investigate the impact of resveratrol on oral neoplastic parameters through a systematic review and meta-analysis. Resveratrol, a naturally occurring polyphenol, has shown promising potential as a therapeutic agent in various cancer types, including oral neoplasms. Understanding the collective findings from existing studies can shed light on the efficacy and mechanisms of resveratrol in oral cancer management. The systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A comprehensive search was performed to identify relevant studies from various databases, registers, websites, and citation searches. The inclusion criteria encompassed in-vivo studies investigating the impact of resveratrol on oral neoplastic parameters in animal models. After screening and assessment, a total of five eligible studies were included in the meta-analysis. The meta-analysis of the selected studies revealed that resveratrol treatment exhibited a potential impact on reducing oral neoplastic proliferation and promoting neoplastic apoptosis. The combined analysis showed a statistically significant decrease in neoplastic parameters with an overall effect size (ES) of 0.85 (95% CI: [0.74, 0.98]). Subgroup analyses were conducted to explore potential variations among different cellular types and exposure compounds, providing further insights into the efficacy of resveratrol in specific contexts. This systematic review and meta-analysis support the potential of resveratrol as a promising therapeutic agent in oral cancer management. The findings indicate that resveratrol may effectively modulate neoplastic proliferation and apoptosis in various cellular types within animal models of oral cancer. However, further well-controlled studies and clinical trials are warranted to validate these observations and elucidate the underlying mechanisms of resveratrol's actions. Resveratrol holds promise as a complementary therapeutic approach in the prevention and treatment of oral neoplastic conditions.


Subject(s)
Mouth Neoplasms , Animals , Resveratrol/therapeutic use , Mouth Neoplasms/drug therapy , Apoptosis
18.
Free Radic Biol Med ; 220: 28-42, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38679300

ABSTRACT

Cancer of the head and neck encompasses a wide range of cancers, including oral and oropharyngeal cancers. Oral cancer is often diagnosed at advanced stages and has a dismal prognosis. Piscidin-1, a marine antimicrobial peptide (AMP) containing approximately 22 amino acids, also exhibits significant anticancer properties. We investigated the possible anti-oral cancer effects of piscidin-1 and clarified the mechanisms underlying these effects. We treated the oral squamous cell carcinoma cell lines OC2 and SCC4 with piscidin-1. Cell viability and the expression of different hallmark apoptotic molecules, including reactive oxygen species (ROS), were tested using the appropriate MTT assay, flow cytometry and western blotting assays, and human umbilical vein endothelial cell (HUVEC) wound healing, migration, and tube formation (angiogenesis) assays. Piscidin-1 increases cleaved caspase 3 levels to induce apoptosis. Piscidin-1 also increases ROS levels and intensifies oxidative stress in the endoplasmic reticulum and mitochondria, causing mitochondrial dysfunction. Additionally, it decreases the oxygen consumption rates and activity of mitochondrial complexes I-V. As expected, the antioxidants MitoTEMPOL and N-acetylcysteine reduce piscidin-1-induced ROS generation and intracellular calcium accumulation. Piscidin-1 also inhibits matrix metalloproteinase (MMP)-2/-9 expression in HUVECs, affecting migration and tube formation angiogenesis. We demonstrated that piscidin-1 can promote apoptosis via both intrinsic and extrinsic apoptotic pathways and findings indicate that piscidin-1 has anti-proliferative and anti-angiogenic properties in oral cancer treatment. Our study on piscidin-1 thus provides a basis for future translational anti-oral cancer drug research and a new theoretical approach for anti-oral cancer clinical research.


Subject(s)
Antimicrobial Cationic Peptides , Apoptosis , Carcinoma, Squamous Cell , Fish Proteins , Human Umbilical Vein Endothelial Cells , Mouth Neoplasms , Neovascularization, Pathologic , Reactive Oxygen Species , Humans , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Mouth Neoplasms/pathology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Antimicrobial Cationic Peptides/pharmacology , Cell Line, Tumor , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Fish Proteins/pharmacology , Fish Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Oxidative Stress/drug effects , Angiogenesis Inhibitors/pharmacology , Animals , Angiogenesis
19.
J Dent Res ; 103(6): 612-621, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38684484

ABSTRACT

Oral squamous cell carcinoma (OSCC) is the most common type of oral cancer, characterized by invasiveness, local lymph node metastasis, and poor prognosis. Traditional treatment and medications have limitations, making the specific inhibition of OSCC growth, invasion, and metastasis a challenge. The tumor microenvironment exhibits mildly acidity and high concentrations of H2O2, and its exploitation for cancer treatment has been widely researched across various cancers, but research in the oral cancer field is relatively limited. In this study, by loading ultra-small Prussian blue nanoparticles (USPBNPs) into mesoporous calcium-silicate nanoparticles (MCSNs), we developed an acid-responsive iron-based nanocomposite, USPBNPs@MCSNs (UPM), for the OSCC treatment. UPM demonstrated excellent dual enzyme activities, generating toxic ·OH in a mildly acidic environment, effectively killing OSCC cells and producing O2 in a neutral environment to alleviate tissue hypoxia. The results showed that UPM could effectively inhibit the proliferation, migration, and invasion of OSCC cells, as well as the growth of mice solid tumors, without obvious systemic toxicity. The mechanisms may involve UPM inducing ferroptosis of OSCC cells by downregulating the xCT/GPX4/glutathione (GSH) axis, characterized by intracellular iron accumulation, reactive oxygen species accumulation, GSH depletion, lipid peroxidation, and abnormal changes in mitochondrial morphology. Therefore, this study provides empirical support for ferroptosis as an emerging therapeutic target for OSCC and offers a valuable insight for future OSCC treatment.


Subject(s)
Cell Proliferation , Iron , Mouth Neoplasms , Nanocomposites , Tumor Microenvironment , Nanocomposites/chemistry , Animals , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Mice , Humans , Cell Proliferation/drug effects , Tumor Microenvironment/drug effects , Cell Line, Tumor , Ferrocyanides/therapeutic use , Silicates/therapeutic use , Silicates/pharmacology , Hydrogen-Ion Concentration , Cell Movement/drug effects , Nanoparticles , Reactive Oxygen Species/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Mice, Nude , Ferroptosis/drug effects , Hydrogen Peroxide , Xenograft Model Antitumor Assays , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Mice, Inbred BALB C
20.
Cancer Lett ; 591: 216897, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38631664

ABSTRACT

Small extracellular vesicles (sEVs) residing at tumor tissues are valuable specimens for biopsy. Tumor heterogeneity is common across all cancer types, but the heterogeneity of tumor tissue-derived sEVs (Ti-sEVs) is undefined. This study aims to discover the spatial distributions of Ti-sEVs in oral squamous cell carcinoma (OSCC) tissues and explore how these vesicle distributions affect the patients' prognosis. Multi-regional sampling enabled us to uncover that Ti-sEVs' accumulation at peritumoral sites correlates with a higher disease-free survival rate, and conversely, sparse peritumoral Ti-sEVs tend to forecast a higher risk of relapse. Of those relapsed patients, Ti-sEVs strongly bind to extracellular matrix and subsequently degrade it for allowing themselves enter the bloodstream rather than staying in situ. In advanced OSCC patients, the quantity and spatial distribution of Ti-sEVs prior to anti-PD-1 treatment, as well as the temporal variance of Ti-sEVs before and after immunotherapy, strongly map the clinical response and can help to distinguish the patients with shrinking tumors from those with growing tumors. Our work elucidates the correlation of spatiotemporal features of Ti-sEVs with patients' therapeutic outcomes and exhibit the potential for using Ti-sEVs as a predictor to forecast prognosis and screen the responders to anti-PD-1 therapy.


Subject(s)
Extracellular Vesicles , Mouth Neoplasms , Neoplasm Recurrence, Local , Humans , Extracellular Vesicles/metabolism , Mouth Neoplasms/pathology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/immunology , Mouth Neoplasms/metabolism , Neoplasm Recurrence, Local/pathology , Female , Male , Middle Aged , Prognosis , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Tumor Microenvironment , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Aged , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Disease-Free Survival , Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...