Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
AME Case Rep ; 8: 7, 2024.
Article in English | MEDLINE | ID: mdl-38234361

ABSTRACT

Background: Giant keratoacanthoma, a rare variant solitary of keratoacanthoma is characterized by a diameter exceeding 20 mm, rapid growth, and destruction of underlying tissue. Traditionally, it has been considered to be a self-resolving or low-grade squamous proliferation. Due to their atypical appearance, Giant keratoacanthomas may present a diagnostic challenge. Histopathological examination remains the reference standard for distinguishing squamous cell carcinoma (SCC) from keratoacanthoma. Surgical management is still the standard treatment for solitary keratoacanthoma. Case Description: We present the case of a 75-year-old man with a giant keratoacanthoma of the cheek, with a transverse length of 25 mm and a longitudinal length of 30 mm located 30 mm below the earlobe that was surgically excised with no obvious recurrence during follow-up. We used the Intraoperative frozen-section examination, which played a crucial role in ensuring clear margins and complete tumor removal. Conclusions: This case demonstrates that keratoacanthoma can be considered surgically curable and should be distinguished from SCC and other crateriform tumors. After surgical resection of giant isolated keratoacanthoma on the face, the high risk of cosmetic problems should be considered. We suggest free-flap transplantation be performed for functional repair, when the invasion range of the tumor is large and a perforation defect is formed.

2.
BMC Cancer ; 23(1): 1062, 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37923984

ABSTRACT

BACKGROUND: This study aimed to find out the characteristics in relation to tumor recurrence in diffused-tenosynovial giant cell tumor of temporomandibular joint and to develop and validate the prognostic model for personalized prediction. METHODS: From April 2009 to January 2021, patients with diffused-tenosynovial giant cell tumor of temporomandibular joint at a single center were included in this study. The clinical features and local recurrence-free survival were assessed through the expression of the Ki-67 index and colony-stimulating factor 1 receptor expression. Both univariate and multivariate analyses were performed on the prognostic factors for local recurrence-free survival. An independent predictor nomogram and pertinent tumor characteristics were included. RESULTS: The retrospective study enrolling seventy eligible patients at the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine. During the follow-up time, eleven patients suffered tumor recurrence. Age was an independent risk factor for local recurrence-free survival (P = 0.032). The Ki-67 index varied significantly in different sites (P = 0.034) and tumor volume (P = 0.017). Multivariate logistic regression was used to develop the prediction model using both statistical significance and prognostic indicators. The C-index of the nomogram based on age, site, Ki-67, and colony-stimulating factor 1 receptor was 0.833. These variates provided good predicted accuracy for a nomogram on local recurrence-free survival. Diffused-tenosynovial giant cell tumor from the temporomandibular joint is extremely uncommon, and certain clinical traits are linked to the tumor proliferation index. CONCLUSIONS: We identified the risk indicators and developed a nomogram in this study to forecast the likelihood of local recurrence-free survival in patients with diffused-tenosynovial giant cell tumor from temporomandibular joint.


Subject(s)
Giant Cell Tumor of Tendon Sheath , Neoplasm Recurrence, Local , Humans , Retrospective Studies , Neoplasm Recurrence, Local/epidemiology , Neoplasm Recurrence, Local/pathology , Macrophage Colony-Stimulating Factor , Ki-67 Antigen , China , Giant Cell Tumor of Tendon Sheath/pathology , Temporomandibular Joint/pathology
3.
Int J Surg ; 109(8): 2220-2227, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37288582

ABSTRACT

BACKGROUND: Locally advanced oral squamous cell carcinoma (LAOSCC) is associated with a high rate of recurrence and poor survival. Given the recent successes of neoadjuvant immunochemotherapy (NAICT) in solid tumors, it is promising to use this treatment modality to achieve a better pathological response and improve the survival of LAOSCC, and clinical evidence is needed to assess its safety and efficacy. PATIENTS AND METHODS: A prospective trial of NAICT with toripalimab (PD-1 inhibitor) and albumin paclitaxel/cisplatin (TTP) was conducted in patients with clinical stage III and IVA OSCC. Intravenous albumin paclitaxel (260 mg/m 2 ), cisplatin (75 mg/m 2 ), and toripalimab (240 mg) were given in sequence on day 1 of each 21 day cycle for two cycles, followed by radical surgery and risk-adapted adjuvant (chemo)radiotherapy. The primary endpoints were safety and major pathological response (MPR). Targeted next generation sequencing and multiplex immunofluorescence were performed to assess clinical molecular characteristics and the tumor immune microenvironment in the pre-NAICT and post-NAICT tumor samples. RESULTS: Twenty patients were enrolled. NAICT was well-tolerated with a low incidence of grades 3-4 adverse events in three patients. The completion rates of NAICT and subsequent R0 resection were 100%. The MPR rate was 60%, including a 30% pathological complete response. MPR was achieved in all four patients with a combined positive score of PD-L1>10. The density of tertiary lymphatic structure in post-NAICT tumor samples predicted the pathological response to NAICT. During the median 23-month follow-up, the disease-free survival was 90%, and the overall survival was 95%. CONCLUSIONS: NAICT with the TTP protocol in LAOSCC is feasible and well tolerated, with a promising MPR and no obstruction on subsequent surgery. This trial is supportive of further randomized trials using NAICT in LAOSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/surgery , Neoadjuvant Therapy/adverse effects , Cisplatin , Squamous Cell Carcinoma of Head and Neck/chemically induced , Squamous Cell Carcinoma of Head and Neck/drug therapy , Mouth Neoplasms/drug therapy , Mouth Neoplasms/surgery , Treatment Outcome , Prospective Studies , Antineoplastic Combined Chemotherapy Protocols , Paclitaxel , Albumins/therapeutic use , Tumor Microenvironment
4.
Curr Med Imaging ; 2023 Mar 06.
Article in English | MEDLINE | ID: mdl-36876843

ABSTRACT

The objectives study aims to assess the position and route of the alveolar antral artery and the lateral wall thickness of the maxillary sinus using cone-beam computed tomography (CBCT), reducing the risk of complications and improving the success rate of surgery. MATERIALS AND METHODS: This study included CBCT scans from 238 patients. The detection diameter of AAA and distance of the lower border of AAA to the maxillary sinus floor at the first premolar, second premolar, first molar, and second molar locations were evaluated. The route of AAA was observed with novel classification. Furthermore, the distance from the maxillary sinus floor to the alveolar crest at four posterior tooth locations was measured respectively. Moreover, the lateral wall thickness at four locations was assessed. Data were subjected to statistical analysis. RESULTS: AAA was observed in 62.18% of all sinuses. The mean diameter was 0.99±0.21 mm, with significant differences within gender. Half of the route of AAA was intrasinus intraosseous type. The mean distance between the maxillary sinus floor and AAA was 8.00±2.68 mm, with a significant difference between dentate and edentulous status at the first molar location. Distance from the sinus floor to the alveolar ridge crest in edentulous status negatively correlated with the distance from the sinus floor to AAA at the first molar location. The mean thickness of the lateral wall was 2.03±0.91 mm, and the difference in thickness between males and females at the four locations was statistically significant. CONCLUSION: intrasinus-intraosseous type, is the most common route. Special care should be taken at the first molar location during a lateral window sinus floor elevation. CBCT is highly recommended to before lateral wall maxillary sinus floor elevation.

5.
Int J Surg ; 109(4): 879-886, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36999830

ABSTRACT

BACKGROUND: Surgery and postoperative adjuvant therapy comprise the standard treatment for locally advanced resectable oral squamous cell carcinoma (LAROSCC), while preoperative neoadjuvant therapy is being explored without sufficient confirmation of improved survival. De-escalation regimens after neoadjuvant therapy, such as those omitting adjuvant radiotherapy, may provide comparable or better outcomes, suggesting rigorous assessment of adjuvant therapy outcomes is needed in LAROSCC patients. The authors thus performed this retrospective study in LAROSCC patients who received neoadjuvant therapy and surgery, to compare the outcomes for overall survival (OS) and locoregional recurrence-free survival (LRFS) between the adjuvant radiotherapy (radio) and nonradiotherapy (nonradio) cohorts. MATERIALS AND METHODS: Patients diagnosed with LAROSCC who received neoadjuvant therapy and surgery were enrolled and divided into radio and nonradio cohorts to determine whether adjuvant radiotherapy could be omitted after neoadjuvant therapy and surgery. RESULTS: From 2008 to 2021, 192 patients were enrolled. No significant differences were found in OS or LRFS between the radio and nonradio patient cohorts. The 10-year estimated OS rates were 58.9 versus 44.1% in radio versus nonradio cohorts, while 10-year estimated LRFS rates were 55.4 versus 48.2%, respectively. For clinical stage III patients, 10-year OS rates were 62.3 versus 62.6% (radio vs. nonradio), and estimated 10-year LRFS rates were 56.5 versus 60.7% (radio vs. nonradio). Multivariate Cox regression modeling of postoperative variables showed pathologic response of primary tumor and pathologic regional lymph nodes staging were associated with survival, while the adjuvant radiotherapy exposure was not included in the model due to nonsignificance. CONCLUSION: These findings support further prospective evaluation of adjuvant radiotherapy omission, and suggest that de-escalation trials are warranted for LAROSCC surgery patients who received neoadjuvant therapy.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Humans , Radiotherapy, Adjuvant , Retrospective Studies , Neoadjuvant Therapy , Carcinoma, Squamous Cell/radiotherapy , Carcinoma, Squamous Cell/surgery , Mouth Neoplasms/radiotherapy , Mouth Neoplasms/surgery , Neoplasm Recurrence, Local , Chemotherapy, Adjuvant , Neoplasm Staging
6.
Nat Commun ; 13(1): 5378, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36104359

ABSTRACT

Novel neoadjuvant therapy regimens are warranted for oral squamous cell carcinoma (OSCC). In this phase I trial (NCT04393506), 20 patients with locally advanced resectable OSCC receive three cycles of camrelizumab (200 mg, q2w) and apatinib (250 mg, once daily) before surgery. The primary endpoints are safety and major pathological response (MPR, defined as ≤10% residual viable tumour cells). Secondary endpoints include 2-year survival rate and local recurrence rate (not reported due to inadequate follow-up). Exploratory endpoints are the relationships between PD-L1 combined positive score (CPS, defined as the number of PD-L1-stained cells divided by the total number of viable tumour cells, multiplied by 100) and other immunological and genomic biomarkers and response. Neoadjuvant treatment is well-tolerated, and the MPR rate is 40% (8/20), meeting the primary endpoint. All five patients with CPS ˃10 achieve MPR. Post-hoc analysis show 18-month locoregional recurrence and survival rates of 10.5% (95% CI: 0%-24.3%) and 95% (95% CI: 85.4%-100.0%), respectively. Patients achieving MPR show more CD4+ T-cell infiltration than those without MPR (P = 0.02), and decreased CD31 and ɑ-SMA expression levels are observed after neoadjuvant therapy. In conclusion, neoadjuvant camrelizumab and apatinib is safe and yields a promising MPR rate for OSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Antibodies, Monoclonal, Humanized , B7-H1 Antigen/metabolism , Carcinoma, Squamous Cell/pathology , Humans , Mouth Neoplasms/drug therapy , Neoadjuvant Therapy , Neoplasm Recurrence, Local , Pilot Projects , Pyridines , Squamous Cell Carcinoma of Head and Neck
7.
Front Psychiatry ; 13: 956566, 2022.
Article in English | MEDLINE | ID: mdl-36061304

ABSTRACT

Purpose: To examine the association of preoperative dental anxiety with the severity of postoperative symptoms among patients undergoing lower third molar (LM3) extraction surgery. Materials and methods: We conducted a hospital-based prospective study with a sample size of 213 patients. All the patients underwent LM3 extraction surgery at the Stomatology Hospital of Tianjin Medical University. Preoperative dental anxiety was measured using the Dental Anxiety Scale for Third Molar Surgery (DAS-TMS) and classified into four categories: No anxiety, Some unease, Anxious, and Very anxious. The primary outcome was defined using the postoperative symptom severity scale on the seventh day after surgery. The patients' clinical characteristics, radiologic features, and surgery-related variables were used as control variables. Bivariate analysis involved Fisher's exact test and Kruskal-Wallis test. Multivariable logistic analysis was used to assess preoperative dental anxiety in relation to the severity of postoperative symptoms. We applied a two-piecewise regression model to examine the potential non-linear associations. Results: The mean (SD) dental anxiety score was 10.56 (3.84). The proportion of dental anxiety was as follows: No anxiety, 7.5%; Some unease, 46.9%; Anxious, 31.0%; Very anxious, 14.6%. The multivariable-adjusted ORs with 95% CIs of postoperative symptoms were 1.00 for No anxiety, 3.63 (0.90-14.68) for Some unease, 5.29 (1.25-22.33) for Anxious, and 4.75 (1.02-22.18) for Very anxious (P for trend = 0.047). The risk of serious postoperative symptoms increased with the dental anxiety level up to 7 points (adjusted OR 1.94, 95% CI 1.12-3.74; P = 0.012). When the dental anxiety level exceeded 7 points, the level of DAS-TMS was not associated with the risk of serious postoperative symptoms (OR 0.98, 95% CI 0.88-1.08; P = 0.756). Conclusions: Findings suggest that dental anxiety is associated with a risk of serious postoperative symptoms following LM3 removal. The degree of dental anxiety in patients before LM3 extraction surgery should be of concern to clinicians.

8.
BMC Oral Health ; 22(1): 265, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35768820

ABSTRACT

BACKGROUND: Schwannomas or neurilemmomas are well-encapsulated, benign, solitary, and slow-growing tumors that originate from Schwann cells of the nerve sheath. Extracranial schwannoma is reported to have a relatively high incidence in the tongue while an extremely low incidence in the floor of mouth. In the current study, we presented the first case series of hypoglossal nerve-derived schwannoma in the floor of mouth in Asia. METHODS: A retrospective study of 9 surgical cases of hypoglossal nerve-derived schwannoma in the floor of mouth was performed. The patient and tumor characteristics were evaluated by physical, radiological and pathological examination. Details of operation and complications were also recorded. RESULTS: Hypoglossal nerve-derived schwannoma in the floor of mouth showed a well-defined boundary with a firm texture, smooth surface and good mobility on palpation. The median maximum diameter of the tumors was 4.3 cm (range 2.8-7.0 cm). The median operative time and bleeding volumes were 89.4 min (range 47-180 min) and 99.2 mL (range 15-200 mL), respectively. All cases received complete surgical excision. CONCLUSION: In this study, we presented the diagnosis and management of hypoglossal nerve-derived schwannoma in the floor of mouth for the first time in Asia. The study provided us with a recommendation for consideration of the diagnosis of hypoglossal schwannoma when a patient presents with a mass in the floor of mouth.


Subject(s)
Cranial Nerve Neoplasms , Hypoglossal Nerve Diseases , Neurilemmoma , Cranial Nerve Neoplasms/diagnosis , Cranial Nerve Neoplasms/pathology , Cranial Nerve Neoplasms/surgery , Humans , Hypoglossal Nerve/pathology , Hypoglossal Nerve/surgery , Hypoglossal Nerve Diseases/diagnosis , Hypoglossal Nerve Diseases/etiology , Hypoglossal Nerve Diseases/surgery , Mouth Floor/pathology , Mouth Floor/surgery , Neurilemmoma/diagnostic imaging , Neurilemmoma/surgery , Retrospective Studies
9.
Cancer Sci ; 113(2): 478-488, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34826159

ABSTRACT

The aim of this study was to: (a) explore the potential mechanism of cancer cell sensitivity to cisplatin, docetaxel, and 5-fluorouracil (TPF) in oral squamous cell carcinoma (OSCC) patients overexpressing growth differentiation factor 15 (GDF15); and (b) identify potential alternative agents for patients who might not benefit from inductive TPF chemotherapy. The results indicated that OSCC cells overexpressing GDF15 were sensitive to TPF through a caspase-9-dependent pathway both in vitro and in vivo. Immunoprecipitation combined with mass spectrometry revealed that the erbB2 protein was a potential GDF15-binding protein, which was verified by coimmunoprecipitation. Growth differentiation factor 15 overexpression promoted OSCC cell proliferation through erbB2 phosphorylation, as well as downstream AKT and Erk signaling pathways. When GDF15 expression was blocked, the phosphorylation of both the erbB2 and AKT/Erk pathways was downregulated. When OSCC cells with GDF15 overexpression were treated with the erbB2 phosphorylation inhibitor, CI-1033, cell proliferation and xenograft growth colony formation were significantly blocked (P < .05). Thus, GDF15-overexpressing OSCC tumors are sensitive to TPF chemoagents through caspase-9-dependent pathways. Growth differentiation factor 15 overexpression promotes OSCC proliferation through erbB2 phosphorylation. Thus, ErbB2 inhibitors could represent potential targeted drugs or an alternative therapy for OSCC patients with GDF15 overexpression.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Growth Differentiation Factor 15/metabolism , Mouth Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Animals , Apoptosis , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cisplatin/pharmacology , Fluorouracil/pharmacology , Humans , Mice , Morpholines/pharmacology , Phosphorylation/drug effects , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction/drug effects , Taxoids/pharmacology
10.
Am J Cancer Res ; 11(6): 2960-2974, 2021.
Article in English | MEDLINE | ID: mdl-34249438

ABSTRACT

Apatinib is an oral tyrosine kinase inhibitor that targets VEGFR2 signaling and shows potent antitumor effects in various cancers. In this study, we explored the efficacy of apatinib against oral squamous cell carcinoma (OSCC). The relationships between VEGFR2 protein expression and clinical variables were investigated in OSCC patients. OSCC tissues had higher VEGFR2 levels than paracancerous tissues. Compared to patients with low VEGFR2 expression, patients with high VEGFR2 expression had poorer overall survival (OS) and disease-free survival (DFS). Apatinib significantly induced G0/G1 phase arrest and apoptosis, inhibited cell growth and colony formation ability, and blocked autophagic flux by downregulating p-AKT and p-mTOR signaling via the VEGFR2/AKT/mTOR pathway in vitro. Moreover, the inhibition of ERK phosphorylation increased apatinib-induced apoptosis in vitro and in vivo. Apatinib synergized with SCH772984 to achieve a more significant suppression of tumor growth than individual treatment, suggesting the combination of apatinib and SCH772984 as a potent OSCC therapy.

11.
J Oral Pathol Med ; 50(8): 795-802, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34157171

ABSTRACT

BACKGROUND: Annexin A1, a member of the Annexin superfamily, has been shown to play a vital role in a broad range of molecular and cellular processes. This study aims to explore the relationship between the Annexin A1 expression and the clinical response to cisplatin, docetaxel and 5-fluorouracil (TPF) as induction chemotherapy in patients with oral squamous cell carcinoma (OSCC). METHODS: This study recruited two hundred thirty-two patients from a III/IVA OSCC trial. Immunohistochemistry was used to assess the level of Annexin A1 expression. Overexpression and knockdown methods in HB96, HN4 and CAL27 cell lines were used to assess the role of Annexin A1 in the neoplastic cellular response to chemotherapy. RESULTS: We found that reduced expression of Annexin A1 conferred a prognostic benefit from induction chemotherapy based on the TPF drug combination in patients with moderately/poorly differentiated disease. Using an in vitro model, we found that low Annexin A1 enhanced cellular proliferation by activating the EGFR/AKT signalling pathway and inhibiting p27 expression. Furthermore, low Annexin A1 initiated a significant decrease in cell viability after treatment with TPF agents. In addition, downregulation of Annexin A1 promoted apoptosis induced by docetaxel, cisplatin and 5-fluorouracil, and upregulation of Annexin A1 inhibited apoptosis. CONCLUSION: Annexin A1 may be of prognostic value in patients with locally advanced OSCC who are managed with TPF chemotherapy, as low Annexin A1 promotes chemosensitivity to TPF chemotherapy in oral cancer cells via enhanced caspase-dependent apoptosis.


Subject(s)
Annexin A1 , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Annexin A1/genetics , Annexin A1/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Cisplatin/pharmacology , Cisplatin/therapeutic use , Docetaxel/pharmacology , Docetaxel/therapeutic use , Fluorouracil/therapeutic use , Head and Neck Neoplasms/drug therapy , Humans , Induction Chemotherapy , Mouth Neoplasms/drug therapy , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Taxoids/therapeutic use
12.
Am J Cancer Res ; 11(4): 1503-1521, 2021.
Article in English | MEDLINE | ID: mdl-33948370

ABSTRACT

Former clinical trials and experimental research have indicated that Interferon-gamma therapy does not achieve an ideal effect in solid tumors. Autophagy has been associated with tumor chemoresistance. The aim of this study was to explore the efficacy of Interferon-gamma and autophagy inhibitor in the combination treatment of oral squamous cell carcinoma. Interferon-gamma-induced apoptosis was evaluated by the expression of relative proteins (cleaved-PARP and caspase-3) and flow cytometry. Interferon-gamma induced autophagy was assessed by the expression of Beclin1, LC3B, and P62. The synergistic effect of interferon-gamma and autophagy inhibitor (chloroquine) was evaluated in vitro and in vivo. Interferon-gamma induced anti-proliferation, apoptosis, and autophagy in oral squamous cell carcinoma cells. Autophagy-related protein 5 was a key feature in Interferon-gamma-induced autophagy flux. Interferon-gamma and chloroquine had obvious synergistic effects on cellular growth inhibition and apoptosis promotion in oral squamous cell carcinoma cells and xenograft models. Our findings suggest that Interferon-gamma-induced autophagy plays a cellular protective role, and blocking autophagy flux can promote Interferon-gamma mediated oral squamous cell carcinoma cell apoptosis. The combination of Interferon-gamma and autophagy inhibitors represents a novel strategy for oral squamous cell carcinoma therapy.

13.
J Nanobiotechnology ; 19(1): 157, 2021 May 26.
Article in English | MEDLINE | ID: mdl-34039370

ABSTRACT

BACKGROUND: Smart nanoscale drug delivery systems that target acidic tumor microenvironments (TME) could offer controlled release of drugs and modulate the hypoxic TME to enhance cancer therapy. The majority of previously reported MnO2 nanostructures are nanoparticles, nanosheets, or nanocomposites incorporated with other types of nanoparticles, which may not offer the most effective method for drug loading or for the controlled release of therapeutic payloads. Previous studies have designed MnO2 nanoshells that achieve tumor-specific and enhanced combination therapy for localized advanced cancer. However, the therapeutic effect of MnO2 nanoshells on metastatic cancer is still uncertain. RESULT: Here, intelligent "theranostic" platforms were synthesized based on hollow mesoporous MnO2 (H-MnO2) nanoshells that were loaded with chemotherapy agents docetaxel and cisplatin (TP) to form H-MnO2-PEG/TP nanoshells, which were designed to alleviate tumor hypoxia, attenuate angiogenesis, trigger the dissolution of Mn2+, and synergize the efficacy of first-class anticancer chemotherapy. The obtained H-MnO2-PEG/TP nanoshells decomposed in the acidic TME, releasing the loaded drugs (TP) and simultaneously attenuated tumor hypoxia and hypoxia-inducible factor-1α (HIF-1α) expression by inducing endogenous tumor hydrogen peroxide (H2O2) decomposition. In vitro experiments showed that compared with the control group, the proliferation, colony formation and migration ability of CAL27 and SCC7 cells were significantly reduced in H-MnO2-PEG/TP group, while cell apoptosis was enhanced, and the expression of hypoxia-inducible factor-1α(HIF-1α) was down-regulated. In vivo experiments showed that tumor to normal organ uptake ratio (T/N ratio) of mice in H-MnO2-PEG/TP group was significantly higher than that in TP group alone (without the nanoparticle), and tumor growth was partially delayed. In the H-MnO2-PEG/TP treatment group, HE staining showed that most of the tumor cells were severely damaged, and TUNEL assay showed cell apoptosis was up-regulated. He staining of renal and liver sections showed no obvious fibrosis, necrosis or hypertrophy, indicating good biosafety. Fluorescence staining showed that HIF-1α expression was decreased, suggesting that the accumulation of MnO2 in the tumor caused the decomposition of H2O2 into O2 and alleviated the hypoxia of the tumor. CONCLUSION: In conclusion, a remarkable in vivo and in vitro synergistic therapeutic effect is achieved through the combination of TP chemotherapy, which simultaneously triggered a series of antiangiogenic and oxidative antitumor reactions.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Drug Therapy/methods , Hypoxia/drug therapy , Manganese Compounds/chemistry , Mouth Neoplasms/drug therapy , Nanoshells/chemistry , Squamous Cell Carcinoma of Head and Neck/drug therapy , Tumor Microenvironment/drug effects , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Delivery Systems , Head and Neck Neoplasms/drug therapy , Humans , Hydrogen Peroxide/metabolism , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Oxides/chemistry , Theranostic Nanomedicine/methods , Tumor Hypoxia/drug effects
14.
Transl Oncol ; 14(6): 101074, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33744726

ABSTRACT

PURPOSE: Carrimycin is a newly synthesized macrolide antibiotic with good antibacterial effect. Exploratory experiments found its function in regulating cell physiology, proliferation and immunity, suggesting its potential anti-tumor capacity. The aim of this study is to investigate the anti-tumor effect of carrimycin against human oral squamous cell carcinoma cells in vitro and in vivo. METHODS: Human oral squamous cell carcinoma cells (HN30/HN6/Cal27/HB96 cell lines) were treated with gradient concentration of carrimycin. Cell proliferation, colony formation and migration ability were analyzed. Cell cycle and apoptosis were assessed by flow cytometry. The effect of carrimycin on OSCC in vivo was investigated in tumor xenograft models. Immunohistochemistry, western blot assay and TUNEL assays of tissue samples from xenografts were performed. The key proteins in PI3K/AKT/mTOR pathway and MAPK pathway were examined by western blot. RESULTS: As the concentration of carrimycin increased, the proliferation, colony formation and migration ability of OSCC cells were inhibited. After treating with carrimycin, cell cycle was arrested in G0/G1 phase and cell apoptosis was promoted. The tumor growth of xenografts was significantly suppressed. Furthermore, the expression of p-PI3K, p-AKT, p-mTOR, p-S6K, p-4EBP1, p-ERK and p-p38 were down-regulated in vitro and in vivo. CONCLUSIONS: Carrimycin can inhibit the biological activities of OSCC cells in vitro and in vivo, and regulate the PI3K/AKT/mTOR and MAPK pathways.

15.
Article in English | MEDLINE | ID: mdl-32981877

ABSTRACT

OBJECTIVES: The aim of this study was to retrospectively analyze the clinical characteristics, surgical treatment, and prognosis of patients with diffuse-type tenosynovial giant cell tumor (D-TGCT) involving the temporomandibular joint (TMJ) and the skull base. STUDY DESIGN: A retrospective study was performed in patients with D-TGCT involving the TMJ and the skull base at our institute from April 2009 to August 2018. Data on clinical characteristics, surgical treatment, and prognosis were collected and analyzed. A literature search on D-TGCT involving the TMJ was conducted and the data analyzed. RESULTS: The study included 22 patients (14 males and 8 females), with an average age of 44 years. The main symptoms were headache and hearing limitation, accompanied by a swelling in the TMJ area. Magnetic resonance imaging (MRI) showed low signals on T1- and T2-weighted images. All lesions were completely removed. Temporal bone flap, titanium mesh, and temporal muscle flap were used for reconstruction. The recurrence rate was 4.5%. In the literature, 115 cases were reported. Surgery alone was performed in 88 cases; postoperative radiotherapy was performed in 19 cases; the tumor recurrence rates were 9.1% and 15.8% for the 2 procedures, respectively. All patients were alive at the end of the follow-up period. CONCLUSIONS: D-TGCT involving the TMJ and the skull base is a locally aggressive but benign lesion necessitating complete resection and has a good prognosis.


Subject(s)
Giant Cell Tumor of Tendon Sheath , Neoplasm Recurrence, Local , Adult , Female , Giant Cell Tumor of Tendon Sheath/diagnostic imaging , Giant Cell Tumor of Tendon Sheath/surgery , Humans , Male , Retrospective Studies , Skull Base/diagnostic imaging , Skull Base/surgery , Temporomandibular Joint/diagnostic imaging , Temporomandibular Joint/surgery
16.
Curr Drug Deliv ; 18(2): 234-245, 2021.
Article in English | MEDLINE | ID: mdl-32811397

ABSTRACT

OBJECTIVE: This study aimed to fabricate Hyaluronic Acid (HA)/parecoxib-loaded PLGA microspheres for the treatment of Temporomandibular Disorders (TMD) and investigate the in vitro and in vivo effect of the microsphere system to solve the issues of poor drug delivery and short duration on drug concentration in conventional TMD therapy. METHODS: The microspheres were prepared by the double emulsion (w/o/w) method. Various formulations were compared in terms of particle size, drug loading rate and encapsulation rate. Scanning Electron Microscopy (SEM), Differential Scanning Calorimetry (DSC) and FT-IR spectroscopy were performed to evaluate physicochemical properties. The drug release behavior of microspheres and toxicity assay on synovial cells were investigated. The in vitro anti-inflammatory effect on inflammatory markers, such as IL-1ß, TNF-α and COX-2, was assessed by real-time PCR. Then, the in vivo therapeutic effect of microspheres was investigated using mechanically-induced rat synovitis model. Protein levels of inflammatory cytokines (IL-1ß, TNF-α and COX-2) from TMJ periarticular tissues were quantified by Enzyme-Linked Immunosorbent Assay (ELISA). RESULTS: The results showed that microspheres were morphologically regular, smooth and non-cohesive. The average particle size of the microspheres was (25.32 ± 1.01) µm. The drug loading rate of parecoxib was 17.12%-20.95% with encapsulation efficiency reaching 51.9%-54.7%. In vitro drug release tests showed a successful sustained release over 28 days with a burst of 19.98% of the total drug substance. Treatment with HA/parecoxib-loaded PLGA microspheres declined the mRNA expression of IL-1ß, TNF-α and COX-2 induced by LPS in articular synovial cells. Moreover, in vivo results demonstrated that the intra-articular microspheres significantly reduced protein levels of inflammatory cytokines (IL-1ß, TNF-α and COX-2) for more than two weeks and stopped the mechanically-induced synovitis in its tracks in rat models. CONCLUSION: The study presented new and potential insights into treatments of TMD using PLGA microspheres loaded with HA and parecoxib as a successful drug delivery system.


Subject(s)
Hyaluronic Acid , Isoxazoles/pharmacology , Temporomandibular Joint Disorders , Animals , Isoxazoles/chemistry , Microspheres , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Spectroscopy, Fourier Transform Infrared , Temporomandibular Joint Disorders/drug therapy
17.
Oncol Lett ; 20(5): 154, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32934722

ABSTRACT

Induction chemotherapy has been previously demonstrated to downgrade locally advanced or aggressive cancers and increase the likelihood of primary lesion eradication. Based on our previous phase 3 trial on TPF (docetaxel, cisplatin and fluorouracil) induction chemotherapy in patients with oral squamous cell carcinoma (OSCC), in which short-term prognostic and predictive values of cyclin D1 expression were reported, the present study aimed to determine the long-term predictive value of cyclin D1 expression in the same patients with OSCC who were eligible to receive TPF induction chemotherapy. In addition, the present study investigated the potential association between cyclin D1 expression and chemosensitivity to TPF agents during OSCC cell intervention, and the underlying apoptotic mechanism of action. In total, 232 patients with locally advanced OSCC from our previous trial with a median follow-up of 5 years were included for survival analysis using the Kaplan-Meier method and the log-rank test in the present study, where cyclin D1 expression in their tissues was detected by immunohistochemistry. Cyclin D1 knockdown, cytotoxicity assays assessing the efficacy of the TPF chemotherapeutic agents and measurements of caspase-3 and PARP activity in HB96, CAL27 and HN30 cell lines were performed. Patients with OSCC in the low cyclin D1 expression group exhibited significantly superior long-term clinical outcomes compared with those in patients in the high cyclin D1 expression group [overall survival (OS), P=0.001; disease-free survival, P=0.003; local recurrence-free survival, P=0.004; distant metastasis-free survival (DMFS), P=0.001]. Furthermore, patients with stage clinical nodal stage 2 (cN2) OSCC in the high cyclin D1 expression group benefitted from TPF induction chemotherapy (OS, P=0.024; DMFS, P=0.024), whilst patients with cN2 OSCC in the low cyclin D1 expression group did not benefit from this chemotherapy. Overexpression of cyclin D1 expression was found to enhance chemosensitivity to TPF chemotherapeutic agents in OSCC by mediating caspase-3-dependent apoptosis. Based on these findings, TPF induction chemotherapy can benefit patients with cN2 OSCC and high cyclin D1 expression in terms of long-term survival from compared with standard treatment. In addition, OSCC cell lines overexpressing cyclin D1 are more sensitive to TPF chemotherapeutic agents in a caspase-3-dependent manner (clinical trial. no. NCT01542931; February 2012).

18.
Biochem Biophys Res Commun ; 531(3): 282-289, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32800541

ABSTRACT

B7H3 is a member of B7 family of immunoregulatory transmembrane glycoproteins associated with maintaining immune tolerance, tumor cell proliferation, migration, invasion and metabolism, drug resistance, and stem cell differentiation. Neural crest-derived Multipotent Stem Cells (MSCs) from the dental pulp has become a good choice for tissue regeneration because it is easily obtainable and has strong regeneration potentials. Although there have been many studies investigating the role of B7H3 in cancer cells and immune cells, its role in the dental pulp stem cells regeneration is unknown. In this study, we chose SHEDs (stem cells from human exfoliated deciduous teeth) as a research model to analyze the expression and function of B7H3. The result showed that SHEDs were B7H3/CD90, B7H3/CD73, B7H3/CD105 double positive, and the expression of B7H3 is primarily located within the membrane. Downregulation of B7H3 expression significantly accelerated the expansion of SHEDs through the SHP1/AKT signal axis while upregulation of B7H3 expression decreased the proliferation of SHEDs. Hence, this study indicates that B7H3 is a stem cell surface molecule and might be used as a SHEDs marker whereby its downregulation enhances the proliferation of SHEDs via the activation of B7H3/SHP1/AKT signaling pathway.


Subject(s)
B7 Antigens/metabolism , Gene Knockdown Techniques , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Cell Membrane/metabolism , Cell Proliferation , Child , Child, Preschool , Humans , Stem Cells , Tooth, Deciduous
19.
Bioact Mater ; 5(4): 844-858, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32637748

ABSTRACT

Pure collagen is biocompatible but lacks inherent osteoinductive, osteoimmunomodulatory and antibacterial activities. To obtain collagen with these characteristics, we developed a novel methodology of doping bioactive elements into collagen through the synchronous self-assembly/mineralization (SSM) of collagen. In the SSM model, amorphous mineral nanoparticles (AMN) (amorphous SrCO3, amorphous Ag3PO4, etc.) stabilized by the polyampholyte, carboxymethyl chitosan (CMC), and collagen molecules were the primary components under acidic conditions. As the pH gradually increased, intrafibrillar mineralization occurred via the self-adaptive interaction between the AMNs and the collagen microfibrils, which were self-assembling; the AMNs wrapped around the microfibrils became situated in the gap zones of collagen and finally transformed into crystals. Sr-doped collagen scaffolds (Sr-CS) promoted in vitro cell proliferation and osteogenic differentiation of rat bone marrow mesenchymal stromal cells (rBMSCs) and synergistically improved osteogenesis of rBMSCs by altering the macrophage response. Ag-doped collagen scaffolds (Ag-CS) exhibited in vitro antibacterial effects on S. aureus, as well as cell/tissue compatibility. Moreover, Sr-CS implanted into the calvarial defect of a rat resulted in improved bone regeneration. Therefore, the SSM model is a de novo synthetic strategy for doping bioactive elements into collagen, and can be used to fabricate multifunctional collagen scaffolds to meet the clinical challenges of encouraging osteogenesis, boosting the immune response and fighting severe infection in bone defects.

20.
J Cell Mol Med ; 24(13): 7163-7174, 2020 07.
Article in English | MEDLINE | ID: mdl-32469152

ABSTRACT

The up-regulation of EMT regulator Twist1 has been implicated in vasculogenic mimicry (VM) formation in human triple-negative breast cancer (TNBC). Twist1 targets the Claudin15 promoter in hepatocellular carcinoma cells. Claudin family members are related with TNBC. However, the relationship between Claudin15 and VM formation is not clear. In this study, we first found that Claudin15 expression was frequently down-regulated in human TNBC, and Claudin15 down-regulation was significantly associated with VM and Twist1 nuclear expression. Claudin15 down-regulation correlated with shorter survival compared with high levels. Claudin15 silence significantly enhanced cell motility, invasiveness and VM formation in the non-TNBC MCF-7 cells. Conversely, an up-regulation of Claudin15 remarkably reduced TNBC MDA-MB-231 cell migration, invasion and VM formation. We also showed that down-regulation of Claudin15 was Twist1-dependent, and Twist1 repressed Claudin15 promoter activity. Furthermore, GeneChip analyses of mammary glands of Claudin15-deficient mice indicated that Claudin18 and Jun might be downstream factors of Twist1-Claudin15. Our results suggest that Twist1 induced VM through Claudin15 suppression in TNBC, and Twist1 inhibition of Claudin15 might involve Claudin18 and Jun expression.


Subject(s)
Claudins , Gene Expression Regulation, Neoplastic , Nuclear Proteins , Triple Negative Breast Neoplasms , Twist-Related Protein 1 , Animals , Female , Humans , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/metabolism , Claudins/deficiency , Claudins/genetics , Claudins/metabolism , Kaplan-Meier Estimate , Mammary Glands, Animal/pathology , Neoplasm Invasiveness , Nuclear Proteins/metabolism , Phenotype , Transcription, Genetic , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Twist-Related Protein 1/metabolism , Up-Regulation/genetics , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...