Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
BMC Med ; 22(1): 314, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39075531

RESUMO

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).


Assuntos
Cisplatino , Docetaxel , Integrina beta1 , Neoplasias Bucais , Humanos , Docetaxel/uso terapêutico , Docetaxel/administração & dosagem , Feminino , Masculino , Pessoa de Meia-Idade , Cisplatino/uso terapêutico , Cisplatino/administração & dosagem , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/terapia , Idoso , Adulto , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/terapia , Biomarcadores Tumorais , Antineoplásicos/uso terapêutico , Resultado do Tratamento
2.
Adv Healthc Mater ; 13(19): e2304639, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38642071

RESUMO

The management of oral squamous cell carcinoma (OSCC) poses significant challenges, leading to organ impairment and ineffective treatment of deep-seated tumors, adversely affecting patient prognosis. A cascade nanoreactor that integrates photodynamic therapy (PDT) and chemodynamic therapy (CDT) for comprehensive multimodal OSCC treatment is introduced. Utilizing iron oxide and mesoporous silica, the FMMSH drug delivery system, encapsulating the photosensitizer prodrug δ-aminolevulinic acid (δ-ALA), is developed. Triphenylphosphine (TPP) modification facilitates mitochondrial targeting, while tumor cell membrane (TCM) coating provides homotypic targeting. The dual-targeting δ-ALA@FMMSH-TPP-TCM demonstrate efficacy in eradicating both superficial and deep tumors through synergistic PDT/CDT. Esterase overexpression in OSCC cells triggers δ-ALA release, and excessive hydrogen peroxide in tumor mitochondria undergoes Fenton chemistry for CDT. The synergistic interaction of PDT and CDT increases cytotoxic ROS levels, intensifying oxidative stress and enhancing apoptotic mechanisms, ultimately leading to tumor cell death. PDT/CDT-induced apoptosis generates δ-ALA-containing apoptotic bodies, enhancing antitumor efficacy in deep tumor cells. The anatomical accessibility of oral cancer emphasizes the potential of intratumoral injection for precise and localized treatment delivery, ensuring focused therapeutic agent delivery to maximize efficacy while minimizing side effects. Thus, δ-ALA@FMMSH-TPP-TCM, tailored for intratumoral injection, emerges as a transformative modality in OSCC treatment.


Assuntos
Ácido Aminolevulínico , Mitocôndrias , Neoplasias Bucais , Fotoquimioterapia , Fármacos Fotossensibilizantes , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Fotoquimioterapia/métodos , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Ácido Aminolevulínico/química , Ácido Aminolevulínico/farmacologia , Camundongos , Dióxido de Silício/química , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/metabolismo , Apoptose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Compostos Organofosforados/química , Compostos Organofosforados/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Camundongos Nus
3.
MedComm (2020) ; 4(4): e312, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37492783

RESUMO

Pembrolizumab with cisplatin and 5-fluorouracil showed survival benefit but relatively high occurrence of treatment-related adverse events (TRAEs) for recurrent/metastatic oral squamous cell carcinoma (R/M OSCC). A more tolerable regime is needed. This trial enrolled 20 R/M OSCC patients with previously untreated and PD-L1 positive. Patients were administered camrelizumab with docetaxel and cisplatin every 3 weeks for six cycles, followed by camrelizumab monotherapy every 3 weeks until disease progression or intolerable toxicity. The primary endpoint was occurrence of grade ≥ 3 TRAEs, secondary endpoints included overall survival (OS), progression-free survival (PFS), and overall response rate (ORR). 45% patients experienced grade ≥ 3 TRAEs, which the most common were anemia (15%), stomatitis (15%), and neutropenia (10%). The most common potential immune-related adverse events were reactive cutaneous capillary endothelial proliferation (RCCEP; 60%), hypothyroidism (35%), and pneumonitis (15%). No treatment-related deaths occurred. The median OS, PFS, and ORR was 14.4 months, 5.35 months, and 40.0% respectively. The study also found RCCEP occurrence, lower FOXP3+ cells, and higher density of intratumor tertiary lymphoid structure were associated with improved efficacy. Our data suggest that camrelizumab with docetaxel/cisplatin as first-line therapy was well tolerable and had potentially favorite efficacy in PD-L1-positive patients with R/M OSCC.

4.
Clin Transl Radiat Oncol ; 40: 100623, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37096116

RESUMO

Objectives: The role of re-irradiation after salvage surgery for recurrent oral cavity cancer (OCC) is controversial. We evaluated the efficacy and safety of adjuvant toripalimab (PD-1 antibody) in this patient setting. Materials and methods: In this phase II study, patients after salvage surgery with OCC occurring in an area of previously irradiated were enrolled. Patients received toripalimab 240 mg once every 3 weeks for 12 months, or combined with S-1 orally for 4-6 cycles. The primary endpoint was 1-year progression-free survival (PFS). Results: Between April 2019 and May 2021, 20 patients were enrolled. Sixty percent patients had ENE or positive margins, 80% were restaged as stage IV, and 80% were previously treated with chemotherapy. The 1-year PFS and overall survival (OS) were 58.2%, and 93.8%, respectively, for patients with CPS ≥ 1, which was significantly better than those of the real-world reference cohort (p = 0.001 and 0.019). No grade 4-5 toxicities were reported, and only one patient experienced grade 3 immune related adrenal insufficiency and discontinued treatment. The 1-year PFS and OS were significantly different for patients with CPS < 1, CPS 1-19 and CPS ≥ 20 (p = 0.011 and 0.017, respectively). The peripheral blood B cell proportion was also correlated with PD in 6 months (p = 0.044). Conclusion: Adjuvant toripalimab or combine with S-1 after salvage surgery showed improved PFS compared with a real-world reference cohort in recurrent, previously irradiated OCC, and favorable PFS were observed in patients with a higher CPS and peripheral B cell proportion. Further randomized trials are warranted.

5.
Int J Oral Sci ; 15(1): 9, 2023 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-36765028

RESUMO

Cancer cell membrane (CCM) derived nanotechnology functionalizes nanoparticles (NPs) to recognize homologous cells, exhibiting translational potential in accurate tumor therapy. However, these nanoplatforms are majorly generated from fixed cell lines and are typically evaluated in cell line-derived subcutaneous-xenografts (CDX), ignoring the tumor heterogeneity and differentiation from inter- and intra- individuals and microenvironments between heterotopic- and orthotopic-tumors, limiting the therapeutic efficiency of such nanoplatforms. Herein, various biomimetic nanoplatforms (CCM-modified gold@Carbon, i.e., Au@C-CCM) were fabricated by coating CCMs of head and neck squamous cell carcinoma (HNSCC) cell lines and patient-derived cells on the surface of Au@C NP. The generated Au@C-CCMs were evaluated on corresponding CDX, tongue orthotopic xenograft (TOX), immune-competent primary and distant tumor models, and patient-derived xenograft (PDX) models. The Au@C-CCM generates a photothermal conversion efficiency up to 44.2% for primary HNSCC therapy and induced immunotherapy to inhibit metastasis via photothermal therapy-induced immunogenic cell death. The homologous CCM endowed the nanoplatforms with optimal targeting properties for the highest therapeutic efficiency, far above those with mismatched CCMs, resulting in distinct tumor ablation and tumor growth inhibition in all four models. This work reinforces the feasibility of biomimetic NPs combining modular designed CMs and functional cores for customized treatment of HNSCC, can be further extended to other malignant tumors therapy.


Assuntos
Neoplasias de Cabeça e Pescoço , Terapia Fototérmica , Animais , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia , Xenoenxertos , Biomimética , Modelos Animais de Doenças , Neoplasias de Cabeça e Pescoço/terapia , Linhagem Celular Tumoral , Microambiente Tumoral
6.
BMC Pediatr ; 22(1): 611, 2022 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-36271345

RESUMO

BACKGROUND: Care practices for very preterm infants and the mortality and morbidity of the infants vary widely among countries and regions with different levels of economic development, including the different areas in China. We aimed to compare the obstetric and delivery room practices of two representative tertiary newborn centers in the northwestern and southern regions of China and the mortality and morbidity of their very preterm infants. METHODS: A retrospective cohort study was conducted. Very preterm infants born between 220/7 and 316/7 weeks of gestation, and admitted to Qinghai Red Cross Hospital (QHH) and Shenzhen Baoan Women's and Children's Hospital (SZH) from January 1, 2018 to December 31, 2020, were included. The infants' characteristics and short-term outcomes, and the hospitals' care practices were compared between the two cohorts. RESULTS: Three hundred and two infants in QHH and 505 infants in SZH were enrolled, and the QHH cohort was more mature than the SZH cohort was (gestational age 30.14 (29.14-31.14) vs. 29.86 (27.86-31.00 weeks, respectively), p < 0.001). Fewer antenatal steroids and more tracheal intubations were used in QHH than in SZH [(73.8% vs. 90.9%, p < 0.001) and (68.2% vs. 35.0%, p < 0.001, respectively)]. The odds of mortality [aOR = 10.31, 95%CI: (6.04, 17.61)], mortality or major morbidity [aOR = 5.95, 95%CI: (4.05, 8.74)], mortality despite active treatment [aOR = 3.14, 95%CI: (1.31, 7.53)], mortality or major morbidity despite active treatment [aOR = 3.35, 95%CI: (2.17, 5.17)], moderate or severe bronchopulmonary dysplasia [aOR = 3.66, 95%CI: (2.20, 6.06)], and severe retinopathy of prematurity [aOR = 3.24, 95%CI: (1.19, 8.83)] were higher in the QHH cohort. No significant difference in the rate of severe neurological injury or necrotizing enterocolitis ≥ Stage 2 was found between the cohorts. CONCLUSION: Obstetric and delivery room care practices used in the management of very preterm infants differed considerably between the QHH and SZH cohorts. Very preterm infants born in QHH have higher odds of mortality or severe morbidity compared with those born in SZH.


Assuntos
Doenças do Recém-Nascido , Doenças do Prematuro , Lactente , Criança , Recém-Nascido , Feminino , Humanos , Gravidez , Adulto , Lactente Extremamente Prematuro , Estudos Retrospectivos , Doenças do Prematuro/epidemiologia , Doenças do Prematuro/terapia , Recém-Nascido de muito Baixo Peso , Idade Gestacional , Estudos de Coortes , Retardo do Crescimento Fetal , Mortalidade Infantil
7.
Sci Transl Med ; 14(661): eabo5987, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-36070368

RESUMO

Head and neck squamous cell carcinoma (HNSCC) is a common and frequently lethal cancer with few therapeutic options. In particular, there are few effective targeted therapies. Development of highly effective therapeutic strategies tailored to patients with HNSCC remains a pressing challenge. To address this, we present a pharmacogenomic study to facilitate precision treatments for patients with HNSCC. We established a large collection of 56 HNSCC patient-derived cells (PDCs), which recapitulated the molecular features of the original tumors. Pharmacological assessment of HNSCCs was conducted using a three-tiered high-throughput drug screening using 2248 compounds across these PDC models and an additional 18 immortalized cell lines. We integrated genomic, transcriptomic, and pharmacological analysis to predict biomarkers, gene-drug associations, and validated biomarkers. These results supported drug repurposing for multiple HNSCC subtypes, including the JAK2 inhibitor fedratinib, for low KRT18-expressing HNSCC cases, and the topoisomerase inhibitor mitoxantrone, for IL6R-activated HNSCC cases. Our results demonstrated concordance between susceptibility predictions from the PDCs and the matched patients' responses to standard clinical medication. Moreover, we identified and experimentally confirmed that high expression of ITGB1 elicited therapeutic resistance to docetaxel and high SOD1 expression conferred resistance to afatinib. We further validated ITGB1 as a predictive biomarker for the efficacy of docetaxel therapy in a phase 2 clinical trial. In summary, our study shows that this HNSCC cell resource, as well as the resulting pharmacogenomic profiles, is effective for biomarker discovery and for guiding precision oncology therapies in HNSCCs.


Assuntos
Neoplasias de Cabeça e Pescoço , Farmacogenética , Docetaxel , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/genética , Humanos , Medicina de Precisão , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética
8.
Front Immunol ; 13: 813732, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35371031

RESUMO

Background: Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignant cancers. The treatment of HNSCC remains challenging despite recent progress in targeted therapies and immunotherapy. Research on predictive biomarkers in clinical settings is urgently needed. Methods: Next-generation sequencing analysis was performed on tumor samples from 121 patients with recurrent or metastatic HNSCC underwent sequencing analysis. Clinicopathological information was collected, and the clinical outcomes were assessed. Progression-free survival (PFS) was estimated using the Kaplan-Meier method and cox regression model was used to conduct multivariate analysis. Fisher's exact tests were used to calculate clinical benefit. A p value of less than 0.05 was designated as significant (p < 0.05). Results: Chromosome 11q13 amplification (CCND1, FGF3, FGF4, and FGF19) and EGFR mutations were significantly associated with decreased PFS and no clinical benefits after treatment with a programmed death 1 (PD-1) inhibitor. The same results were found in the combined positive score (CPS) ≥ 1 subgroup. In patients who were treated with an EGFR antibody instead of a PD-1 inhibitor, a significant difference in PFS and clinical benefits was only observed between patients with CPS ≥ 1 and CPS < 1. Conclusion: Chromosome 11q13 amplification and EGFR mutations were negatively correlated with anti-PD-1 therapy. These markers may serve as potential predictive biomarkers to identify patients for whom immunotherapy may be unsuitable.


Assuntos
Neoplasias de Cabeça e Pescoço , Imunoterapia , Biomarcadores , Receptores ErbB/genética , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Imunoterapia/métodos , Mutação , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia
9.
Signal Transduct Target Ther ; 7(1): 73, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35260570

RESUMO

Cetuximab is a widely used drug for treating head and neck squamous cell carcinomas (HNSCCs); however, it provides restricted clinical benefits, and its response duration is limited by drug resistance. Here, we conducted randomized "Phase II-like clinical trials" of 49 HNSCC PDX models and reveal multiple informative biomarkers for intrinsic resistance to cetuximab (e.g., amplification of ANKH, up-regulation of PARP3). After validating these intrinsic resistance biomarkers in another HNSCC PDX cohort (61 PDX models), we generated acquired cetuximab resistance PDX models and analyzed them to uncover resistance mechanisms. Whole exome sequencing and transcriptome sequencing revealed diverse patterns of clonal selection in acquired resistant PDXs, including the emergence of subclones with strongly activated RAS/MAPK. Extending these insights, we show that a combination of a RAC1/RAC3 dual-target inhibitor and cetuximab could overcome acquired cetuximab resistance in vitro and in vivo. Beyond revealing intrinsic resistance biomarkers, our PDX-based study shows how clonal architecture changes underlying acquired resistance can be targeted to expand the therapeutic utility of this important drug to more HNSCC patients.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neoplasias de Cabeça e Pescoço , Carcinoma de Células Escamosas de Cabeça e Pescoço , Biomarcadores , Cetuximab/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/genética , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética
10.
Am J Transl Res ; 13(5): 4949-4958, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34150079

RESUMO

OBJECTIVE: This study was designed to explore the efficacy of budesonide (BUD) in preventing and treating bronchopulmonary dysplasia (BPD) in premature infants and its effect on pulmonary function. METHODS: A total of 94 premature infants with BPD who were born in our hospital were selected as the research subjects and divided into the control group (47 cases) for routine treatment and the research group (47 cases) for BUD therapy on the basis of routine treatment according to the random number table method. The incidence of BPD and the time of oxygen inhalation, ventilator ventilation, extubation and hospitalization were recorded in the two groups. In addition, arterial blood gas indexes (arterial oxygen saturation (SaO2), arterial partial pressure of oxygen (PaO2), arterial partial pressure of carbon dioxide (PaCO2)), inflammatory response indicators (interleukin-6 (IL-6), interleukin-8 (IL-8), tumor necrosis factor (TNF-α)), pulmonary function indexes (the ratio of time taken to reach peak expiratory flow to total expiratory time (TPTEF/TE), the ratio of peak expiratory volume to total expiratory volume (VPEF/VE), tidal expiratory flow at 25%/50%/75% remaining tidal volume (TEF25, TEF50, TEF75), and the incidence of complications were observed and compared. RESULTS: After treatment, SaO2 and PaO2 increased in both groups, and their values in the research group were higher than those in the control group; PaCO2 decreased in both groups, and the PaCO2 in the research group was lower than that in the control group (P<0.05). The post-treatment TNF-α, IL-6 and IL-8 levels decreased in both groups, and their levels in the research group were lower than those in the control group (all P<0.001). TPTEE/TE, VPEF/VE, TEF25, TEF50 and TEF75 increased in both groups after treatment, and their values in the research group were higher than those in the control group (all P<0.01). The research group required shorter oxygen inhalation time, ventilator ventilation time, time to extubation and hospitalization time than the control group (all P<0.001). The incidence of BPD and other complications in the research group were lower than that in the control group (8.51%, 6.38% vs. 23.40%, 21.28%; P=0.049, 0.036). CONCLUSION: BUD is effective in the prevention and treatment of BPD in premature infants, which can effectively reduce the incidence of BPD and other complications, improve blood gas indexes, reduce inflammatory reactions and promotes good pulmonary function in children.

11.
Cell Discov ; 7(1): 32, 2021 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-33972509

RESUMO

Substitution of lysine 36 with methionine in histone H3.3 (H3.3K36M) is an oncogenic mutation that inhibits SETD2-mediated histone H3K36 tri-methylation in tumors. To investigate how the oncohistone mutation affects the function of SETD2 at the nucleosome level, we determined the cryo-EM structure of human SETD2 associated with an H3.3K36M nucleosome and cofactor S-adenosylmethionine (SAM), and revealed that SETD2 is attached to the N-terminal region of histone H3 and the nucleosome DNA at superhelix location 1, accompanied with the partial unwrapping of nucleosome DNA to expose the SETD2-binding site. These structural features were also observed in the previous cryo-EM structure of the fungal Set2-nucleosome complex. By contrast with the stable association of SETD2 with the H3.3K36M nucleosome, the EM densities of SETD2 could not be observed on the wild-type nucleosome surface, suggesting that the association of SETD2 with wild-type nucleosome might be transient. The linker histone H1, which stabilizes the wrapping of nucleosome DNA at the entry/exit sites, exhibits an inhibitory effect on the activities of SETD2 and displays inversely correlated genome distributions with that of the H3K36me3 marks. Cryo-EM analysis of yeast H3K36 methyltransferase Set2 complexed with nucleosomes further revealed evolutionarily conserved structural features for nucleosome recognition in eukaryotes, and provides insights into the mechanism of activity regulation. These findings have advanced our understanding of the structural basis for the tumorigenesis mechanism of the H3.3K36M mutation and highlight the effect of nucleosome conformation on the regulation of histone modification.

12.
Nat Commun ; 12(1): 1275, 2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33627666

RESUMO

Synthetic lethality is emerging as an important cancer therapeutic paradigm, while the comprehensive selective treatment opportunities for various tumors have not yet been explored. We develop the Synthetic Lethality Knowledge Graph (SLKG), presenting the tumor therapy landscape of synthetic lethality (SL) and synthetic dosage lethality (SDL). SLKG integrates the large-scale entity of different tumors, drugs and drug targets by exploring a comprehensive set of SL and SDL pairs. The overall therapy landscape is prioritized to identify the best repurposable drug candidates and drug combinations with literature supports, in vitro pharmacologic evidence or clinical trial records. Finally, cladribine, an FDA-approved multiple sclerosis treatment drug, is selected and identified as a repurposable drug for treating melanoma with CDKN2A mutation by in vitro validation, serving as a demonstrating SLKG utility example for novel tumor therapy discovery. Collectively, SLKG forms the computational basis to uncover cancer-specific susceptibilities and therapy strategies based on the principle of synthetic lethality.


Assuntos
Mutações Sintéticas Letais/genética , Linhagem Celular Tumoral , Cladribina/uso terapêutico , Inibidor p16 de Quinase Dependente de Ciclina/genética , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Modelos Teóricos , Mutação/genética
13.
Endocr Relat Cancer ; 26(7): 643-658, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31117050

RESUMO

Pancreatic neuroendocrine neoplasms (pNENs) are endocrine tumors arising in pancreas and is the most common neuroendocrine tumors. Mounting evidence indicates lncRNA H19 could be a determinant of tumor progression. However, the expression and mechanism of H19 and the relevant genes mediated by H19 in pNENs remain undefined. Microarray analysis was conducted to identify the differentially expressed lncRNAs in pNENs. H19 expression was analyzed in 39 paired pNEN tissues by qPCR. The biological role of H19 was determined by functional experiments. RNA pulldown, mass spectroscopy and RNA immunoprecipitation were performed to confirm the interaction between H19 and VGF. RNA-seq assays were performed after knockdown H19 or VGF. H19 was significantly upregulated in pNEN tissues with malignant behaviors, and the upregulation predicted poor prognosis in pNENs. In vitro and in vivo data showed that H19 overexpression promoted tumor growth and metastasis, whereas H19 knockdown led to the opposite phenotypes. H19 interacted with VGF, which was significantly upregulated in pNENs, and higher VGF expression was markedly related to poor differentiation and advanced stage. Furthermore, VGF was downregulated when H19 was knocked down, and VGF promoted cell proliferation, migration and invasion. Mechanistic investigations revealed that H19 activated PI3K/AKT/CREB signaling and promoted pNEN progression by interacting with VGF. These findings indicate that H19 is a promising prognostic factor in pNENs with malignant behaviors and functions as an oncogene via the VGF-mediated PI3K/AKT/CREB pathway. In addition, our study implies that VGF may also serve as a candidate prognostic biomarker and therapeutic target in pNENs.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fatores de Crescimento Neural/metabolismo , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/metabolismo , Adulto , Idoso , Animais , Progressão da Doença , Feminino , Humanos , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Transdução de Sinais , Células Tumorais Cultivadas
14.
Carbohydr Polym ; 204: 111-123, 2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-30366522

RESUMO

Pancreatic ductal adenocarcinoma is a highly malignant gastrointestinal tumor. Molecular targeting therapy for pancreatic cancer is still limited. High expressed Galectin-3 in pancreatic cancer is positively correlated with disease progression, indicating that Galectin-3 can be employed as a predictor of poor prognosis. From safflower, we isolated and purified a homogeneous polysaccharide, HH1-1, which could bind to and inhibit Galectin-3. HH1-1 could block the interaction between Galectin-3 and EGFR. Following HH1-1 treatment, the binding ability between EGFR and Galectin-3 was reduced by 245.28 folds. HH1-1 could suppress pancreatic cancer cell proliferation, arrest the cell cycle in S phase, induce cell apoptosis, inhibit angiogenesis and impede tumor cell migration and invasion. Moreover, HH1-1 affected the Galectin-3/EGFR/AKT/FOXO3 signaling pathway and possessed anti-pancreatic cancer activity in vitro and in vivo, especially in patient-derived xenografts. Further study suggested that HH1-1 had almost no toxicity both in vitro and in vivo. This adds new evidence to suggest that HH1-1 could be a promising therapeutic agent and support the pursuit of the Galectin-3 as a target in pancreatic cancer treatment.


Assuntos
Antineoplásicos/uso terapêutico , Galactanos/uso terapêutico , Galectina 3/antagonistas & inibidores , Neoplasias Pancreáticas/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Sanguíneas , Carthamus tinctorius/química , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/metabolismo , Proteína Forkhead Box O3/metabolismo , Galactanos/farmacologia , Galectina 3/metabolismo , Galectinas , Humanos , Camundongos Nus , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos
15.
Polymers (Basel) ; 9(10)2017 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-30965840

RESUMO

RN1, a polysaccharide from flowers of Panax pseudo-ginsieng Wall. Var. notoginseng (Burkill) Hoo & Tseng, is a potential multi-targeting drug candidate for pancreatic cancer treatment. However, the active targeting domain of RN1 is still unknown. Herein, three RN1 derived branches were synthesized via [3+2] or [2+2] strategies, efficiently. Two pentasaccharides, 18 and 27, showed similar inhibition effect on pancreatic cancer BxPC-3 cells to that of RN1 at same concentration. Interestingly, tetrasaccharide 21 potently inhibited gemcitabineresistant cell line Panc-1 at high concentration. These suggest that the branches of RN1 might be the active targeting domain and tetrasaccharide 21 might be a potential leading compound for pancreatic cancer with gemcitabine resistance.

16.
Cancer Lett ; 382(1): 44-52, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27569654

RESUMO

Fucoidan may inhibit angiogenesis. However, its functional target molecule and the underlying mechanism are still vague. In the present study, we showed that sulfated fucoidan FP08S2 from Sargassum fusiforme inhibited tube formation as well as migration and invasion of human microvascular endothelial cells (HMEC-1). In addition, FP08S2 was confirmed to disrupt VEGF-induced angiogenesis both in vitro and in vivo. Further study indicated that FP08S2 could bind to both VEGF and VEGFR2 to interfere with VEGF-VEGFR2 interaction. Moreover, VEGFR2/Erk/VEGF signaling pathway was blocked by FP08S2 in HMEC-1 cells. Importantly, FP08S2 impeded the growth and microvessel formation of A549 cancer cell xenograft in nude mice. These results suggested that FP08S2 presented remarkable anti-angiogenic activity via blocking VEGF signaling and could be a potential novel leading compound to inhibit lung cancer cell growth.


Assuntos
Inibidores da Angiogênese/farmacologia , Proliferação de Células/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neovascularização Patológica , Polissacarídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Células A549 , Animais , Movimento Celular/efeitos dos fármacos , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Relação Dose-Resposta a Droga , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Neovascularização Fisiológica/efeitos dos fármacos , Fatores de Tempo , Carga Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Sci Rep ; 6: 28833, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27349455

RESUMO

Gallbladder carcinoma (GBC) is a highly lethal malignancy of the gastrointestinal tract. Despite extensive research, the underlying molecular mechanism of GBC remains largely unclear. Stathmin 1 (STMN1) is an important cytosolic protein associated with microtubule stability that was reported to be involved in tumorigenesis. Up to our knowledge, its role in gallbladder carcinoma has not been analyzed. In this study, we found that STMN1 was significantly highly expressed in GBC by immunohistochemistry (IHC). Further research demonstrated that silencing of STMN1 inhibited cell growth in vitro. Moreover, knockdown of STMN1 induced apoptosis and delayed G2/M phase transformation in GBC cells. Our data support a rationale for further studies that the silencing of STMN1 may regulate the activity of p38 MAPK kinase and p53/p21 signal pathway. Besides, xenografted gallbladder carcinoma cells growth were significantly impaired after STMN1 was silenced in vivo. These results suggested that STMN1 played an important role in cell proliferation and migration. This provided a potential clue for investigating the therapeutic target in GBC.


Assuntos
Proliferação de Células/genética , Neoplasias da Vesícula Biliar/genética , Interferência de RNA , Estatmina/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação para Baixo , Feminino , Neoplasias da Vesícula Biliar/metabolismo , Neoplasias da Vesícula Biliar/terapia , Humanos , Imuno-Histoquímica , Camundongos Endogâmicos BALB C , Terapêutica com RNAi/métodos , Estatmina/biossíntese , Carga Tumoral/genética
18.
Oncotarget ; 7(18): 26388-99, 2016 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27049919

RESUMO

Malignant gliomas are the most common form of intrinsic primary brain tumors worldwide. Alterations in microRNAs play a role in highly invasive malignant glioma, but detail mechanism still unknown. In this study, the role and mechanism of microRNA-149 (miR-149) in glioma are investigated. We show that miR-149 is expressed at substantially higher levels in glioma than in normal tissues. Stable overexpression of miR-149 augments potent prosurvival activity, as evidenced by promotion of cell viability, inhibition of apoptosis, and induced xenografted tumor growth in vivo. We further show that Caspase-2 is identified as a functional target of miR-149 and expression of caspase-2 is inversely associated with miR-149 in vitro. In addition, miR-149 promotes tumor survival in the U87-MG and A172 cell lines and it targets caspase-2 via inactivation of the p53 and p21 pathways. There results support a special role for miR-149 by targeting Caspase-2 to impact on p53 signaling pathway. We speculate that miR-149 has distinct biological functions in p53 wild type cells and p53 mutation cells, and the mechanisms involved remain to be explored in future. Our study suggests that targeting miR-149 may be a novel therapy strategy for treating p53 wild type glioma tumors in humans.


Assuntos
Neoplasias Encefálicas/patologia , Caspase 2/metabolismo , Cisteína Endopeptidases/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Glioma/patologia , MicroRNAs/metabolismo , Adulto , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Caspase 2/genética , Cisteína Endopeptidases/genética , Progressão da Doença , Feminino , Glioma/genética , Glioma/metabolismo , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade
19.
Anal Bioanal Chem ; 387(1): 303-9, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17089098

RESUMO

Different types of carbon nanotube material (single-walled carbon nanotubes (SWCNTs) and multi-walled carbon nanotubes (MWCNTs) of different internal diameter) have been used for preparation of CNT-modified glassy-carbon electrodes. Redox reactions involving ferricyanide and hydrogen peroxide were examined at the CNT-modified electrodes. Electrodes modified with SWCNTs usually had better electron-transfer properties than MWCNT-modified electrodes. Glucose biosensors were also prepared with electropolymerized polyphenylenediamine films, CNT materials, and glucose oxidase. Amperometric behavior in glucose determination was examined. SWCNT-modified glucose biosensors usually had a wider dynamic range (from 0.1 to 5.5 mmol L-1) and greater sensitivity in glucose determination. The detection limit was estimated to be 0.05 mmol L-1.


Assuntos
Técnicas Biossensoriais/métodos , Condutividade Elétrica , Glucose/análise , Nanotubos de Carbono/química , Catálise , Eletroquímica , Eletrodos , Ferricianetos/química , Glucose/química , Glucose Oxidase/química , Peróxido de Hidrogênio/química , Oxirredução , Fenilenodiaminas/química , Reprodutibilidade dos Testes
20.
Shanghai Kou Qiang Yi Xue ; 12(5): 331-3, 2003 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-14966604

RESUMO

OBJECTIVE: To investigate the changes of tooth pain in patients with orthodontic tooth movement, to study the changes of bioactivators in GCF in patients with orthodontic tooth movement, and explore the correlation between pain and bioactivators in orthodontic patients. METHODS: 50 patients were included, each having one treatment tooth and one contralateral control tooth. The levels of PGE(2), P substances, IL-6 and GM-CSF in GCF of upper lateral incisor were investigated before activation and at 12th hour, 24th hour, 48th hour and 72th hour after the applying labial orthodontic force, by using highly sensitive radioimmunassay. To record the changes of pain in orthodontic patients at 12th hour, 24th hour,48th hour and 72th hour after applying stress. RESULTS: At experimental sides, total amount of GCF PGE(2), P substances(SP), IL-6 and GM-CSF levels was significantly elevated compared with that before activation. The concentration of PGE(2) and SP in GCF increased significantly and reached peak point at 24th hour in patients, while the levels of IL-6 and GM-CSF in GCF remained at higher baseline through the experiment. There was an rhythm change of periodontal pain during orthodontic tooth movement. CONCLUSION: There was an rhythm changes of pain after stress in patients with orthodontic tooth movement, and the rhythm pain was correlated to the changes of some activator such as P substances and PGE(2) in periodontal ligament.


Assuntos
Líquido do Sulco Gengival/química , Ortodontia Corretiva/efeitos adversos , Odontalgia/etiologia , Adolescente , Adulto , Criança , Dinoprostona/análise , Fator Estimulador de Colônias de Granulócitos e Macrófagos/análise , Humanos , Interleucina-6/análise , Estresse Mecânico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA