Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Fish Dis ; 44(11): 1865-1873, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34287946

RESUMO

Galectin-9, an important pathogen recognition receptor (PRR), could recognize and bind pathogen-associated molecular patterns (PAMPs) on the surface of invading microorganisms, initiating the innate immune responses. A galectin-9 was identified from Qihe crucian carp Carassius auratus and designated as CaGal-9. The predicted CaGal-9 protein contained two non-identical carbohydrate recognition domains (CRDs), namely, N-CRD and C-CRD. The recombinant proteins (rCaGal-9, rN-CRD and rC-CRD) were purified from Escherichia coli BL21 (DE3) and exhibited strong agglutinating activity with erythrocytes of rabbit. The haemagglutination was inhibited by D-galactose, α-lactose and N-acetyl-D-galactose. Results of microbial agglutination assay showed that three recombinant proteins agglutinated Gram-negative bacterium Aeromonas hydrophila and Gram-positive bacterium Staphylococcus aureus. With regard to the binding activity, each recombinant protein could bind to LPS, PGN and the examined microorganisms (A. hydrophila and S. aureus) with different binding affinities. The integrated analyses suggested that CaGal-9 with two CRD domains could play an important role in immune defence against pathogenic microorganisms for C. auratus.


Assuntos
Carpas/imunologia , Proteínas de Peixes/genética , Galectinas/genética , Aeromonas hydrophila , Aglutinação , Sequência de Aminoácidos , Animais , Carpas/genética , Proteínas de Peixes/imunologia , Galectinas/imunologia , Imunidade Inata , Moléculas com Motivos Associados a Patógenos , Proteínas Recombinantes/imunologia , Alinhamento de Sequência , Staphylococcus aureus
2.
Br J Cancer ; 123(11): 1616-1624, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32958820

RESUMO

BACKGROUND: To report the long-term outcomes of a phase III trial designed to test two hypotheses: (1) elective nodal irradiation (ENI) is superior to conventional field irradiation (CFI), and (2) chemoradiotherapy plus erlotinib is superior to chemoradiotherapy in locally advanced oesophageal squamous cell cancer (ESCC). METHODS: Patients with locally advanced ESCC were randomly assigned (1:1:1:1 ratio) to one of the four groups: A: radiotherapy adoption of ENI with two cycles of concurrent TP chemotherapy (paclitaxel and cisplatin) plus erlotinib; B: radiotherapy adoption of ENI with two cycles of concurrent TP; C: radiotherapy adoption of CFI with two cycles of concurrent TP plus erlotinib and D: radiotherapy adoption of CFI with two cycles of concurrent TP. A total of 60 Gy of radiation doses was delivered over 30 fractions. We explored the impact of epidermal growth factor receptor (EGFR) expression on the efficacy of erlotinib plus chemoradiotherapy. RESULTS: A total of 352 patients (88 assigned to each treatment group) were enrolled. The 5-year survival rates were 44.9%, 34.8%, 33.8% and 19.6% in groups A, B, C and D, respectively (P = 0.013). ENI significantly improved OS compared with standard CFI (median, 38.5 vs 22.6 months; HR, 0.74; P = 0.018). The addition of erlotinib significantly improved OS (median, 39.4 vs 27.4 months; HR, 0.75; P = 0.025). Patients with overexpressing EGFR treated with erlotinib had a better OS and PFS than those without erlotinib. CONCLUSIONS: Concurrent chemoradiotherapy with ENI and/or erlotinib improved long-term survival in locally advanced ESCC. CLINICAL TRIAL REGISTRATION: Trial registration: NCT00686114.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Quimiorradioterapia/métodos , Cloridrato de Erlotinib/administração & dosagem , Neoplasias Esofágicas/terapia , Carcinoma de Células Escamosas do Esôfago/terapia , Adulto , Idoso , Cisplatino/administração & dosagem , Feminino , Humanos , Linfonodos/efeitos da radiação , Masculino , Pessoa de Meia-Idade , Paclitaxel/administração & dosagem
3.
BMC Cancer ; 20(1): 278, 2020 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-32252680

RESUMO

BACKGROUND: There is no consensus on the therapeutic approach to ECOG 2 patients with locally advanced non-small-cell lung cancer (LA-NSCLC), despite the sizable percentage of these patients in clinical practice. This study focused on the efficacy, toxicity and the optimal chemotherapy regimen of CCRT in ECOG 2 patients in a phase III trial. METHODS: Patients capable of all self-care with bed rest for less than 50% of daytime were classified as ECOG 2 subgroup. A subgroup analysis was performed for ECOG 2 patients recruited in the phase III trial receiving concurrent EP (etoposide + cisplatin)/PC (paclitaxel + carboplatin) chemotherapy with intensity-modulated radiation therapy (IMRT) or three-dimensional conformal external beam radiation therapy (3D-CRT). RESULTS: A total of 71 ECOG 2 patients were enrolled into the study. Forty-six (64.8%) patients were treated with IMRT technique. The median overall survival (OS) and progression free survival (PFS) for ECOG 2 patients were 16.4 months and 9 months, respectively. No difference was observed in treatment compliance and toxicities between ECOG 2 patients and ECOG 0-1 patients. Within the ECOG 2 group (31 in the EP arm and 40 in the PC arm), median OS and 3-year OS were 15.7 months and 37.5% for the EP arm, and 16.8 months and 7.5% for the PC arm, respectively (p = 0.243). The incidence of grade ≥ 3 radiation pneumonitis was higher in the PC arm (17.5% vs. 0.0%, p = 0.014) with 5 radiation pneumonitis related deaths, while the incidence of grade 3 esophagitis was numerically higher in the EP arm (25.8% vs. 10.0%, p = 0.078). CONCLUSIONS: CCRT provided ECOG 2 patients promising outcome with acceptable toxicities. EP might be superior to PC in terms of safety profile in the setting of CCRT for ECOG 2 patients. Prospective randomized studies based on IMRT technique are warranted to validate our findings. TRIAL REGISTRATION: ClinicalTrials.gov registration number: NCT01494558. (Registered 19 December 2011).


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/terapia , Quimiorradioterapia/mortalidade , Neoplasias Pulmonares/terapia , Adulto , Idoso , Carboplatina/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/patologia , Quimiorradioterapia/métodos , Cisplatino/administração & dosagem , Ensaios Clínicos Fase III como Assunto , Etoposídeo/administração & dosagem , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Estudos Multicêntricos como Assunto , Paclitaxel/administração & dosagem , Prognóstico , Estudos Prospectivos , Ensaios Clínicos Controlados Aleatórios como Assunto , Taxa de Sobrevida
4.
BMC Genomics ; 20(1): 611, 2019 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-31345182

RESUMO

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) cells are heterogeneous, easily develop radioresistance, and recur. Single-cell RNA-seq (scRNA-seq) is a next-generation sequencing method that can delineate diverse gene expression profiles of individual cells and mining their heterogeneous behaviors in response to irradiation. Our aim was using scRNA-seq to describe the difference between parental cells and cells that acquired radioresistance, and to investigate the dynamic changes of the transcriptome of cells in response to FIR. RESULTS: We sequenced ESCC cell lines KYSE180 with and without fractionated irradiation (FIR). A total of 218 scRNA-seq libraries were obtained from 88 cells exposed to 12 Gy (KYSE-180-12 Gy), 89 exposed to 30 Gy (KYSE-180-30 Gy), and 41 parental KYSE-180 cells not exposed to FIR. Dynamic gene expression patterns were determined by comprehensive consideration of genes and pathways. Biological experiments showed that KYSE-180 cells became radioresistant after FIR. PCA analysis of scRNA-seq data showed KYSE-180, KYSE-180-12 Gy and KYSE-180-30 Gy cells were discrete away from each other. Two sub-populations found in KYSE-180-12 Gy and only one remained in KYSE-180-30 Gy. This sub-population genes exposure to FIR through 12 Gy to 30 Gy were relevant to the PI3K-AKT pathway, pathways evading apoptosis, tumor cell migration, metastasis, or invasion pathways, and cell differentiation and proliferation pathways. We validated DEGs, such as CFLAR, LAMA5, ITGA6, ITGB4, and SDC4 genes, in these five pathways as radioresistant genes in bulk cell RNA-seq data from ESCC tissue of a ESCC patient treated with radiotherapy and from KYSE-150 cell lines. CONCLUSIONS: Our results delineated the divergent gene expression patterns of individual ESCC cells exposure to FIR, and displayed genes and pathways related to development of radioresistance.


Assuntos
Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Tolerância a Radiação , Transcriptoma , Linhagem Celular Tumoral/efeitos da radiação , Humanos , Redes e Vias Metabólicas , RNA-Seq
5.
Oncologist ; 24(4): 461-e136, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30850562

RESUMO

LESSONS LEARNED: Definitive concurrent chemoradiotherapy based on oxaliplatin and endostatin was effective with the objective response rate exceeding 80%, and the treatment-related toxicities were acceptable.The treatment compliance of the current combination was much higher, without significant reduction in survival outcomes, than historical reports. BACKGROUND: This phase II trial aimed at assessing the efficiency and safety of definitive concurrent chemoradiotherapy (dCRT) using oxaliplatin (OHP) and endostatin in patients with inoperable esophageal squamous cell carcinoma (ESCC). METHODS: Radiotherapy was delivered with a daily fraction of 2.0 Gy to a total dose of 60.0 Gy over 6 weeks. Endostatin and OHP were both intravenously administered at doses of 7.5 mg/m2 daily for 2 weeks and 135 mg/m2 on day 1, respectively, every 3 weeks. The primary endpoint was the objective response rate (ORR). RESULTS: The analysis included 37 patients. The median age was 63 years (range: 49-71 years), and all patients were stage III-IVA. Of these patients, 97.3% (36/37) completed the dCRT course with an ORR of 83.8%, including 10 (27.0%) patients with complete response and 21 (56.8%) patients with partial response. The median overall survival (OS) time was 18.5 months (95% confidence interval [CI]: 10.6-26.4) with a 2-year OS rate of 39.6% (95% CI: 0.202-0.590). The median progression-free survival (PFS) time was 11.5 months (95% CI: 7.6-15.4) with a 2-year PFS rate of 20.2% (95% CI: 0.049-0.355). Grade 3 toxicities included esophagitis (five patients) and leukocytopenia (three patients). Grade 4 leukopenia was observed in one patient. Late toxicity was infrequent, and no treatment-related death occurred. Posttreatment dysphagia scores were significantly improved when compared with baseline (p < .001). CONCLUSION: dCRT based on OHP and endostatin resulted in high treatment compliance with manageable toxicities. This combination resulted in encouraging ORR without compromising survival outcomes. It should be validated in future clinical studies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimiorradioterapia/métodos , Neoplasias Esofágicas/terapia , Carcinoma de Células Escamosas do Esôfago/terapia , Idoso , Estudos de Casos e Controles , Endostatinas/administração & dosagem , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/patologia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Oxaliplatina/administração & dosagem , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida
6.
Cell Mol Life Sci ; 74(5): 869-880, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27738745

RESUMO

Single-cell sequencing (SCS) is a fast-growing, exciting field in genomic medicine. It enables the high-resolution study of cellular heterogeneity, and reveals the molecular basis of complicated systems, which facilitates the identification of new biomarkers for diagnosis and for targeting therapies. It also directly promotes the next generation of genomic medicine because of its ultra-high resolution and sensitivity that allows for the non-invasive and early detection of abnormalities, such as aneuploidy, chromosomal translocation, and single-gene disorders. This review provides an overview of the current progress and prospects for the diagnostic applications of SCS, specifically in pre-implantation genetic diagnosis/screening, non-invasive prenatal diagnosis, and analysis of circulating tumor cells. These analyses will accelerate the early and precise control of germline- or somatic-mutation-based diseases, particularly single-gene disorders, chromosome abnormalities, and cancers.


Assuntos
Técnicas de Diagnóstico Molecular/métodos , Análise de Sequência de DNA/métodos , Análise de Célula Única/métodos , Pesquisa Translacional Biomédica , Animais , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Medicina de Precisão , Diagnóstico Pré-Implantação
7.
Cancer Sci ; 108(9): 1828-1833, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28665042

RESUMO

Elective nodal irradiation (ENI) might improve overall survival in patients with inoperable esophageal cancer. We conducted a retrospective analysis to assess the long-term survival and toxicity of esophageal cancer patients treated with ENI versus conventional-field irradiation (CFI). All data in the present study were based on our institutional experience from 2000 to 2005 of patients with inoperable esophageal cancer treated with ENI or CFI plus two concurrent cycles of paclitaxel/cisplatin. Based on the inclusion and exclusion criteria, 89 patients were included in the analysis. Of these patients, 51 were treated with ENI, whereas 38 were treated with CFI. For the per-protocol population, the patients in the ENI group significantly improved in terms of their 10-year disease-specific overall survival (43.1% vs 10.5%, P = 0.019), 10-year disease-free survival (36.7% vs 10.2%, P = 0.040) and 10-year local recurrence-free survival (47.2% vs 17.2%, P = 0.018) compared with the CFI group. Aside from radiation esophagitis, the incidence of grade 3 or greater acute toxicities did not differ between the two groups. Multivariate analysis showed that radiation field, tumor length and clinical stage were independent prognostic factors associated with OS. Concurrent chemoradiotherapy with ENI improves both disease-specific overall survival and loco-regional control in patients with inoperable esophageal cancer receiving per-protocol treatment. The regimen has a manageable tolerability profile.


Assuntos
Adenocarcinoma/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Escamosas/terapia , Neoplasias Esofágicas/terapia , Adenocarcinoma/mortalidade , Adenocarcinoma/secundário , Idoso , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/secundário , Quimiorradioterapia , Cisplatino/administração & dosagem , Intervalo Livre de Doença , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Feminino , Humanos , Estimativa de Kaplan-Meier , Linfonodos/patologia , Irradiação Linfática , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Paclitaxel/administração & dosagem , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Terapia de Salvação , Resultado do Tratamento
8.
Lancet ; 388(10054): 1883-1892, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27567279

RESUMO

BACKGROUND: Outcomes are poor for patients with recurrent or metastatic nasopharyngeal carcinoma and no well established first-line chemotherapy is available for the disease. We compared the efficacy and safety of gemcitabine plus cisplatin versus fluorouracil plus cisplatin in patients with recurrent or metastatic nasopharyngeal carcinoma. METHODS: In this multicentre, randomised, open-label, phase 3 trial, patients with recurrent or metastatic nasopharyngeal carcinoma were recruited from 22 hospitals in China. Key inclusion criteria were Eastern Cooperative Oncology Group performance status of 0 or 1, adequate organ function, and measurable lesions according to Response Evaluation Criteria in Solid Tumors version 1.1. Patients were randomly assigned in a 1:1 ratio to receive either gemcitabine (1 g/m2 intravenously on days 1 and 8) and cisplatin (80 mg/m2 intravenously on day 1), or fluorouracil (4 g/m2 in continuous intravenous infusion over 96 h) and cisplatin (80 mg/m2 on day 1 given intravenously) once every 3 weeks for a maximum of six cycles. The randomisation was done centrally via an interactive phone response system using block randomisation with a size of six. The primary endpoint was progression-free survival assessed by the independent image committee in the intention-to-treat population. Safety analyses were done in patients who received at least one cycle of study drug. This study is ongoing and is registered with ClinicalTrials.gov, number NCT01528618. FINDINGS: Between Feb 20, 2012, and Oct 30, 2015, 362 patients were randomly assigned to a group (181 to the gemcitabine [plus cisplatin] group and 181 to the fluorouracil [plus cisplatin] group). Median follow-up time for progression-free survival was 19·4 months (IQR 12·1-35·6). The median progression-free survival was 7·0 months (4·4-10·9) in the gemcitabine group and 5·6 months (3·0-7·0) in the fluorouracil group (hazard ratio [HR] 0·55 [95% CI 0·44-0·68]; p<0·0001). A total of 180 patients in the gemcitabine group and 173 patients in the fluorouracil group were included in the safety analysis. Significantly different treatment-related grade 3 or 4 adverse events between the gemcitabine and fluorouracil groups were leucopenia (52 [29%] vs 15 [9%]; <0·0001), neutropenia (41 [23%] vs 23 [13%]; p=0·0251), thrombocytopenia (24 [13%] vs three [2%]; p=0·0007), and mucosal inflammation (0 vs 25 [14%]; <0·0001). Serious treatment-related adverse events occurred in seven (4%) patients in the gemcitabine group and ten (6%) in the fluorouracil group. Six (3%) patients in the gemcitabine group and 14 (8%) patients in the fluorouracil group discontinued treatment because of drug-related adverse events. No treatment-related deaths occurred in either group. INTERPRETATION: Gemcitabine plus cisplatin prolongs progression-free survival in patients with recurrent or metastatic nasopharyngeal carcinoma. The results establish gemcitabine plus cisplatin as the standard first-line treatment option for this population. FUNDING: Sun Yat-Sen University Clinical Research 5010 Programme, Chinese National Natural Science Foundation project (grant numbers 81372502 and 81201917), the National High Technology Research and Development Program of China (863 program numbers 2012AA02A501 and 2012AA02A502), and the Natural Science Foundation of Guangdong (grant number S2013010016564).


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Nasofaríngeas/tratamento farmacológico , Neoplasias Nasofaríngeas/patologia , Adulto , Idoso , Carcinoma , China , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Desoxicitidina/administração & dosagem , Desoxicitidina/efeitos adversos , Desoxicitidina/análogos & derivados , Intervalo Livre de Doença , Esquema de Medicação , Feminino , Fluoruracila/administração & dosagem , Humanos , Infusões Intravenosas , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Razão de Chances , Seleção de Pacientes , Fatores de Risco , Resultado do Tratamento , Gencitabina
9.
Mol Carcinog ; 56(3): 1150-1163, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27769097

RESUMO

Previous studies on the mechanisms underlying ESCC (esophageal squamous cell carcinoma) chemoresistance only focused on tumor cells while tumor microenvironment has been completely ignored. Our study aimed to clarify the effect of CAFs (cancer-associated fibroblasts), one major component of tumor microenvironment, on the chemoresistance of ESCC. By primary culture, two pairs of CAFs and matched NFs (normal fibroblasts) were isolated from tumor tissues of ESCC patients and matched normal esophageal epithelial tissues, respectively. The association of CAFs and chemoresistance was assessed in esophageal carcinoma cells, in xenograft tumor models and in clinical specimens of ESCC patients. We found CAFs conferred ESCC cells significant resistance to several common chemotherapeutic drugs including cisplatin, taxol, irinotecan (CPT-11), 5-fluorouracil (5-Fu), carboplatin, docetaxel, pharmorubicin, and vincristine. Mechanism studies revealed that blockage of CAFs-secreted TGFß1 signaling by its receptor TGFßR1 inhibitor LY2157299 significantly reversed the chemoresistance in vitro and in vivo. Furthermore, the crosstalk of CAFs and ESCC cells enhanced the expression and activation of FOXO1, a member of the forkhead transcription factors in the O-box sub-family, inducing TGFß1 expression in an autocrine/paracrine signaling loop. In 130 ESCC patients, the expression of TGFß1 in CAFs was significantly associated with overall survival of patients treated with chemoradiotherapy. Together, our study highlighted TGFß1 expressed in CAFs as an attractive target to reverse tumor chemoresistance, and can be used as an independent prognostic factor of ESCC patients treated with chemoradiotherapy. © 2016 Wiley Periodicals, Inc.


Assuntos
Antineoplásicos/administração & dosagem , Fibroblastos Associados a Câncer/efeitos dos fármacos , Carcinoma de Células Escamosas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Neoplasias Esofágicas/tratamento farmacológico , Proteína Forkhead Box O1/metabolismo , Pirazóis/administração & dosagem , Quinolinas/administração & dosagem , Fator de Crescimento Transformador beta1/metabolismo , Adulto , Idoso , Animais , Antineoplásicos/farmacologia , Fibroblastos Associados a Câncer/metabolismo , Carcinoma de Células Escamosas/metabolismo , Comunicação Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Pirazóis/farmacologia , Quinolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Tumour Biol ; 39(6): 1010428317705334, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28618934

RESUMO

Curcumin is a potent anti-cancer drug in several types of human cancers. Despite of several preclinical and clinical studies of curcumin, the precise mechanism of curcumin in cancer prevention has remained unclear. In our study, we for the first time investigated whole transcriptome alteration in A549 non-small cell lung cancer (NSCLC) cell lines after treatment with curcumin using RNA sequencing. We found that lots of genes and signaling pathways were significantly altered after curcumin treatment in A549 cells. With bioinformatics approaches (gene ontology, Kyoto Encyclopedia of Genes and Genomes, and STRING), we found that those curcumin altered genes were not only the genes that induce cell death but also those extracellular matrix receptors and mitogen-activated protein kinase signaling pathway genes which regulate cell migration and proliferation. Among those significantly altered genes, eight genes ( COL1A1, COL4A1, COL5A1, LAMA5, ITGA3, ITGA2B, DDIT3, and DUSP1) were further examined by quantitative reverse transcription polymerase chain reaction and western blot analysis in four non-small cell lung cancer cell lines. Both in cell lines and in mouse model, the extracellular matrix receptors including the integrin ( ITGA3 and ITGA2B), collagen ( COL5A1), and laminin ( LAMA5) were significantly inhibited by curcumin at messenger RNA and protein levels. Functional studies confirmed that curcumin not only induced A549 cell death but also repressed cell proliferation and migration by regulating extracellular matrix receptors. Collectively, our study suggests that curcumin may be used as a promising drug candidate for intervening lung cancer in future studies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Colágeno Tipo V/biossíntese , Curcumina/administração & dosagem , Integrina alfa2/biossíntese , Integrina alfa3/biossíntese , Laminina/biossíntese , Células A549 , Animais , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo V/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Integrina alfa2/genética , Integrina alfa3/genética , Laminina/genética , Camundongos , RNA Mensageiro/biossíntese , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Dis Esophagus ; 30(2): 1-9, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26918886

RESUMO

Concurrent chemoradiotherapy (CCRT) has been accepted as the standard non-surgical treatment for esophageal cancer. However, no consistent conclusions have been reached whether elective lymph node irradiation (ENI) should be delivered. Therefore, we performed a systematic review and pooled analysis to evaluate the value of CCRT with ENI. A literature search based on PubMed, Embase and Google Scholar was carried out and all of the studies were evaluated carefully regarding with survival outcomes, response rates, patterns of failure rates and acute/late toxicities. Twenty-two studies were identified based on the criteria: median overall survival time was 21.0 months; pooled response rates were 56.8% (CR) and 85.8% (CR+PR), respectively; residual disease rate, local-regional recurrence rate, distant failure rate and both (local-regional recurrence plus distant failure) rate was 28%, 21%, 11%, and 7%, respectively; hematologic toxicities were the most sever acute toxicities and esophagus-related toxicity was the most common radiation-related toxicity both in acute (15.7%) and late (6.2%) phase. In conclusion, ENI is feasible with acceptable toxicities in esophageal carcinoma and the efficacy should be verified in randomized trials.


Assuntos
Carcinoma de Células Escamosas/terapia , Quimiorradioterapia/métodos , Neoplasias Esofágicas/terapia , Linfonodos/efeitos da radiação , Adulto , Idoso , Carcinoma de Células Escamosas/patologia , Quimiorradioterapia/efeitos adversos , Intervalo Livre de Doença , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Feminino , Humanos , Linfonodos/patologia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Lesões por Radiação/epidemiologia , Lesões por Radiação/etiologia , Dosagem Radioterapêutica , Resultado do Tratamento
12.
Biochem Biophys Res Commun ; 471(4): 596-602, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26876573

RESUMO

Circulating tumor DNA (ctDNA) is becoming an important biomarker in noninvasive diagnosis and monitoring of tumor dynamics. This study tested the feasibility of plasma ctDNA for the non-invasive analysis of tumor mutations in esophageal squamous cell carcinoma (ESCC) by sequencing of tumor, tumor-adjacent, and normal tissue, as well as pre-surgery and post-surgery plasma. Exome sequencing of eight patients identified between 29 and 134 somatic mutations in ESCCs, many of which were also determined in ctDNA. Comparison of pre-surgery and post-surgery plasma has shown that mutations had reduced frequency or disappeared after surgery treatment. We further evaluated the TruSight Cancer sequencing panel by using it to detect mutations in the plasma of three patients. Tumor mutations were only found in one of them. To design a sequencing panel with improved targeting, we identified significantly mutated genes by meta-analysis of 532 ESCC genomes. Our results confirmed the well-known driver genes and found several uncharacterized genes. The new panel consisted of 90 recurrent genes, which theoretically achieved 94% and 75% of sensitivity when detecting at least 1 and 2 mutant genes in ESCC patients, respectively. Our results demonstrate the feasibility of using ctDNA to detect ESCCs and monitor treatment effect. The low-cost and sensitive target panel could facilitate clinical usage of ctDNA as a noninvasive biomarker.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/diagnóstico , DNA de Neoplasias/genética , Neoplasias Esofágicas/diagnóstico , Biomarcadores Tumorais/sangue , Carcinogênese/genética , Carcinoma de Células Escamosas/sangue , Carcinoma de Células Escamosas/genética , Análise Mutacional de DNA/métodos , DNA de Neoplasias/sangue , Neoplasias Esofágicas/sangue , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago , Genes Neoplásicos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mutação , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/genética
13.
Tumour Biol ; 37(8): 10097-105, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26820130

RESUMO

This study is aimed to investigate the differentially expressed long non-coding RNAs (lncRNAs) in colorectal cancer and its potential biological function. Colorectal adenoma is the precancerous lesions of colorectal cancer, so in this study, we used colorectal adenoma as negative control. The global lncRNA expression profile in colorectal cancer and adenoma was evaluated by bioinformatics. The biological functions and potential mechanism of AK027294 were investigated in HCT116, HCT8, and SW480 colorectal cancer cells. A total of 135 lncRNAs were found to be differentially expressed in colorectal cancer and adenoma tissues. Among them, 71 lncRNAs were up-regulated and 64 lncRNAs were down-regulated. Especially, AK027294 was found to be highly expressed in colorectal cancer tissues compared with colorectal adenoma tissues (fold change is 184.5). Our results indicated that AK027294 down-regulation significantly inhibited colorectal cancer cells proliferation and migration, but promoted cell apoptosis (P < 0.05). The potential mechanism of AK027294 might be associated with the regulation of caspase-3, caspase-8, Bcl-2, MMP12, MMP9, and TWIST. The lncRNA expression profile in colorectal cancer suggests lncRNAs may play important roles in the occurrence and progression of colorectal cancer. AK027294 is highly expressed in colorectal cancer and closely correlates with colorectal cells proliferation, migration, and apoptosis.


Assuntos
Adenocarcinoma/patologia , Neoplasias Colorretais/patologia , RNA Longo não Codificante/genética , RNA Neoplásico/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Apoptose , Ciclo Celular , Divisão Celular , Linhagem Celular Tumoral , Movimento Celular , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Replicação do DNA , Perfilação da Expressão Gênica , Humanos , Invasividade Neoplásica , Proteínas de Neoplasias/fisiologia , Interferência de RNA , RNA Longo não Codificante/biossíntese , RNA Neoplásico/biossíntese , RNA Interferente Pequeno/genética , Análise Serial de Tecidos , Transfecção , Cicatrização
14.
Anticancer Drugs ; 26(5): 531-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25646744

RESUMO

The study aimed to clarify the relationship between ß-elemene, a long noncoding RNA (lncRNA), and human telomerase reverse transcriptase (hTERT) in esophageal carcinoma ECA-109 cells. The proliferation of ECA-109 cells was measured using a CCK-8 kit and flow cytometry. PCR microarray and real-time RT-PCR were designed to determine lncRNA expression in ECA-109 cells before and after treatment with ß-elemene. Western blot was used to detect the hTERT level after the differentially expressed lncRNAs in ECA-109 cells were interfered with small interfering RNA (siRNA). On treatment with ß-elemene, the proliferation of ECA-109 cells was notably inhibited, and about 85% of the lncRNAs showed higher expression levels in ECA-109 cells than in those untreated cells, from which, CDKN2B-AS1 was screened out. A specific siRNA (si-CDKN2B-AS1) that targets the ß-elemene-mediated lncRNA CDKN2B-AS1 was designed, synthesized, and applied to treat ECA-109 cells. Its interference efficiency reached as high as 89.6%. When ECA-109 cells were transfected with the siRNA, the hTERT level was increased by 84.7%. The CCK-8 assay showed that the proliferation of ECA-109 cells treated with ß-elemene was significantly promoted after siRNA transfection (P<0.01). It was also shown by flow cytometry that, compared with the scramble-treated group (negative control), the proliferation index value of ECA-109 cells in the si-CDKN2B-AS1 treatment group was notably increased (25.7 vs. 51.7%) and the TERT protein level was increased by 67.25% after the cells were treated with si-CDKN2B-AS1. The chemotherapeutic drug ß-elemene suppressed the proliferation of esophageal carcinoma ECA-109 cells by regulating the inhibition of hTERT expression by lncRNA CDKN2B-AS1.


Assuntos
Antineoplásicos/farmacologia , RNA Longo não Codificante/metabolismo , Sesquiterpenos/farmacologia , Telomerase/metabolismo , Carcinoma de Células Escamosas , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Esofágicas , Regulação da Expressão Gênica , Humanos , RNA Longo não Codificante/genética , Telomerase/genética
15.
Jpn J Clin Oncol ; 44(3): 241-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24453273

RESUMO

OBJECTIVE: The incidence of radiation-induced late xerostomia varies greatly in nasopharyngeal carcinoma patients treated with radiotherapy. The single-nucleotide polymorphisms in genes involved in DNA repair and fibroblast proliferation may be correlated with such variability. The purpose of this paper was to evaluate the association between the risk of developing radiation-induced late xerostomia and four genetic polymorphisms: TGFß1 C-509T, TGFß1 T869C, XRCC3 722C>T and ATM 5557G>A in nasopharyngeal carcinoma patients treated with Intensity Modulation Radiated Therapy. METHODS: The severity of late xerostomia was assessed using a patient self-reported validated xerostomia questionnaire. Polymerase chain reaction-ligation detection reaction methods were performed to determine individual genetic polymorphism. The development of radiation-induced xerostomia associated with genetic polymorphisms was modeled using Cox proportional hazards, accounting for equivalent uniform dose. RESULTS: A total of 43 (41.7%) patients experienced radiation-induced late xerostomia. Univariate Cox proportional hazard analyses showed a higher risk of late xerostomia for patients with XRCC3 722 TT/CT alleles. In multivariate analysis adjusted for clinical and dosimetric factors, XRCC3 722C>T polymorphisms remained a significant factor for higher risk of late xerostomia. CONCLUSIONS: To our knowledge, this is the first study that demonstrated an association between genetic polymorphisms and the risk of radiation-induced late xerostomia in nasopharyngeal carcinoma patients treated with Intensity Modulation Radiated Therapy. Our findings suggest that the polymorphisms in XRCC3 are significantly associated with the risk of developing radiation-induced late xerostomia.


Assuntos
Proteínas de Ligação a DNA/genética , Neoplasias Nasofaríngeas/radioterapia , Polimorfismo de Nucleotídeo Único , Radioterapia de Intensidade Modulada/efeitos adversos , Fator de Crescimento Transformador beta1/genética , Xerostomia/etiologia , Adulto , Idoso , Carcinoma , Fatores de Confusão Epidemiológicos , Feminino , Frequência do Gene , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo , Reação em Cadeia da Polimerase , Fatores de Risco , Autorrelato , Índice de Gravidade de Doença , Inquéritos e Questionários , Xerostomia/genética
16.
Zhonghua Yi Xue Za Zhi ; 94(19): 1507-11, 2014 May 20.
Artigo em Zh | MEDLINE | ID: mdl-25143175

RESUMO

OBJECTIVE: To explore the effects of differencial expression of WISP1 upon the radiosenstivity and migration in glioma cell lines M059K and M059J, as well as WISP1 knockdown cell line M059K-WISP1-. METHODS: The expression of WISP1 in different cell lines was detected by Western blot assay. The lentivirus mediated-RNA interference technology was employed to knockdown the endogenous expression of gene WISP1 in human glioma cells(M059K). A stable cell line was established by infecting M059K cells with the lentivirus particles, the transfection efficiency was examined by fluorescence microscopy. The radiosensitivity and migration ability of cells were assessed by Colony-forming and Scratch Wound Assays respectively. RESULTS: WISP1 was highly expressed in M059K cells compared with M059J cells. M059K cells were less radiosensitive than M059J cells , and had a better ability of migration. M059K cells were transfected into by the lentiviral vector pGLV3/H1/GFP+puro vector -WISP1. And M059K-WISP1- cell line with stable WISP1 downregulation was established successfully. Western blot demonstrated significantly downregulated WISP1 expression in M059K-WISP1- cells, which showed obviously improved radiosensitivity in Colony-forming assay and suppressed migration ability in Scratch Wound Assay. Colony-forming assay based on the muti-target/single-hit model indicated that radiosensitivity of M059K-WISP1- was significantly decreased. CONCLUSION: Compared with WISP1 low expression cell line M059J , WISP1 high expression cell line M059K was less radiosensitive and had a better ability of migration.We have successfully established a glioma cell line M059K-WISP1- stably downregulating WISP1, which shows a better radiosensitivity and a lowered migration activity in vitro. This may provide a novel therapeutic target for the treatment of human glioma.


Assuntos
Proteínas de Sinalização Intercelular CCN/genética , Glioma/patologia , Proteínas Proto-Oncogênicas/genética , Tolerância a Radiação/genética , Linhagem Celular Tumoral , Movimento Celular , Técnicas de Silenciamento de Genes , Vetores Genéticos , Humanos , Lentivirus/genética , Transfecção
17.
Int J Womens Health ; 16: 755-767, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38706691

RESUMO

Objective: Immune cells play a key role in tumor microenvironment. The purpose of this study was to investigate the infiltration and clinical indication of immune cells including their combined prognostic value in microenvironment of triple negative breast cancer. Methods: We investigated 100 patients with triple negative breast cancer by Opal/Tyramide Signal Amplification multispectral immunofluorescence between 2003 and 2017 at Zhejiang Provincial people's Hospital. Intratumoral and stromal immune cells of triple negative breast cancer were classified and quantitatively analyzed. Survival outcomes were compared using the Kaplan-Meier method and further analyzed with multivariate analysis. Results: Infiltration level of stromal B lymphocytes, stromal and intratumoral CD8+ T cells, stromal CD4+ T cells, stromal PD-L1 and intratumoral tumor associated macrophages 2 cells were shown as independent factors affecting disease-free survival and overall survival in univariate analysis. Stromal B lymphocytes, T stage, N stage and pathological type were independent predictive factors for both DFS and OS in multivariate analysis. We firstly found that patients with B lymphocytes-enriched subtypes have a better prognosis than those with T lymphocytes-enriched subtypes and tumor-associated macrophage-enriched subtypes. Conclusion: The present study identified a bunch of immune targets and subtypes, which could be exploited in future combined immunotherapy/chemotherapy strategies for triple negative breast cancer patients.

18.
Technol Cancer Res Treat ; 22: 15330338231169873, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37066520

RESUMO

Esophageal cancer is one of the most lethal malignancies worldwide. For locally advanced diseases, radiotherapy is the main treatment. The survival rates, however, are still appalling, which is partially due to radioresistance. In 6% of cases of esophageal cancer, the Hedgehog pathway is reactivated, and this pathway is crucial for the growth, progression, treatment resistance, and metastasis of esophageal cancer. Here, we conducted a comprehensive review of the current research on Hedgehog pathway in esophageal cancer.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Esofágicas , Humanos , Proteínas Hedgehog , Neoplasias Esofágicas/patologia , Transdução de Sinais , Carcinoma de Células Escamosas/patologia , beta Catenina/metabolismo , Tolerância a Radiação
19.
Nat Commun ; 14(1): 892, 2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36807354

RESUMO

Intratumoral heterogeneity (ITH) has been linked to decreased efficacy of clinical treatments. However, although genomic ITH has been characterized in genetic, transcriptomic and epigenetic alterations are hallmarks of esophageal squamous cell carcinoma (ESCC), the extent to which these are heterogeneous in ESCC has not been explored in a unified framework. Further, the extent to which tumor-infiltrated T lymphocytes are directed against cancer cells, but how the immune infiltration acts as a selective force to shape the clonal evolution of ESCC is unclear. In this study, we perform multi-omic sequencing on 186 samples from 36 primary ESCC patients. Through multi-omics analyses, it is discovered that genomic, epigenomic, and transcriptomic ITH are underpinned by ongoing chromosomal instability. Based on the RNA-seq data, we observe diverse levels of immune infiltrate across different tumor sites from the same tumor. We reveal genetic mechanisms of neoantigen evasion under distinct selection pressure from the diverse immune microenvironment. Overall, our work offers an avenue of dissecting the complex contribution of the multi-omics level to the ITH in ESCC and thereby enhances the development of clinical therapy.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Humanos , Carcinoma de Células Escamosas do Esôfago/genética , Neoplasias Esofágicas/genética , Multiômica , Transcriptoma , Perfilação da Expressão Gênica , Microambiente Tumoral
20.
J Immunol ; 184(9): 5047-54, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20363964

RESUMO

CD1d-expressing cells present lipid Ag to CD1d-restricted NKT cells, which play an important role in immune regulation and tumor rejection. Lymphoid enhancer-binding factor-1 (LEF-1) is one of the regulators of the Wnt signaling pathway, which is a powerful regulator in cellular growth, differentiation, and transformation. There is little evidence connecting Wnt signaling to CD1d expression. In this study, we have identified LEF-1 as a regulator of the expression of the gene encoding the human CD1d molecule (CD1D). We found that LEF-1 binds specifically to the CD1D promoter. Overexpression of LEF-1 in K562 or Jurkat cells suppresses CD1D promoter activity and downregulates endogenous CD1D transcripts, whereas knockdown of LEF-1 using LEF-1-specific small interfering RNA increases CD1D transcripts in K562 and Jurkat cells but there are different levels of surface CD1d on these two cell types. Chromatin immunoprecipitation showed that the endogenous LEF-1 is situated at the CD1D promoter and interacts with histone deacetylase-1 to facilitate the transcriptional repressor activity. Knockdown of LEF-1 using small interfering RNA potentiates an acetylation state of histone H3/H4, supporting the notion that LEF-1 acts as a transcriptional repressor for the CD1D gene. Our finding links LEF-1 to CD1D and suggests a role of Wnt signaling in the regulation of the human CD1D gene.


Assuntos
Antígenos CD1d/genética , Regulação para Baixo/imunologia , Fator 1 de Ligação ao Facilitador Linfoide/fisiologia , Regiões Promotoras Genéticas/imunologia , Proteínas Repressoras/genética , Antígenos CD1d/biossíntese , Antígenos CD1d/metabolismo , Regulação para Baixo/genética , Técnicas de Silenciamento de Genes , Humanos , Células Jurkat , Células K562 , Fator 1 de Ligação ao Facilitador Linfoide/antagonistas & inibidores , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Ligação Proteica/genética , Ligação Proteica/imunologia , RNA Interferente Pequeno/genética , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Proteínas Wnt/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa