Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
2.
JAMA Oncol ; 8(2): 242-250, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-34913959

RESUMO

IMPORTANCE: Patients with radioactive iodine-refractory differentiated thyroid cancer (RAIR-DTC) have a poor prognosis and limited treatment options. OBJECTIVE: To assess the efficacy and safety of apatinib, a highly selective vascular endothelial growth factor (VEGFR-2) inhibitor, in patients with progressive locally advanced or metastatic RAIR-DTC. DESIGN, SETTING, AND PARTICIPANTS: This randomized, double-blind, placebo-controlled, phase 3 trial (Efficacy of Apatinib in Radioactive Iodine-refractory Differentiated Thyroid Cancer [REALITY]) was conducted in 92 patients with progressive locally advanced or metastatic RAIR-DTC between February 17, 2017, and March 2, 2020, at 21 sites within China, and the data cutoff date for this analysis was March 25, 2020. INTERVENTIONS: Patients were randomly assigned (1:1) to apatinib, 500 mg/d, or placebo. Patients who developed progression while receiving placebo were allowed to cross over to apatinib. MAIN OUTCOMES AND MEASURES: The primary end point was investigator-assessed progression-free survival (PFS). Secondary end points included overall survival, objective response rate (ORR), disease control rate (DCR), duration of response, time to objective response, and safety. Intention-to-treat analyses were performed to evaluate efficacy. RESULTS: Of the 92 patients included in the trial, 56 were women (60.9%); mean (SD) age at baseline was 55.7 (10.6) years. Patients were randomized to the apatinib (n = 46) or placebo (n = 46) group. The median follow-up duration was 18.1 (IQR, 12.7-22.2) months. The median PFS was 22.2 (95% CI, 10.91-not reached) months for apatinib vs 4.5 (95% CI, 1.94-9.17) months for placebo (hazard ratio, 0.26; 95% CI, 0.14-0.47; P < .001). The confirmed ORR was 54.3% (95% CI, 39.0%-69.1%) and the DCR was 95.7% (95% CI, 85.2%-99.5%) in the apatinib group vs an ORR of 2.2% (95% CI, 0.1%-11.5%) and DCR of 58.7% (95% CI, 43.2%-73.0%) in the placebo group. The median overall survival was not reached for apatinib (95% CI, 26.25-not reached) and was 29.9 months (95% CI, 18.96-not reached) for placebo (hazard ratio, 0.42; 95% CI, 0.18-0.97; P = .04). The most common grade 3 or higher-level treatment-related adverse events in the apatinib group were hypertension (16 [34.8%]), hand-foot syndrome (8 [17.4%]), proteinuria (7 [15.2%]), and diarrhea (7 [15.2%])-none of which occurred in the placebo group. CONCLUSIONS AND RELEVANCE: The REALITY trial met its primary end point of PFS at the prespecified interim analysis. Apatinib showed significant clinical benefits in both prolonged PFS and overall survival with a manageable safety profile in patients with progressive locally advanced or metastatic RAIR-DTC. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT03048877.


Assuntos
Antineoplásicos , Piridinas , Neoplasias da Glândula Tireoide , Idoso , Antineoplásicos/uso terapêutico , Feminino , Humanos , Radioisótopos do Iodo , Masculino , Pessoa de Meia-Idade , Piridinas/uso terapêutico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/radioterapia
3.
Lung Cancer ; 130: 135-142, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30885334

RESUMO

BACKGROUND: High concordance has been observed between Ventana D5F3 ALK immunohistochemistry (IHC) and fluorescence in-situ hybridization (FISH) in lung adenocarcinoma (LADC). However, whether a similar conclusion can be applied to lung squamous-cell carcinoma (LSCC) has remained unclear. We therefore evaluated the ALK (anaplastic lymphoma kinase) status and the therapeutic effect of an ALK tyrosine kinase inhibitor (TKI) in IHC- or FISH-positive LSCC. MATERIALS AND METHODS: A total of 2403 LSCC patients from three institutions were screened for ALK aberration by IHC. All IHC-positive cases were subjected to FISH (with an approximately equal number of negative cases as a control group) and next-generation sequencing (NGS). Clinical efficacy was evaluated for the patients who received TKI therapy. RESULTS: In 2403 cases of LSCC, 37 cases were identified as ALK-positive by IHC. After quality control, 28 cases were succeeded by FISH (six with insufficient tissue, three with lack of signals) and 13 by NGS (24 failed due to insufficient samples or poor DNA quality); the percentage of non-diagnostic tests was 24.3% (9/37) and 64.9% (24/37), respectively. Four cases (4/2394, 0.17%) analyzed by FISH were determined as ALK-positive. For the control group (40 ALK IHC), FISH demonstrated no samples with ALK gene fusion. The concordance between ALK IHC- and ALK FISH-positive results was 14.3% (4/28). In the 13 cases studied by NGS, two cases showed ALK-EML4 fusion (consistent with two FISH-positive results), and two cases were interpreted as harboring an ALK-association gene mutation. Among four patients (two FISH-positive and two IHC-positive only cases) receiving TKI therapy, two patients had stable disease and the other two had progressive disease. CONCLUSIONS: The positive concordance rate of ALK IHC and FISH in LSCC is far less than that reported for LADC. Therefore, ALK IHC detection in LSCC cannot be used as a diagnostic method for ALK rearrangement.


Assuntos
Quinase do Linfoma Anaplásico/genética , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/diagnóstico , Crizotinibe/uso terapêutico , Neoplasias Pulmonares/diagnóstico , Inibidores de Proteínas Quinases/uso terapêutico , Adulto , Idoso , Quinase do Linfoma Anaplásico/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Mutação/genética , Reprodutibilidade dos Testes
4.
Cancer Manag Res ; 10: 115-121, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29403309

RESUMO

BACKGROUND: The AURA3 clinical trial has shown that advanced non-small cell lung cancer (NSCLC) patients with EGFR T790M mutations in circulating tumor DNA (ctDNA) could benefit from osimertinib. PURPOSE: The aim of this study was to assess the usefulness of QuantStudio™ 3D Digital PCR System platform for the detection of plasma EGFR T790M mutations in NSCLC patients, and compare the performances of 3D Digital PCR and ARMS-PCR. PATIENTS AND METHODS: A total of 119 Chinese patients were enrolled in this study. Mutant allele frequency of plasma EGFR T790M was detected by 3D Digital PCR, then 25 selected samples were verified by ARMS-PCR and four of them were verified by next generation sequencing (NGS). RESULTS: In total, 52.94% (69/119) had EGFR T790M mutations detected by 3D Digital PCR. In 69 positive samples, the median mutant allele frequency (AF) was 1.09% and three cases presented low concentration (AF <0.1%). Limited by the amount of plasma DNA, 17 samples (AF <2.5%) and eight samples (T790M-) were selected for verification by ARMS-PCR. Four of those samples were verified by NGS as a third verification method. Among the selected 17 positive cases, ten samples presented mutant allele frequency <0.5%, and seven samples presented intermediate mutant allele frequency (0.5% AF 2.5%). However, only three samples (3/17) were identified as positive by ARMS-PCR, namely, P6 (AF =1.09%), P7 (AF =2.09%), and P8 (AF =2.21%). It is worth mentioning that sample P9 (AF =2.05%, analyzed by 3D Digital PCR) was identified as T790M- by ARMS-PCR. Four samples were identified as T790M+ by both NGS and 3D Digital PCR, and typically three samples (3/4) presented at a low ratio (AF <0.5%). CONCLUSION: Our study demonstrated that 3D Digital PCR is a novel method with high sensitivity and specificity to detect EGFR T790M mutation in plasma.

5.
Biochem Biophys Res Commun ; 482(4): 1400-1406, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27956175

RESUMO

Interleukin 23 (IL-23) is an inflammatory cytokine which plays a vital role in autoimmune diseases as well as in tumorigenesis. However, the role of IL-23 in tumor procession is still controversial and the underlying mechanism remains unclear. Here we established a stable cell line overexpressing IL-23 to prove that IL-23 promoted tumor growth and pulmonary metastasis through induction of tumor-related inflammation and absence of immune surveillance. IL-23 promotes tumor-associate inflammatory response such as infiltration of M2 macrophages, neutrophils and their elevated secretions of immunosuppressive cytokines transforming growth factor-ß (TGF-ß), IL-10 and vascular endothelial growth factor (VEGF) into tumor tissues, meanwhile the increase of the matrix metalloprotease MMP9. In addition, IL-23 increases the expression of the endothelial marker CD31 and proliferative marker Ki67 in tumors. Moreover, IL23 induces immunosuppression though reducing the infiltration of CD4+and CD8+T cells into tumor tissues. In conclusion, IL-23 is a considerable molecular in tumor progression, which simultaneously facilitates processes of pro-tumor inflammation, such as angiogenesis, immunosuppressive cytokines as well as infiltrations of M2 macrophages and neutrophils, and suppresses antitumor immune responses through reduction of CD4+ T cells and CD8+ T cells.


Assuntos
Interleucina-23/genética , Interleucina-23/metabolismo , Macrófagos/metabolismo , Neoplasias Mamárias Animais/metabolismo , Neutrófilos/metabolismo , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Proliferação de Células , Progressão da Doença , Feminino , Inflamação , Interleucina-10/metabolismo , Antígeno Ki-67/metabolismo , Macrófagos/citologia , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neutrófilos/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Sci Rep ; 6: 32351, 2016 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-27571893

RESUMO

Genetic modification of whole-cell cancer vaccines to augment their efficacies has a history of over two and a half decades. Various genes and gene combinations, targeting different aspects of immune responses have been tested in pursuit of potent adjuvant effects. Here we show that co-expression of two cytokine members of the common cytokine receptor γ-chain family, IL-21 and IL-7, in whole-cell cancer vaccines boosts antitumor immunity in a CD4(+) and CD8(+) T cell-dependent fashion. It also generates effective immune memory. The vaccine-elicited short-term effects positively correlated with enhanced infiltration of CD4(+) and CD8(+) effector T cells, and the long-term effects positively correlated with enhanced infiltration of effector memory T cells, especially CD8(+) effector memory T cells. Preliminary data suggested that the vaccine exhibited good safety profile in murine models. Taken together, the combination of IL-21 and IL-7 possesses potent adjuvant efficacy in whole-cell vaccines. This finding warrants future development of IL-21 and IL-7 co-expressing whole-cell cancer vaccines and their relevant combinatorial regimens.


Assuntos
Vacinas Anticâncer/imunologia , Imunidade Celular/genética , Interleucina-7/imunologia , Interleucinas/imunologia , Neoplasias/prevenção & controle , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Interleucina-7/genética , Interleucinas/genética , Camundongos , Camundongos Knockout , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/prevenção & controle
7.
Sci Rep ; 6: 23764, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-27026065

RESUMO

Overexpression of folate receptor alpha (FRα) and high telomerase activity are considered to be the characteristics of ovarian cancers. In this study, we developed FRα-targeted lipoplexes loaded with an hTERT promoter-regulated plasmid that encodes a matrix protein (MP) of the vesicular stomatitis virus, F-LP/pMP(2.5), for application in ovarian cancer treatment. We first characterized the pharmaceutical properties of F-LP/pMP(2.5). The efficient expression of the MP-driven hTERT promoter in SKOV-3 cells was determined after an in-vitro transfection assay, which was significantly increased compared with a non-modified LP/pMP(2.5) group. F-LP/pMP(2.5) treatment significantly inhibited the growth of tumors and extended the survival of mice in a SKOV-3 tumor model compared with other groups. Such an anti-tumor effect was due to the increased expression of MP in tumor tissue, which led to the induction of tumor cell apoptosis, inhibition of tumor cell proliferation and suppression of tumor angiogenesis. Furthermore, a preliminary safety evaluation demonstrated a good safety profile of F-LP/pMP(2.5) as a gene therapy agent. Therefore, FRα-targeted lipoplexes with therapeutic gene expression regulated by an hTERT promoter might be a promising gene therapy agent and a potential translational candidate for the clinical treatment of ovarian cancer.


Assuntos
Neoplasias Ovarianas/terapia , Animais , Linhagem Celular Tumoral , Feminino , Terapia Genética , Humanos , Lipossomos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Neoplasias Ovarianas/patologia , Plasmídeos/genética , Transfecção , Vesiculovirus/genética , Proteínas da Matriz Viral/genética
8.
Oncotarget ; 7(7): 7761-72, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26760960

RESUMO

Toll-like receptors (TLRs)/NF-κB activation stimulated by lipopolysaccharide (LPS) was associated with diverse biological response in colon cancer, but the underlying mechanism was largely unknown. In the current study, we reported cell proliferation was elevated in adenomatous polyposis coli (APC) mutated- and APC knockdown cell lines, while the proliferation was inhibited in APC wild-type cell lines. Besides, in vivo experiments showed that LPS promoted APC knockdown tumor growth while inhibited proliferation of APC wild type. Further study confirmed that activation of TLRs/NF-κB signaling pathway by LPS cross regulated with APC/GSK-3ß/ß-catenin pathway, which were depend on APC status of cell lines. Taken together, APC genotypes play a key role in LPS induced different colon cancer biological response by cross-regulating ß-catenin and NF-κB, which may provide a novel strategy for carcinogenesis prevention.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Neoplasias do Colo/patologia , Mutação/genética , Receptor 4 Toll-Like/metabolismo , Proteína da Polipose Adenomatosa do Colo/antagonistas & inibidores , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Apoptose , Western Blotting , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Feminino , Citometria de Fluxo , Imunofluorescência , Genótipo , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Técnicas Imunoenzimáticas , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Nus , NF-kappa B/metabolismo , RNA Interferente Pequeno/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/metabolismo
9.
Sci Rep ; 5: 16437, 2015 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-26561336

RESUMO

Myeloid-derived suppressor cells (MDSCs) are known to play important roles in the development of immunosuppressive tumor microenvironment. A20 is a zinc-finger protein which could negatively regulate apoptosis in several cell types. However, the role of A20 in tumor microenvironment remains largely unknown. In this study, we found that A20 was over-expressed in MDSCs. The treatment of tumor-bearing mice with small interfering RNA targeting A20 (si-A20) inhibited the growth of tumors. The infiltration of MDSCs was dramatically reduced after si-A20 treatment, as compared to control groups, whereas the numbers of dendritic cells and macrophages were not affected. Also, injection of si-A20 improved T cell mediated tumor-specific immune response. Depletion of MDSCs with anti-Gr1 antibody showed similar antitumor effect and improved T cell response. TNF-α was highly expressed after si-A20 injection. Furthermore, si-A20 induced apoptosis of MDSCs in the presence of TNF-α both in vivo and in vitro. Cleaved Caspase-3 and Caspase-8 were elevated with the activation of JNK pathway after the induction of MDSC apoptosis by si-A20. Thus, our findings suggested that knockdown of A20 in tumor site inhibited tumor growth at least through inducing the apoptosis of MDSCs. A20 might be a potential target in anticancer therapy.


Assuntos
Apoptose/genética , Apoptose/imunologia , Cisteína Endopeptidases/genética , Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Células Mieloides/imunologia , Células Mieloides/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Tolerância Imunológica , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Sistema de Sinalização das MAP Quinases , Melanoma Experimental , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , RNA Interferente Pequeno/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa
10.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 46(4): 542-7, 2015 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-26480655

RESUMO

OBJECTIVE: To determine the role of infiltrated T cells in the accumulation of myeloid-derived suppressor cells (MDSCs) in tumor microenvironment. METHODS: T-cell-deficient nude mice models were established using BALB/c mice. Growth of tumors was compared between those with and without adoptive transfer of T cells. Pathological changes of the tumors were examined with HE histological analysis. The levels of MDSCs were detected with flow cytometry (FACS). RESULTS: Tumor growth was promoted in T-cell-deficient nude mice, which was accompanied with lower levels of MDSCs compared with BALB/c mice (P < 0.05). T cell transfer increased the level of MDSCs significantly (P < 0.05). T cells depletion decreased the level of MDSCs (P < 0.05). CONCLUSION: Infiltrated T cells induce the accumulation of MDSCs in tumor microenvironment, and influence tumor growth.


Assuntos
Células Mieloides/citologia , Neoplasias/patologia , Linfócitos T/citologia , Microambiente Tumoral , Animais , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus
11.
Int J Oncol ; 47(5): 1901-11, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26398907

RESUMO

Human umbilical endothelial cells (HUVECs) have been proven to be effective in tumor anti-angiogenesis but the mechanism remained to be further demonstrated. The restricted ability of HUVECs to proliferate in vitro also limits their application on a large scale. In the present study, we immortalized HUVECs with hTERT genes by lentiviral infection and explored the antitumor immunity of hTERT-expressing HUVECs (HUVEC-TERTs). Results showed that HUVEC-TERTs maintained high telomere activity and expressed CD31, VEGFR-II and integrin α5. Passage-30 HUVEC-TERTs were able to form vascular tubes in vitro without showing signs of senescence. In vivo HUVEC-TERTs elicited antitumor immunity in mouse LL2 and CT26 models protectively and therapeutically. Both humoral and cellular immunity participated in the tumor anti-angiogenesis as HUVEC-neutralizing sera antibodies and HUVEC-specific CTL were detected. The subsets of activated spleen T lymphocytes included both CD4(+) T cells and CD8(+) T cells. Moreover, MDSCs and Tregs were decreased while T lymphocytes were aggregated in the tumor microenvironment. Collectively, the present study is the first to confirm the antitumor immunity of hTERT-immortalized HUVECs. Both anti-angiogenesis and tumor microenvironmental regulation participated in the antitumor activity. Transducing hTERT genes might be a new strategy to allow HUVECs to be applied on a large scale in cancer immunotherapy.


Assuntos
Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Neovascularização Patológica/imunologia , Telomerase/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Neoplasias/patologia , Neovascularização Patológica/terapia , Linfócitos T Citotóxicos/imunologia , Telomerase/genética , Telômero/genética
12.
J Hematol Oncol ; 8: 67, 2015 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-26063074

RESUMO

BACKGROUND: SUMO-activating enzyme subunit 2 (SAE2) is the sole E1-activating enzyme required for numerous important protein SUMOylation, abnormal of which is associated with carcinogenesis. SAE2 inactivation was recently reported to be a therapeutic strategy in cancers with Myc overexpression. However, the roles of SAE2 in small cell lung cancer (SCLC) are largely unknown. METHODS: Stably SAE2 knockdown in H446 cells were established with a lentiviral system. Cell viability, cell cycle, and apoptosis were analyzed using MTT assay and flow cytometric assay. Expression of SAE2 mRNA and protein were detected by qPCR, western blotting, and immunohistochemical staining. Cell invasion and migration assay were determined by transwell chamber assay. H446 cells with or without SAE2 knockdown, nude mice models were established to observe tumorigenesis. RESULTS: SAE2 was highly expressed in SCLC and significantly correlated with tumorigenesis in vivo. Cancer cells with RNAi-mediated reduction of SAE2 expression exhibited growth retardation and apoptosis increasing. Furthermore, down-regulation of SAE2 expression inhibited migration and invasion, simultaneously increased the sensitivity of H446 to etoposide and cisplatin. CONCLUSIONS: SAE2 plays an important role in tumor growth, metastasis, and chemotherapy sensitivity of H446 and is a potential clinical biomarker and therapeutic target in SCLC with high c-Myc expression.


Assuntos
Transformação Celular Neoplásica/genética , Neoplasias Pulmonares/genética , Carcinoma de Pequenas Células do Pulmão/genética , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Imuno-Histoquímica , Camundongos
13.
Biochem Biophys Res Commun ; 459(3): 398-404, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25735977

RESUMO

The echinoderm microtubule-associated protein-like 4(EML4)--anaplastic lymphoma kinase (ALK) fusion gene has been identified as a driver mutation in non-small-cell lung cancer (NSCLC). However, the role of EML4-ALK in malignant transformation is not entirely clear. Here, for the first time, we showed that H1299 NSCLC cells stably expressing EML4-ALK acquire EMT phenotype, associated with enhanced invasive migration and increased expression of EMT-inducing transcription factors. H1299-EML4-ALK cells also displayed cancer stem cell-like properties with a concomitant up-regulation of CD133 and enhanced ability of mammospheres formation. Moreover, we found that inhibition of ERK1/2 reversed EMT induced by EML4-ALK in H1299 cells. Taken together, these results suggested that EML4-ALK induced ERK activation is mechanistically associated with EMT phenotype. Thus, inhibition of ERK signaling pathway could be a potential strategy in treatment of NSCLC patients with EML4-ALK translocation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas de Fusão Oncogênica/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Movimento Celular , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal/genética , Humanos , Neoplasias Pulmonares/genética , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Fusão Oncogênica , Proteínas de Fusão Oncogênica/genética , Fenótipo , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia
14.
Cell Res ; 25(2): 237-53, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25613571

RESUMO

Nanocarriers with positive surface charges are known for their toxicity which has limited their clinical applications. The mechanism underlying their toxicity, such as the induction of inflammatory response, remains largely unknown. In the present study we found that injection of cationic nanocarriers, including cationic liposomes, PEI, and chitosan, led to the rapid appearance of necrotic cells. Cell necrosis induced by cationic nanocarriers is dependent on their positive surface charges, but does not require RIP1 and Mlkl. Instead, intracellular Na(+) overload was found to accompany the cell death. Depletion of Na(+) in culture medium or pretreatment of cells with the Na(+)/K(+)-ATPase cation-binding site inhibitor ouabain, protected cells from cell necrosis. Moreover, treatment with cationic nanocarriers inhibited Na(+)/K(+)-ATPase activity both in vitro and in vivo. The computational simulation showed that cationic carriers could interact with cation-binding site of Na(+)/K(+)-ATPase. Mice pretreated with a small dose of ouabain showed improved survival after injection of a lethal dose of cationic nanocarriers. Further analyses suggest that cell necrosis induced by cationic nanocarriers and the resulting leakage of mitochondrial DNA could trigger severe inflammation in vivo, which is mediated by a pathway involving TLR9 and MyD88 signaling. Taken together, our results reveal a novel mechanism whereby cationic nanocarriers induce acute cell necrosis through the interaction with Na(+)/K(+)-ATPase, with the subsequent exposure of mitochondrial damage-associated molecular patterns as a key event that mediates the inflammatory responses. Our study has important implications for evaluating the biocompatibility of nanocarriers and designing better and safer ones for drug delivery.


Assuntos
Apoptose/efeitos dos fármacos , Portadores de Fármacos/toxicidade , Nanoestruturas/toxicidade , Necrose , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Cátions/química , Linhagem Celular Tumoral , Quitosana/química , Quitosana/toxicidade , DNA Mitocondrial/metabolismo , Portadores de Fármacos/química , Humanos , Inflamação/patologia , Lipossomos/química , Lipossomos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Nanoestruturas/química , Ouabaína/farmacologia , Polietilenoimina/química , Polietilenoimina/toxicidade , Transdução de Sinais/efeitos dos fármacos , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/genética , Receptor Toll-Like 9/metabolismo
15.
J Immunol ; 194(1): 429-37, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25429070

RESUMO

Hemorrhage is a common clinical manifestation in patients with cancer. Intratumor hemorrhage has been demonstrated to be a poor prognostic factor for cancer patients. In this study, we investigated the role of RBCs and hemoglobin (Hb) in the process of tumor progression and therapeutical response. RBCs and Hb potently promoted tumor cell proliferation and syngenic tumor growth. RBCs and Hb activated the reactive oxygen species-NF-κB pathway in both tumor cells and macrophages. RBCs and Hb also induced chemoresistance mediated, in part, by upregulating ABCB1 gene expression. Tumor growth induced by RBCs was accompanied by an inflammatory signature, increased tumor vasculature, and influx of M2 macrophages. In both the peritoneal cavity and tumor microenvironment, extravascular RBCs rapidly recruited monocyte-macrophages into the lesion sites. In addition, RBCs and Hb increased several nucleotide-binding oligomerization domain-like receptors' expression and induced IL-1ß release. Our results provide novel insights into the protumor function of RBCs and Hb as endogenous danger signals, which can promote tumor cell proliferation, macrophage recruitment, and polarization. Hemorrhage may represent a useful prognostic factor for cancer patients because of its role in tumor promotion and chemoresistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Eritrócitos/patologia , Hemoglobinas/farmacologia , Hemorragia/patologia , Neoplasias/patologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/biossíntese , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Movimento Celular/imunologia , Proliferação de Células , Cisplatino/farmacologia , Doxorrubicina/farmacologia , Inflamação/imunologia , Interleucina-1beta/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neovascularização Patológica , Estresse Oxidativo/efeitos dos fármacos , Paclitaxel/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição RelA/biossíntese , Fator de Transcrição RelA/metabolismo , Microambiente Tumoral/imunologia
16.
J Cancer Res Clin Oncol ; 140(11): 1815-23, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24927808

RESUMO

PURPOSE: Lipopolysaccharide (LPS) is a major component of the outer surface membrane of Gram-negative bacteria which has been proved an effective immune enhancer. Here, we investigated the anti-tumor effect of irradiated tumor cells that stimulated by LPS in mouse xenografts models. METHODS: Tumor cells were irradiated after stimulation with 1 µg/mL LPS for 48 h. The C57BL/6 mice were immunized subcutaneously with irradiated tumor cells. The anti-tumor effect of lymphocytes of immunized mice was investigated. The cytotoxicity of spleen lymphocytes from immunized mice was determined by a standard (51)Cr-release assay. The roles of immune cell subsets in anti-tumor activity were assessed by injected intraperitoneally with monoclonal antibodies. RESULTS: We observed that the vaccine of irradiated tumor cell with LPS-stimulated elicited a stronger protective anti-tumor immunity than other controls. Adoptive transfer of lymphocytes of immunized mice showed that the cellular immune response was involved in the anti-tumor effect. And this effect was achieved by activation of antigen-specific CD8(+) T cell response and reduction of myeloid-derived suppressor cells (MDSCs, Gr1(+) CD11b (+) ), which were confirmed by depletion of immune cell subsets and flow cytometry analysis. CONCLUSIONS: In summary, our study showed that stimulation of LPS was able to enhance anti-tumor immunity of vaccination with tumor cells after irradiation treatment, which might be a new strategy for cancer therapy.


Assuntos
Vacinas Anticâncer/imunologia , Lipopolissacarídeos/farmacologia , Melanoma Experimental/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos da radiação , Linhagem Celular Tumoral , Feminino , Hibridomas , Imunoterapia Adotiva , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Baço/imunologia , Carga Tumoral/imunologia , Vacinação , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Breast Cancer ; 17(1): 25-32, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24744794

RESUMO

PURPOSE: The universal organic solvent dimethyl sulfoxide (DMSO) can be used as a differentiation inducer of many cancer cells and has been widely used as a solvent in laboratories. However, its effects on breast cancer cells are not well understood. The aim of this study is to investigate the effect and associated mechanisms of DMSO on mouse breast cancer. METHODS: We applied DMSO to observe the effect on tumors in a mouse breast cancer model. Tumor-associated macrophages (TAMs) were tested by flow cytometry. Ex vivo tumor microenvironment was imitated by 4T1 cultured cell conditioned medium. Enzyme-linked immunosorbent assays were performed to detect interleukin (IL)-10 and IL-12 expression in medium. To investigate the cytotoxicity of DMSO on TAMs, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays were performed. RESULTS: We found that DMSO produced tumor retardation when injected into mouse peritoneal cavities in a certain concentration range (0.5-1.0 mg/g). Furthermore, as detected by flow cytometry, TAM subtypes were found to be transformed. We further imitated a tumor microenvironment in vitro by using 4T1 cultured cell conditioned medium. Similarly, by using low concentration DMSO (1.0%-2.0% v/v), TAMs were induced to polarize to the classically activated macrophage (M1-type) and inhibited from polarizing into the alternatively activated macrophage (M2-type) in the conditioned medium. IL-10 expression in tumors was reduced, while IL-12 was increased compared with the control. Furthermore, we reported that 2.0% (v/v) DMSO could lead to cytotoxicity in peritoneal macrophages after 48 hours in MTT assays. CONCLUSION: Our findings suggest that DMSO could exert antitumor effects in 4T1 cancer-bearing mice by reversing TAM orientation and polarization from M2- to M1-type TAMs. These data may provide novel insight into studying breast cancer immunotherapy.

18.
FEBS Lett ; 588(10): 1921-9, 2014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24713430

RESUMO

Here, we investigated the molecular mechanism underlying the changes in the distribution of nucleolin. Our study identified PI3K/Akt signaling as an essential pathway regulating the distribution of nucleolin. Furthermore, nucleolin can interact with phospho-PI3K-p55, and changes in the distribution of nucleolin were related to its phosphorylation. Subsequently, we analyzed the correlation of VEGF and nucleolin, and found that distribution of nucleolin related to metastatic potential. Finally, blocking cell surface nucleolin influences the process of epithelial-mesenchymal transitions. This indicates that nucleolin may be a novel cancer therapy target and a predictive marker for tumor migration in colorectal carcinoma.


Assuntos
Neoplasias Colorretais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Western Blotting , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Movimento Celular , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Células HCT116 , Humanos , Masculino , Microscopia de Fluorescência , Pessoa de Meia-Idade , Metástase Neoplásica , Fosforilação , Ligação Proteica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Nucleolina
19.
Bioorg Med Chem Lett ; 24(5): 1426-31, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24491459

RESUMO

A series of novel benzamide derivatives were prepared and evaluated using cell-based measurements. Among these compounds, 10f significantly inhibited Hedgehog signaling and showed equivalent or more potency than GDC-0449 in different tests. Furthermore, compound 10f potently inhibited the proliferation of Daoy, a medulloblastoma cell line that is reported to be resistant to GDC-0449, which indicated a promising prospect in the treatment of Hedgehog signaling pathway related cancer in clinical trial.


Assuntos
Benzamidas/síntese química , Benzamidas/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Sulfonas/síntese química , Sulfonas/farmacologia , Animais , Benzamidas/química , Linhagem Celular Tumoral , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Relação Estrutura-Atividade , Sulfonas/química , Peixe-Zebra
20.
Support Care Cancer ; 22(7): 1741-55, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24510195

RESUMO

PURPOSE: The Distress Thermometer (the DT) is a commonly used screening tool to detect distress in cancer patients. This meta-analysis aims to examine the diagnostic role and the optimal cut-off score of the DT compared with various different reference standards. METHODS: We searched PubMed and Embase from 1997 to September 2013 for relevant studies. After extracting data, we estimated the pooled sensitivity, specificity, likelihood ratios, diagnostic odds ratios, and constructed summary receiver operating characteristics curves to determine the optimal cut-off score. RESULTS: Forty-two relevant studies and 14,808 patients were included in total. When we pooled all the results together, the DT showed a good balance between pooled sensitivity (0.81, 95% confidence intervals (CI) 0.79-0.82) and pooled specificity (0.72, 95% CI 0.71-0.72) at the cut-off score of 4. The value of area under the curve (AUC) is 0.8321. When the DT is compared with the HADS-Total, the cut-off score of 4 maximized the balance between the pooled sensitivity (0.82, 95% CI 0.80-0.84) and pooled specificity (0.73, 95% CI 0.72-0.74). The AUC is 0.8432. CONCLUSIONS: This meta-analysis suggests that the DT is a valid tool to detect potential distress in cancer patients. According to our results, 4 as the optimal cut-off, is recommended. Further studies are needed to be done to examine the accuracy and optimal cut-off score in different regions globally and different cancer subtypes to guide the use of the DT for different patients.


Assuntos
Neoplasias/psicologia , Psicometria/métodos , Estresse Psicológico/diagnóstico , Feminino , Humanos , Funções Verossimilhança , Masculino , Programas de Rastreamento/métodos , Pessoa de Meia-Idade , Neoplasias/diagnóstico , Razão de Chances , Curva ROC , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA