Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cancer Sci ; 112(10): 4139-4150, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34058054

RESUMO

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) (eg, gefitinib) exert potent therapeutic efficacy in non-small-cell lung cancer (NSCLC) harboring EGFR-activating mutations. However, the resistance to EGFR TKIs limits their clinical therapeutic efficacy. TIP30, a newly identified tumor suppressor, appears to be involved in the regulation of cytoplasmic and nuclear EGFR signaling in NSCLC. Our previous study demonstrated that TIP30 regulated EGF-dependent cyclin D1 transcription in human lung adenocarcinoma and suppressed tumorigenesis. In the present study, the involvement of TIP30 in combating gefitinib resistance in NSCLC was determined for the first time in vitro and in vivo. Gain and loss of function studies showed that overexpression of TIP30 effectively sensitized cells to gefitinib in vitro, whereas TIP30 inhibition promoted gefitinib cell resistance. Moreover, TIP30 negatively regulated the activation of the p-AKT and p-MEK signaling pathways in PC9/GR. Importantly, PC9/GR harbored high levels of nuclear EGFR, and overexpression of TIP30 restored irregular EGFR trafficking and degradation from early endosomes to the late endosomes, decreasing the nuclear accumulation of EGFR, which may partly or totally inhibit EGFR-mediated induction of c-Myc transcription. Xenographic tumors induced by overexpression of TIP30 by PC9/GR cells in nude mice were suppressed compared with their original counterparts. Overall, it was revealed that TIP30 overexpression restored gefitinib sensitivity in NSCLC cells and attenuated the cytoplasmic and nuclear EGFR signaling pathways and may be a promising biomarker in gefitinib resistance in NSCLC.


Assuntos
Acetiltransferases/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias Pulmonares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Núcleo Celular/metabolismo , Ciclina D1/metabolismo , Citoplasma/metabolismo , Endossomos/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Gefitinibe/farmacologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Lisossomos/metabolismo , MAP Quinase Quinase 1/metabolismo , Camundongos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Int J Colorectal Dis ; 31(4): 805-11, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26847617

RESUMO

PURPOSE: We wished to evaluate the effectiveness of laparoscopic and open surgery for patients with rectum cancer through a meta-analysis. METHODS: We searched PubMed, EMBASE, and Cochrane database until June 30, 2015, to identify eligible studies. Randomized controlled trials comparing laparoscopic with open surgery for rectum cancer were included. Meta-analysis was performed using the search strategy following the requirement of the Cochrane Library Handbook. Three-year overall survival (OS) and disease-free survival (DFS) were the main endpoints. RESULTS: Eight randomized controlled trials comprising 3145 patients matched the selection criteria. Meta-analysis showed no significant difference between laparoscopic and open surgery in 3-year overall survival (OS) and disease-free survival (DFS) (hazard ratio (HR)3-year OS = 0.83, 95 % CI [0.68-1.01]; P = 0.06; HR3-year DFS = 0.89, 95 % CI [0.75,1.05]; P = 0.16). No evidence of publication bias was observed. CONCLUSION: Our meta-analysis supported the notion that based on the 3-year DFS and OS, oncological outcomes are comparable after laparoscopic and open surgery for rectal cancer.


Assuntos
Laparoscopia , Neoplasias Retais/cirurgia , Idoso , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Viés de Publicação , Ensaios Clínicos Controlados Aleatórios como Assunto , Neoplasias Retais/mortalidade , Taxa de Sobrevida , Resultado do Tratamento
3.
Biochem Biophys Res Commun ; 443(2): 712-7, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24333874

RESUMO

Salinomycin (Sal) is a polyether ionophore antibiotic that has recently been shown to induce cell death in various human cancer cells. However, whether salinomycin plays a functional role in nasopharyngeal carcinoma (NPC) has not been determined to date. The present study investigated the chemotherapeutic efficacy of salinomycin and its molecular mechanisms of action in NPC cells. Salinomycin efficiently inhibited proliferation and invasion of 3 NPC cell lines (CNE-1, CNE-2, and CNE-2/DDP) and activated a extensive apoptotic process that is accompanied by activation of caspase-3 and caspase-9, and decreased mitochondrial membrane potential. Meanwhile, the protein expression level of the Wnt coreceptor lipoprotein receptor related protein 6 (LRP6) and ß-catenin was down-regulated, which showed that the Wnt/ß-catenin signaling was involved in salinomycin-induced apoptosis of NPC cells. In a nude mouse NPC xenograft model, the anti-tumor effect of salinomycin was associated with the downregulation of ß-catenin expression. The present study demonstrated that salinomycin can effectively inhibit proliferation and invasion, and induce apoptosis of NPC cells in vitro and inhibit tumor growth in vivo, probably via the inhibition of Wnt/ß-catenin signaling, suggesting salinomycin as a potential candidate for the chemotherapy of NPC.


Assuntos
Cateninas/metabolismo , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/fisiopatologia , Piranos/administração & dosagem , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Nasofaríngeas/tratamento farmacológico
4.
Cancer Invest ; 30(2): 126-30, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22148972

RESUMO

OBJECTIVE: The current prospective randomized study was designed to evaluate the safety and efficacy of combined intrapleural cisplatin and OK-432 (picibanil) plus hyperthermotherapy in patients with malignant pleural effusion (MPE). METHODS: A total of 358 patients with MPE due to end-stage malignancies were enrolled and randomly divided into two groups, A and B: the intrapleural combination of cisplatin and OK-432 with hyperthermotherapy (n = 179) or without hyperthermotherapy (n = 179), respectively. Mild toxicities such as nausea, vomiting or anorexia, bone marrow depression, and pyrexia were similar in both groups. RESULT: Patients in Group A (with hyperthermotherapy) showed a significantly higher overall response (93.4%) compared to those in Group B (79.8%, χ(2) = 43.11, p < .05). The median survival time for patients in Group A and Group B were 8.9 and 6.2 months, respectively (p > .05). After treatment, the quality of life scores were significantly increased in both groups as compared to prior treatment (p < .05). CONCLUSION: In conclusion, our study suggests that combined intrapleural cisplatin and OK-432 followed by hyperthermotherapy are more effective in the control of MPE and improve patients' quality of life.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Hipertermia Induzida/métodos , Neoplasias Pulmonares/terapia , Derrame Pleural Maligno/terapia , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Terapia Combinada , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Picibanil/administração & dosagem , Picibanil/efeitos adversos , Derrame Pleural Maligno/tratamento farmacológico , Derrame Pleural Maligno/patologia , Estudos Prospectivos , Qualidade de Vida , Análise de Sobrevida
5.
Ann Transl Med ; 10(10): 579, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35722357

RESUMO

Background: Existing research shows that long non-coding RNAs (lncRNAs) have important regulatory effects in gastric cancer (GC). In recent years, focally amplified lncRNA on chromosome 1 (FALEC) has been repeatedly reported to have carcinogenic effects in thyroid carcinoma, colorectal cancer, and endometrial cancer, etc. While the role and mechanism of FALEC during GC tumorigenesis remains unclear. Methods: The levels of FALEC, microRNA-203b (miR-203b), and Recombinant Pim-3 Oncogene (PIM3) were confirmed by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Cell autophagy, proliferation, apoptosis, migration, and invasion were estimated using western blot, transmission electron microscopy (TEM), cell counting kit-8 (CCK-8), flow cytometer, and Transwell assays. The interaction between miR-203b and FALEC or PIM3 was verified using a dual-luciferase reporter assay. Moreover, the involvement of miR-203b and PIM3 in the regulatory effects of FALEC on GC was determined with rescue experiments. Results: The results showed that FALEC and PIM3 were highly expressed, while miR-203b was lowly expressed, in GC. FALEC knockdown repressed GC cell proliferation, migration, and invasion, and promoted apoptosis and autophagy in vitro. Meanwhile, FALEC knockdown prevented growth and induced GC autophagy in vivo. This shows that FALEC upregulated PIM3 by sponging miR-203b in GC cells. Besides, FALEC induced the malignant behaviors of GC cells by regulating the miR-203b/PIM3 axis. Conclusions: The FALEC/miR-203b/PIM3 axis might be a promising therapeutic target for therapy in GC patients.

6.
Pathol Res Pract ; 215(6): 152379, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30878308

RESUMO

BACKGROUND: Epidemiological studies have illustrated that regular aspirin consumption may decrease the risk of non-small cell lung cancer (NSCLC). The present study aims to investigate the mechanism of aspirin-induced inhibition of NSCLC development during hypoxia. METHODS: A549 cells were pre-treated with the vehicle control or aspirin and then subjected to hypoxic culture. Cell viability was monitored by CCK-8 assay, and flow cytometry was performed to detect cell cycle distributions, apoptosis, and proportion of cancer stem cells (CSCs). Flow cytometric cell sorting was used to separate CSCs. Quantitative reverse transcription-polymerase chain reaction and Western blot were used to detect the mRNA and protein levels of stem cell markers and the related signaling molecules. The abundance of prostaglandin E2 was detected by enzyme-linked immunosorbent assay. Exosomes in the cell culture medium were isolated using ExoQuick, and the number of exosomes was quantified by the EXOCET exosome quantification assay kit. Cell migration and angiogenesis were monitored by transwell migration assay and in vitro angiogenesis experiments. RESULTS: Aspirin inhibited cell proliferation and induced G2/M cell cycle arrest in hypoxic A549 cells; it also inhibited hypoxia-enhanced stemness in both A549 and ALDH+ cells. The drug reduced hypoxia-enhanced numbers of exosomes in A549 cells and exerted negative effects on the hypoxia-mediated up-regulation of exosomal HIF-1α/COX-2 and expression of exosomal miR-135b and miR-210. While hypoxic-induced exosomes can promote the proliferation, migration, and angiogenesis of other A549 cells, aspirin can weaken this promotion by reducing the amount of exosome secreted and changing exosome contents. CONCLUSIONS: Aspirin inhibits the hypoxia-induced stemness, hypoxic-mediated exosome release, and malignant paracrine effects of A549 cells.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Exossomos/efeitos dos fármacos , Neoplasias Pulmonares/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células A549 , Hipóxia Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos
7.
Oncol Lett ; 14(1): 751-757, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28693230

RESUMO

Nasopharyngeal carcinoma (NPC) has a high incidence and mortality rate, particularly in Southern China. Apogossypolone (ApoG2) is a novel derivative of gossypol with antitumor activity and less toxicity. The human NPC CNE-2 cell line was studied in the in vitro model; whilst 4 week-old male nude mice (BALB/c-nu) were inoculated subcutaneously with CNE-2 cells, and xenograft tumors were studied in the in vivo model. Graded concentrations of ApoG2 were used in treatment studies. In ApoG2-treated and control in vitro and in vivo tumor cells, cell apoptosis, and autophagy were evaluated and quantified using fluorescent and transmission electron microscopy and flow cytometry. Hoechst-33258 fluorescence staining was used to evaluate apoptosis in treated and non-treated cell culture and xenograft NPC cells. Western blotting was performed on lysed tumor cells using primary antibodies to B-cell lymphoma-2 (Bcl-2), beclin-1, and ß-actin, and flow cytometry results indicated cell apoptosis rates of 3.90±0.34 and 19.52±1.18% in the control and ApoG2-treated cells, respectively (F=485.294, P<0.001). Western blot analysis showed that ApoG2 significantly decreased expression of the Bcl-2 protein in CNE-2 cells, when compared with control cells (F=68.909, P=0.001) and flow cytometry showed cell autophagy rates of 0.92±3.10% of control cells compared with 28.24±7.35% of ApoG2-treated cells (F=31.035, P=0.003). ApoG2 treatment significantly increased beclin-1 protein expression in CNE-2 cells (F=497.906, P<0.001). ApoG2 treatment inhibited NPC xenograft tumor growth by 65.49% (P<0.05). In conclusion, these results support a role for ApoG2 in inhibiting the growth of human NPC cells by inducing apoptosis and autophagy. Future controlled clinical studies could be planned, to define safety, efficacy and dosing regimens for ApoG2 as a potential treatment for patients with NPC.

8.
Mol Med Rep ; 13(2): 1570-6, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26707143

RESUMO

At present, the therapeutic treatment strategies for patients with hepatocellular carcinoma (HCC) remain unsatisfactory, and novel methods are urgently required to treat this disease. Members of the B cell lymphoma (Bcl)-2 family are anti­apoptotic proteins, which are commonly expressed at high levels in certain HCC tissues and positively correlate with the treatment resistance of patients with HCC. ABT-737, an inhibitor of Bcl-2 anti-apoptotic proteins, has been demonstrated to exhibit potent antitumor effects in several types of tumor, including HCC. However, treatment with ABT-737 alone also activates certain pro-survival signaling pathways, which attenuate the antitumor validity of ABT-737. Curcumin, which is obtained from Curcuma longa, is also an antitumor potentiator in multiple types of cancer. In the present study, the synergistic effect of curcumin and ABT-737 on HCC cells was investigated for the first time, to the best of our knowledge. It was found that curcumin markedly enhanced the antitumor effects of ABT-737 on HepG2 cells, which was partially dependent on the induction of apoptosis, according to western blot analysis and flow cytometric apoptosis analysis. In addition, the sustained activation of the ROS-ASK1-c-Jun N-terminal kinase pathway may be an important mediator of the synergistic effect of curcumin and ABT-737. Collectively, these results indicated that the combination of curcumin and ABT-737 can efficaciously induce the death of HCC cells, and may offer a potential treatment strategy for patients with HCC.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Curcumina/administração & dosagem , Neoplasias Hepáticas/tratamento farmacológico , MAP Quinase Quinase 4/biossíntese , MAP Quinase Quinase Quinase 5/biossíntese , Apoptose/efeitos dos fármacos , Compostos de Bifenilo/administração & dosagem , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , MAP Quinase Quinase 4/genética , MAP Quinase Quinase Quinase 5/genética , Nitrofenóis/administração & dosagem , Piperazinas/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Transdução de Sinais , Sulfonamidas/administração & dosagem
9.
Di Yi Jun Yi Da Xue Xue Bao ; 25(12): 1478-81, 2005 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-16354609

RESUMO

OBJECTIVE: To study the molecular mechanism underlying cisplatin resistance in ovarian carcinoma by detecting the expressions of DNA transcription- and repair-related genes in cisplatin-resistant human ovarian carcinoma COC1 cell line. METHODS: The differential expression of DNA transcription- and repair-related genes between the parental COC1 and cisplatin-resistant COC1/DDP cell line was determined using cDNA microarray. RESULTS AND CONCLUSION: Compared with COC1 cells, 143 genes in COC1/DDP cells showed significant differential expression, among which 20 were DNA transcription- and repair-related genes including 13 significantly up-regulated genes and 7 down-regulated ones. Abnormality of DNA transcription and repair might be involved in the development of cisplatin resistance in COC1/DDP cells.


Assuntos
Cisplatino/farmacologia , Reparo do DNA/genética , DNA de Neoplasias/genética , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Ovarianas/genética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Transcrição Gênica
10.
Medicine (Baltimore) ; 94(17): e803, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25929935

RESUMO

The clinical benefits provided by using combined taxanes and anthracyclines in first-line chemotherapy for metastatic breast carcinoma (MBC) remain uncertain. This meta-analysis compares the benefits of using a combination of anthracyclines along with taxanes versus using single-agent-based chemotherapeutic regimens in the treatment of MBC.Relevant clinical trials as well as abstracts from articles presented at major cancer conferences were searched in various databases including PubMed, Embase, and Cochrane Library. The relevant studies had a primary endpoint of overall survival (OS) and secondary endpoints that included progression-free survival (PFS), time-to-treatment failure (TTF), time to progression (TTP), objective response rate (ORR), disease control rate (DCR), and safety. The hazard ratios of OS, PFS, TTF, and TTP, the odds ratios of ORR and DCR, and the risk ratios (RRs) for grades 1-2 and 3-4 toxicities were extracted from the retrieved studies and analyzed using various statistical methods. Meta-analytic estimates were derived from a random-effect model.Fifteen trials were included in the final meta-analysis, and the results suggest that chemotherapy with combined anthracyclines and taxanes does not significantly improve the OS of MBC patients when compared with the OS achieved using separate taxane or anthracycline-based regimens. Compared with taxane-based regimens, combined taxane along with anthracycline regimens failed to significantly improve TTP, ORR, or DCR, but did significantly improve TTP and ORR when compared with anthracycline-based regimens. Furthermore, both individual taxane-based and anthracycline-based regimens produced fewer toxic reactions compared to combined taxane along with anthracycline regimens. Taxane-based regimens had lower RRs for side effects of neutropenia, infection/febrile neutropenia, nausea, and vomiting, whereas patients receiving anthracycline-based regimens had lower RRs for neutropenia, infection/febrile neutropenia, anorexia, stomatitis/mucosal inflammation, diarrhea, and sensory neuropathy. In contrast, patients receiving taxane-based regimens were at higher RRs for hand-foot syndrome and diarrhea, whereas patients receiving anthracycline-based regimens had higher RRs for nausea and vomiting.A taxane-based treatment regimen may be a better option than a combined taxane/anthracycline regimen for managing patients with advanced breast cancer, as it produces equivalent clinical outcomes and has less toxicity compared to other similar regimens.


Assuntos
Antraciclinas/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Taxoides/administração & dosagem , Neoplasias da Mama/patologia , Feminino , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto , Análise de Sobrevida
11.
Di Yi Jun Yi Da Xue Xue Bao ; 24(4): 392-6, 2004 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-15090306

RESUMO

OBJECTIVE: To understand the relation between cytotoxic activity of immunologic effector cells and multidrug resistance of the tumor cells. METHODS: Continuous observation of the morphological changes and MTT colorimetry were employed to evaluate the cytotoxic activity of lymphokine-activated killer (LAK) cells and natural killer (NK) cells against multidrug-resistant (MDR) human oral carcinoma cell line-KBV200 (before and after reversal of MDR) and parental drug-sensitive cell line KB. The morphologic changes of LAK cells and the 3 target cell lines were observed continuously under inverted microscope 3 h after co-culture of LAK cells with one of three target cell lines respectively. The lysis rates of three tumor cell lines in response to co-culture with LAK or NK cells were determined using MTT colorimetry. RESULTS: In comparison with the parental drug-sensitive cell line KB, both KBV200 and its reserved cell line by verapamil (KBVV) showed earlier adherence and greater number of cells lysed by LAK. In MTT colorimetry assay, the cytotoxicity of both LAK and NK cells against the 3 cell lines was associated with the effector-to-target (E/T) cell ratio; the lysis rates of KBV200 and reversed KBV200 cells by verapamil in response to LAK and NK cells were higher than that of KB cells (P<0.05), but KBV200 and KBVV did not significantly differ (P>0.05). At the same E/T ratio, LAK cells possessed stronger cytotoxicity than NK cells against all the tumor cell lines (P<0.05). CONCLUSIONS: Immunologic effector cells possess strong cytotoxic activity against multidrug-resistant cell line KBV200. Modulation of MDR does not decrease the cytotoxic activity of the immunologic effector cells. The results of this study suggest that adoptive cell immunotherapy with immunologic effector cells may be of value in controlling the progress of MDR tumors.


Assuntos
Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Linhagem Celular Tumoral , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Humanos , Células KB/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos
12.
Di Yi Jun Yi Da Xue Xue Bao ; 24(7): 779-81, 2004 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-15257901

RESUMO

OBJECTIVE: To investigate the relationship between the expression of glucosylceramide synthase (GCS) mRNA in vincristine-resistant KBV(200) human cancer cell line and multidrug resistance (MDR) of the cancer cells. METHODS: Reverse transcriptional polymerase chain reaction (RT-PCR) was employed to analyze the differential expression of GCS mRNA between KBV(200) and KB cell lines and the changes in the mRNA expressions of GCS and mdr1 gene in KBV(200) cells after reversion of MDR. The effects of de-phenyl-z-palmaitoylamino-3-morpholine-1-propanol (DL-PPMP) and verapamil in reversing MDR of the cells were evaluated by MTT assay. RESULTS: KBV(200) cells exhibited significantly increased expressions of GCS and mdr1 gene, whereas mdr1 gene failed to be detected in the parental KB cells. DL-PPMP within the concentrations ranging from 5 to 25 micromol/L could inhibit the expression of GCS gene, with the maximum inhibition achieved at 25 micromol/L. Verapamil at the concentration of 10 micromol/L was already sufficient to induce inhibition of GCS expression in KVB(200) cells, which was more manifest with the concentration of 15 micromol/L. DL-PPMP and verapamil were found to inhibit mdr1 gene expression in KBV(200) cells at the mRNA level, and complete inhibition occurred after a 48-hour DL-PPMP treatment at 25 micromol/L. CONCLUSION: The inhibition of GCS and mdr1 gene expressions is positively correlated with the concentrations of DL-PPMP and verapamil, which can reverse MDR by inhibiting synthesis of GCS and mdr1 gene, indicating the positive correlation between the expression of GCS gene and MDR in KBV(200) cells. GCS gene might play an important role in MDR during tumor progression.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Glucosiltransferases/genética , RNA Mensageiro/análise , Vincristina/farmacologia , Linhagem Celular Tumoral , Resistência a Múltiplos Medicamentos , Genes MDR , Humanos
13.
Di Yi Jun Yi Da Xue Xue Bao ; 22(11): 1042-4, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12433646

RESUMO

OBJECTIVE: To synthesize a targeted drug delivery system for 5-fluorouracil (5Fu) using sulfadiazine (SF) as a carrier with reduced side-effects and strong antitumor activity. METHODS: SF-poly (ethylene glycol) (PEG) conjugate was initially synthesized. 5Fu was subjected to reaction with trichloromethyl chloroformate to prepare chloroformyl 5Fu, which was linked to a spacer hydroxyl group of PEG that served as a macromolecular linking arm between SF and 5Fu. The content of 5Fu in the conjugate was determined by ultraviolet spectrophotometry. Spectrum of ultraviolet and infrared along with differential scanning calorimetry were employed to identify the structure of the conjugate of SFPEG-end capped 5Fu. RESULTS: The drug loading content of the conjugate was 3.2 %, and structural analysis confirmed the linkage between 5Fu and SF via PEG. CONCLUSION: Targeted drug delivery system for 5Fu using SF as a carrier has been successfully synthesized by this means.


Assuntos
Antimetabólitos Antineoplásicos/síntese química , Sistemas de Liberação de Medicamentos , Fluoruracila/síntese química , Polietilenoglicóis/síntese química , Sulfadiazina/química , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/química , Portadores de Fármacos , Etilenoglicol/química , Fluoruracila/administração & dosagem , Fluoruracila/análogos & derivados , Fluoruracila/química , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Espectrofotometria Ultravioleta
14.
Di Yi Jun Yi Da Xue Xue Bao ; 23(12): 1297-300, 2003 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-14678895

RESUMO

OBJECTIVE: To observe the effects of three commonly used local ablation methods in the treatment of transplanted hepatocellular carcinoma in mice to provide experimental evidence for treating hepatocellular carcinoma that defies surgical removal. METHODS: Mouse models of hepatocellular carcinoma were established by means of subcutaneous transplantation, and treatment results of the three ablation methods, namely percutaneous ethanol injection therapy (PEIT), percutaneous acetic acid injection therapy (PAIT) and percutaneous local cryosurgery therapy (PLCT), were compared. RESULTS: The tumor inhibition rates of PLCT, PAIT and PEIT were 99.2%, 85.3%, and 72.8%, respectively. The rates for complete necrosis of the tumors were 100% (5/5), 60% (3/5), and 40% (2/5), with the survival time of 88.11+/-5.67, 86.67+/-7.26, and 72.89+/-12.86 days respectively. CONCLUSIONS: All of the three local ablation methods can inhibit the tumor growth to various degrees and prolong the survival time of the tumor-bearing mice. PLCT may yield relatively better result than the other two methods.


Assuntos
Ácido Acético/administração & dosagem , Criocirurgia , Etanol/administração & dosagem , Neoplasias Hepáticas Experimentais/terapia , Administração Cutânea , Animais , Injeções , Neoplasias Hepáticas Experimentais/mortalidade , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Transplante de Neoplasias
15.
Zhong Yao Cai ; 25(12): 917-8, 2002 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-12685354

RESUMO

OBJECTIVE: To study effect of Pill Fufang Zaofan together with radiotherapy and chemotherapy at same time. METHODS: Treat group administrated Pill Fufang Zaofan from 1 week before radiotherapy and chemotherapy, to 4 weeks after radiotherapy and chemotherapy. Control group didn't administrate Pill Fufang Zaofan. Other agents of two groups are no difference. RESULTS: White blood cells (WBC) of treat group were much more than those of control group. The cases of treat group in which WBC was lower than normal level were obviously less than those of control group. CONCLUSION: Pill Fufang Zaofan can protect bone marrow when using with radiotherapy and chemotherapy at same time.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/terapia , Medicamentos de Ervas Chinesas/administração & dosagem , Neoplasias Pulmonares/terapia , Fitoterapia , Adolescente , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Células da Medula Óssea/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Terapia Combinada , Feminino , Hematopoese/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/radioterapia , Masculino , Pessoa de Meia-Idade , Mitomicinas/efeitos adversos
16.
Breast Cancer ; 21(2): 223-30, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22711315

RESUMO

OBJECTIVE: Apogossypolone (ApoG2), a new derivative of gossypol, is a potent cell-growth inhibitor. ApoG2 has been demonstrated to have superior anti-tumor activity than gossypol in Bcl-2 transgenic mice. The purpose of this study was to investigate the inhibitory effect of ApoG2 on breast cancer cell line MCF-7 in vitro and in vivo, and to investigate its anti-tumor mechanism. METHODS: MCF-7 cell line in culture was treated with ApoG2. The inhibitory effects of ApoG2 on cell growth were measured by MTT and colony-formation assay. The cell apoptotic rate and cell cycle were analyzed by use of flow cytometry (FCM). The ultrastructural changes were observed by transmission electron microscopy. Autophagy was detected by acridine orange staining. Expression of Bcl-2, Bax, and Beclin 1 proteins was measured by western blot analysis. RESULTS: The inhibitory effect of ApoG2 on MCF-7 cell proliferation was dose and time-dependent. The maximum effect was observed when cells were incubated for 72 h with 40 µM ApoG2. ApoG2 at 5 µM also inhibited colony formation. FCM assay indicated that ApoG2 induced cell apoptosis and caused cell arrest in the S phase and G2/M phase. Transmission electron microscopic examination and acridine orange staining showed that ApoG2 induced intracellular autolysosome formation. Furthermore, ApoG2 reduced Bcl-2 expression, and enhanced expression of Bax and Beclin 1. Xenografting of MCF-7 cells in mice can also be inhibited by ApoG2. CONCLUSION: ApoG2, a novel anti-apoptotic Bcl-2 agent, inhibits proliferation of breast cancer cell line MCF-7 by inducing cell apoptosis and autophagy.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Gossipol/análogos & derivados , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Gossipol/farmacologia , Humanos , Células MCF-7/efeitos dos fármacos , Células MCF-7/ultraestrutura , Proteínas de Membrana/metabolismo , Camundongos Nus , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína X Associada a bcl-2/metabolismo
18.
Nan Fang Yi Ke Da Xue Xue Bao ; 30(8): 1903-5, 2010 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-20813697

RESUMO

OBJECTIVE: To study the antiangiogenetic and tumor inhibitory effects of endostatin (Es) by intratumoral versus intravenous administration combined with adriamycin (Adm) for treatment of transplanted tumor in mice. METHODS: Forty mice were subjected to subcutaneous implantation of H22 cells and randomly divided into 4 groups by the body weight when the tumor diameter reached 1 cm, namely the control group (with intratumoral and intravenous injection of normal saline), Es intratumoral group (with intratumoral injection Es and intraperitoneal Adm injection), Es vein group (with intravenous Es injection and intraperitoneal Adm injection), and Adm group (with intratumoral saline injection and intraperitoneal Adm injection). The tumor volumes and tumor inhibition rates were calculated, and the expression of vascular endothelial growth factor (VEGF) and the microvessel density (MVD) of the tumors were examined, with the survival time of the mice also observed. RESULTS: The tumor volume was smaller in Es intratumoral group than in the other groups (P<0.05). The expression of VEGF and M VD in Es intratumoral group was significantly decreased as compared with that in the other groups (P<0.05). The survival time was significantly longer in Es intratumoral group and Es vein group than in the other groups (P<0.05), but showed no significant difference between Es intratumoral group and Es vein group (P>0.05). CONCLUSION: In combination with Adm regimen, Es given intratumoral injection produces better effect than intravenous Es injection against angiogenesis and tumor growth, no significant difference can be found in the survival time between them.


Assuntos
Doxorrubicina/uso terapêutico , Endostatinas/administração & dosagem , Neoplasias Hepáticas/tratamento farmacológico , Administração Intravenosa , Animais , Quimioterapia Combinada , Endostatinas/uso terapêutico , Feminino , Injeções Intralesionais , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Nan Fang Yi Ke Da Xue Xue Bao ; 29(5): 887-9, 893, 2009 May.
Artigo em Chinês | MEDLINE | ID: mdl-19460700

RESUMO

OBJECTIVE: To study the effect of adenovirus (Ad)-mediated fusion gene system driven by the KDR promoter on the proliferation of human colon adenocarcinoma SW620 cells. METHODS: The KDR-expressing SW620 cells and LS174T cells not expressing KDR were both infected with AdEasy-KDR-CDglyTK followed by treatment with the prodrugs 5-FC and/or ganciclovir at different concentrations. The effect of the transfection on the cell proliferation was evaluated. RESULTS: The expression of green fluorescent protein (GFP) was observed in 95% of the infected SW620 and LS174T cells with a multiplicity of infection (MOI) of 100. Significant difference was not founded in the growth of SW620 and LS174T cells with or without the transfection. The infected SW620 cells exhibit high sensitivity to the prodrugs, but the infected LS174T cells did not (P<0.01). The CDglyTK fusion gene produced much stronger killing effect of on the target cells than either of the single suicide genes (P<0.01). CONCLUSION: CDglyTK fusion gene system driven by the KDR promoter selectively kills the KDR-CDglyTK SW620 cells and inhibits the cell proliferation.


Assuntos
Adenoviridae/genética , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/patologia , Genes Transgênicos Suicidas/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Adenocarcinoma/patologia , Adenoviridae/metabolismo , Linhagem Celular Tumoral , Citosina Desaminase/genética , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Timidina Quinase/genética , Transfecção , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
20.
Nan Fang Yi Ke Da Xue Xue Bao ; 29(5): 880-3, 2009 May.
Artigo em Chinês | MEDLINE | ID: mdl-19460698

RESUMO

OBJECTIVE: To obtain monoclonal antibodies (mAb) against LI-cadherin and investigate their effects on the proliferation of human hepatocellular carcinoma cells. METHODS: Balb/c mice were immunized with recombinant LI-cadherin, and hybridoma cell lines secreting monoclonal antibodies against LI-cadherin were established with routine cell fusion and subcloning approach. The specificity of these mAbs was determined by enzyme-linked immunosorbent assay (ELISA) and Western blotting. The effect of the mAbs obtained on the growth of HepG2 cells was assessed using inverted microscope and MTT assay. RESULTS: Two hybridoma cell lines (F001 and F002) stably secreting specific mAbs were obtained. Western blot analysis showed that the two antibodies specifically recognized LI-cadherin antigen derived from human eucaryotic cells or tissue. Treatment of the HepG2 cells with the mAbs resulted in reduced viable cell number and changes in the cell morphologies, and the two mAbs inhibited the proliferation of HepG2 cells in a concentration-dependent manner (P<0.05). CONCLUSION: The two specific mAbs obtained can inhibit the proliferation of HepG2 cells in vitro, which facilitates further study of the relationship between LI-cadherin and tumors.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/farmacologia , Caderinas/imunologia , Proliferação de Células/efeitos dos fármacos , Neoplasias Hepáticas/patologia , Animais , Anticorpos Monoclonais/imunologia , Caderinas/farmacologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Hibridomas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA