Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
China Journal of Chinese Materia Medica ; (24): 3472-3484, 2023.
Article in Chinese | WPRIM | ID: wpr-981482

ABSTRACT

Ginsenoside Rg_3, an active component of traditional Chinese medicine(TCM), was used as the substitute for cholesterol as the membrane material to prepare the ginsenoside Rg_3-based liposomes loaded with dihydroartemisinin and paclitaxel. The effect of the prepared drug-loading liposomes on triple-negative breast cancer in vitro was evaluated. Liposomes were prepared with the thin film hydration method, and the preparation process was optimized by single factor experiments. The physicochemical properties(e.g., particle size, Zeta potential, and stability) of the liposomes were characterized. The release behaviors of drugs in different media(pH 5.0 and pH 7.4) were evaluated. The antitumor activities of the liposomes were determined by CCK-8 on MDA-MB-231 and 4T1 cells. The cell scratch test was carried out to evaluate the effect of the liposomes on the migration of MDA-MB-231 and 4T1 cells. Further, the targeting ability of liposomes and the mechanism of lysosome escape were investigated. Finally, H9c2 cells were used to evaluate the potential cardiotoxicity of the preparation. The liposomes prepared were spheroid, with uniform particle size distribution, the ave-rage particle size of(107.81±0.01) nm, and the Zeta potential of(2.78±0.66) mV. The encapsulation efficiency of dihydroartemisinin and paclitaxel was 57.76%±1.38% and 99.66%±0.07%, respectively, and the total drug loading was 4.46%±0.71%. The accumulated release of dihydroartemisinin and paclitaxel from the liposomes at pH 5.0 was better than that at pH 7.4, and the liposomes could be stored at low temperature for seven days with good stability. Twenty-four hours after administration, the inhibition rates of the ginsenoside Rg_3-based liposomes loaded with dihydroartemisinin(70 μmol·L~(-1)) and paclitaxel on MDA-MB-231 and 4T1 cells were higher than those of the positive control(adriamycin) and free drugs(P<0.01). Compared with free drugs, liposomes inhibited the migration of MDA-MB-231 and 4T1 cells(P<0.05). Liposomes demonstrated active targeting and lysosome escape. In particular, liposomes showed lower toxicity to H9c2 cells than free drugs(P<0.05), which indicated that the preparation had the potential to reduce cardiotoxicity. The findings prove that ginsenoside Rg_3 characterized by the combination of drug and excipient is an ideal substitute for lipids in liposomes and promoted the development of innovative TCM drugs for treating cancer.


Subject(s)
Humans , Paclitaxel/pharmacology , Liposomes/chemistry , Ginsenosides/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Cardiotoxicity/drug therapy , Cell Line, Tumor
2.
Rev. Assoc. Med. Bras. (1992) ; 68(2): 159-164, Feb. 2022. tab, graf
Article in English | LILACS | ID: biblio-1365364

ABSTRACT

SUMMARY OBJECTIVE: The objective of this study was to explore the molecular mechanism underlying the occurrence of benign bile duct stricture and the target of low-dose paclitaxel in the prevention of benign bile duct stricture. METHODS: Under the stimulation of transforming growth factor beta 1, the expression of collagen type I and connective tissue growth factor were detected on isolated primary fibroblasts. The phosphorylation levels of JNK and Smad2L were detected using Western blot. The effect of low-dose paclitaxel on the transforming growth factor beta 1-induced inhibition of type I collagen and connective tissue growth factor expression and JNK and Smad2L phosphorylation was also observed. RESULTS: Transforming growth factor beta 1 induced the secretion of type I collagen and connective tissue growth factor as well as JNK phosphorylation in biliary fibroblasts. The JNK inhibitor or siRNA-Smad2 inhibited the transforming growth factor beta 1-induced secretion of type I collagen and connective tissue growth factor. Low-dose paclitaxel inhibited the expression of type I collagen induced by transforming growth factor beta 1 and may inhibit the secretion of collagen in biliary fibroblasts. CONCLUSION: The activation of JNK/Smad2L induced by transforming growth factor beta 1 is involved in the occurrence of benign bile duct stricture that is mediated by the overexpression of type I collagen and connective tissue growth factor, and low-dose paclitaxel may inhibit the phosphorylation of JNK/Smad2L.


Subject(s)
Humans , Paclitaxel/pharmacology , Collagen , MAP Kinase Signaling System , Collagen Type I/metabolism , Collagen Type I/pharmacology , Smad2 Protein , Fibroblasts/metabolism
3.
China Journal of Chinese Materia Medica ; (24): 2457-2464, 2022.
Article in Chinese | WPRIM | ID: wpr-928125

ABSTRACT

In view of the longevity and innate immune escape of red blood cells, this study designed the red blood cell membrane-coated paclitaxel nanosuspension [RBC-(PTX)NS] and investigated its physicochemical properties and antitumor effect in vitro. Paclitaxel nanosuspension [(PTX)NS] was prepared by ultrasonic precipitation and then RBC-(PTX)NS by ultrasonic coating. The formulation of(PTX)NS was optimized with Box-Behnken method and indexes of particle diameter, zeta potential, and stability. The morphology, particle diameter, stability, in vitro dissolution, and antitumor effect of(PTX)NS and RBC-(PTX)NS were characterized. The results showed that the particle diameter and zeta potential were(129.38±0.92) nm and(-22.41±0.48) mV, respectively, for the optimized(PTX)NS, while(142.5±0.68) nm and(-29.85±0.53) mV, respectively, for RBC-(PTX)NS. Under the transmission electron microscope,(PTX)NS was spherical and RBC-(PTX)NS had obvious core-shell structure. RBC-(PTX)NS remained stable for 5 days at 4 ℃. The in vitro dissolution test demonstrated that the cumulative release rate of RBC-(PTX)NS reached 79% within 20 min, which was significantly higher than that(25%) of(PTX)NS(P<0.05). As evidenced by MTT assay, RBC-(PTX)NS highly inhibited the proliferation of HepG2 cells in a dose-dependent manner. The cell membrane-coated nano-preparation preparation method is simple and reproducible. It improves the solubility of PTX and endows RBC-(PTX)NS with higher stability and stronger cytotoxicity. Thus, it is a new method for the delivery of PTX via nanocrystallization.


Subject(s)
Erythrocyte Membrane , Nanoparticles/chemistry , Paclitaxel/pharmacology , Particle Size , Suspensions
4.
Journal of Southern Medical University ; (12): 26-35, 2022.
Article in Chinese | WPRIM | ID: wpr-936281

ABSTRACT

OBJECTIVE@#To construct a polylactic acid-glycolic acid-polyethylene glycol (PLGA-PEG) nanocarrier (N-Pac-CD133) coupled with a CD133 nucleic acid aptamer carrying paclitaxel for eliminating lung cancer stem cells (CSCs).@*METHODS@#Paclitaxel-loaded N-Pac-CD133 was prepared using the emulsion/solvent evaporation method and characterized. CD133+ lung CSCs were separated by magnetic bead separation and identified for their biological behaviors and gene expression profile. The efficiency of paclitaxel-loaded N-Pac-CD133 for targeted killing of lung cancer cells was assessed in vitro. SCID mice were inoculated with A549 cells and received injections of normal saline, empty nanocarrier linked with CD133 aptamer (N-CD133), paclitaxel, paclitaxel-loaded nanocarrier (N-Pac) or paclitaxel-loaded N-Pac-CD133 (n=8, 5 mg/kg paclitaxel) on days 10, 15 and 20, and the tumor weight and body weight of the mice were measured on day 40.@*RESULTS@#Paclitaxel-loaded N-Pac-CD133 showed a particle size of about 100 nm with a high encapsulation efficiency (>80%) and drug loading rate (>8%), and was capable of sustained drug release within 48 h. The CD133+ cell population in lung cancer cells showed the characteristic features of lung CSCs, including faster growth rate (30 days, P=0.001) and high expressions of tumor stem cell markers OV6(P < 0.001), CD133 (P=0.001), OCT3/4 (P=0.002), EpCAM (P=0.04), NANOG (P=0.005) and CD44 (P=0.02). Compared with N-Pac and free paclitaxel, paclitaxel-loaded N-Pac-CD133 showed significantly enhanced targeting ability and cytotoxicity against lung CSCs in vitro (P < 0.001) and significantly reduced the formation of tumor spheres (P < 0.001). In the tumor-bearing mice, paclitaxel-loaded N-Pac-CD133 showed the strongest effects in reducing the tumor mass among all the treatments (P < 0.001).@*CONCLUSION@#CD133 aptamer can promote targeted delivery of paclitaxel to allow targeted killing of CD133+ lung CSCs. N-Pac-CD133 loaded with paclitaxel may provide an effective treatment for lung cancer by targeting the lung cancer stem cells.


Subject(s)
Animals , Mice , Cell Line, Tumor , Drug Carriers , Lung , Mice, SCID , Nanoparticles , Neoplasms , Neoplastic Stem Cells , Paclitaxel/pharmacology , Polyethylene Glycols/pharmacology
5.
Int. j. morphol ; 39(2): 564-570, abr. 2021. ilus, tab, graf
Article in English | LILACS | ID: biblio-1385368

ABSTRACT

SUMMARY: Cancer known as a malignant tumor, is a class of diseases involving abnormal cell growth with the potential to invade or spread to other parts of the body. The Ehrlich tumor is a mammary adenocarcinoma of mice developed in solid and ascitic forms. This study was aimed to investigate the effects of paclitaxel on Netrin 1 and Factor 8 expression and also in tumor cell proliferation, apoptosis, angiogenesis, and development of tumor in Ehrlich solid tumors treated with paclitaxel. In this study, 26 adult Balb/C male mice were used. 6 of them were used as stock. Ehrlich ascites cells taken from animals in stock were injected subcutaneously from the neck area to all animals. The mice were randomly assigned to two groups of ten rats per group. Paclitaxel treatment group 10 mg/kg were administered to mice intraperitoneally (i.p.) 4,9, and 14th days. 15th day the animals were sacrificed and tumor tissues were taken. Paraffin-embedded solid tumor sections were stained Hematoxylin & Eosin, Masson's Trichrome. Also solid tumor sections were stained immunohistochemically with Netrin1 and Factor 8. Tunel method was applied to determine apoptosis. Paclitaxel applied as a therapeutic Ehrlich solid tumor reduced the volume of tumors in the treatment groups. At the end of the experiments, in the treatment groups' significantly reduced the Netrin 1 expression and microvessel density compared to the group control. Also paclitaxel in the treatment group increased the number of apoptotic cells. We suggest that decreasing the expression of Netrin 1 would be reduced vessel density and increased apoptosis.


RESUMEN: El cáncer, conocido como tumor maligno, es una clase de enfermedad que involucra un crecimiento celular anormal con potencial de invadir o diseminarse a otras partes del cuerpo. El tumor de Ehrlich es un adenocarcinoma mamario de ratones desarrollado en formas sólidas y ascíticas. Este estudio tuvo como objetivo investigar los efectos del paclitaxel en la expresión de Netrin 1 y Factor 8 y también en la proliferación de células tumorales, apoptosis, angiogénesis y desarrollo de tumores sólidos de Ehrlich tratados con paclitaxel. En esta investigación se utilizaron 26 ratones machos Balb / C adultos. Seis de ellos se utilizaron como stock. Se inyectaron por vía subcutánea células de ascitis de Ehrlich tomadas de animales en la zona del cuello. Los ratones se asignaron aleatoriamente a dos grupos de diez ratas por grupo. Se administraron 10 mg/kg del grupo de tratamiento con paclitaxel a ratones por vía intraperitoneal (i.p.) 4, 9 y 14 días. El día 15 se sacrificaron los animales y se extrajeron los tejidos tumorales. Las secciones de tumor sólido incluidas en parafina se tiñeron con hematoxilina y eosina y tricrómico de Masson. También se tiñeron inmunohisto-químicamente secciones de tumor sólido con Netrin1 y Factor 8. Se aplicó el método Tunel para determinar la apoptosis. El paclitaxel aplicado como tumor sólido terapéutico de Ehrlich redujo el volumen de tumores en los grupos de tratamiento. Al final de los experimentos, en los grupos de tratamiento se redujo significativamente la expresión de Netrin 1 y la densidad de microvasos en comparación con el grupo control. Además, el paclitaxel en el grupo tratamiento aumentó el número de células apoptóticas. Sugerimos que la disminución de la expresión de Netrin 1 reduciría la densidad de los vasos y aumentaría la apoptosis.


Subject(s)
Animals , Male , Mice , Carcinoma, Ehrlich Tumor/drug therapy , Paclitaxel/administration & dosage , Netrin-1/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/administration & dosage , Factor VIII , Immunohistochemistry , Paclitaxel/pharmacology , Apoptosis , Cell Proliferation/drug effects , Microvascular Density/drug effects , Mice, Inbred BALB C , Neovascularization, Pathologic , Antineoplastic Agents, Phytogenic/pharmacology
6.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 890-897, 2020.
Article in English | WPRIM | ID: wpr-881034

ABSTRACT

Paclitaxel, a tetracyclic diterpenoid compounds, was firstly isolated from the bark of the Pacific yew trees. Currently, as a low toxicity, high efficiency, and broad-spectrum natural anti-cancer drug, paclitaxel has been widely used against ovarian cancer, breast cancer, uterine cancer, and other cancers. As the matter of fact, natural paclitaxel from Taxus species has been proved to be environmentally unsustainable and economically unfeasible. For this reason, researchers from all over the world are devoted to searching for new ways of obtaining paclitaxel. At present, other methods, including artificial cultivation of Taxus plants, microbial fermentation, chemical synthesis, tissue and cell culture have been sought and developed subsequently. Meanwhile, the biosynthesis of paclitaxel is also an extremely attractive method. Unlike other anti-cancer drugs, paclitaxel has its unique anti-cancer mechanisms. Here, the source, production, and anti-cancer mechanisms of paclitaxel were summarized and reviewed, which can provide theoretical basis and reference for further research on the production, anti-cancer mechanisms and utilization of paclitaxel.


Subject(s)
Humans , Antineoplastic Agents, Phytogenic/pharmacology , Neoplasms/drug therapy , Paclitaxel/pharmacology
7.
Journal of Korean Medical Science ; : 360-370, 2016.
Article in English | WPRIM | ID: wpr-95370

ABSTRACT

Cancer stem cells (CSCs) have tumor initiation, self-renewal, metastasis and chemo-resistance properties in various tumors including colorectal cancer. Targeting of CSCs may be essential to prevent relapse of tumors after chemotherapy. Phosphatidylinositol-3-kinase (PI3K) and mammalian target of rapamycin (mTOR) signals are central regulators of cell growth, proliferation, differentiation, and apoptosis. These pathways are related to colorectal tumorigenesis. This study focused on PI3K and mTOR pathways by inhibition which initiate differentiation of SW620 derived CSCs and investigated its effect on tumor progression. By using rapamycin, LY294002, and NVP-BEZ235, respectively, PI3K and mTOR signals were blocked independently or dually in colorectal CSCs. Colorectal CSCs gained their differentiation property and lost their stemness properties most significantly in dual-blocked CSCs. After treated with anti-cancer drug (paclitaxel) on the differentiated CSCs cell viability, self-renewal ability and differentiation status were analyzed. As a result dual-blocking group has most enhanced sensitivity for anti-cancer drug. Xenograft tumorigenesis assay by using immunodeficiency mice also shows that dual-inhibited group more effectively increased drug sensitivity and suppressed tumor growth compared to single-inhibited groups. Therefore it could have potent anti-cancer effects that dual-blocking of PI3K and mTOR induces differentiation and improves chemotherapeutic effects on SW620 human colorectal CSCs.


Subject(s)
Animals , Humans , Male , Mice , AC133 Antigen/genetics , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chromones/pharmacology , Colorectal Neoplasms/drug therapy , Imidazoles/pharmacology , Mice, Inbred BALB C , Mice, Nude , Morpholines/pharmacology , Neoplastic Stem Cells/cytology , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Quinolines/pharmacology , SOXB1 Transcription Factors/genetics , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
8.
Yonsei Medical Journal ; : 1643-1650, 2015.
Article in English | WPRIM | ID: wpr-70407

ABSTRACT

PURPOSE: Bone metastasis invariably increases morbidity and mortality. This study compares the effects of ibandronate and paclitaxel on bone structure and its mechanical properties and biochemical turnover in resorption markers using an immunocompetent Walker 256-Sprague-Dawley model, which was subjected to tumor-induced osteolysis. MATERIALS AND METHODS: Seventy rats were divided equally into 4 groups: 1) sham group (SHAM), 2) tumor group (CANC), 3) ibandronate treated group (IBAN), and 4) paclitaxel treated group (PAC). Morphological indices [bone volume fraction (BV/TV), trabecular number (Tb.N), trabecular thickness (Tb.Th), trabecular separation (Tb.Sp)] and mechanical properties (failure load, stiffness) were evaluated after thirty days of treatment period. Bone resorption rate was analysed using serum deoxypyridinoline (Dpd) concentrations. RESULTS: Morphological indices showed that ibandronate (anti-resorptive drug) had a better effect in treating tumor-induced architectural changes in bone than paclitaxel (chemotherapeutic drug). The deterioration in bone architecture was reflected in the biomechanical properties of bone as studied with decreased failure load (F(x)) and stiffness (S) of the bone on the 30th day postsurgery. Dpd concentrations were significantly lower in the IBAN group, indicating successful inhibition of bone resorption and destruction. CONCLUSION: Ibandronate was found to be as effective as higher doses of paclitaxel in maintaining stiffness of bone. Paclitaxel treatment did not appear to inhibit osteoclast resorption, which is contrary to earlier in-vitro literature. Emphasis should be placed on the use of immunocompetent models for examining drug efficacy since it adequately reflects bone metastasis in clinical scenarios.


Subject(s)
Animals , Male , Rats , Amino Acids , Biomechanical Phenomena/drug effects , Bone Density/drug effects , Bone Neoplasms/drug therapy , Bone Resorption/chemically induced , Diphosphonates/pharmacology , Immunocompetence , Neoplasm Metastasis , Osteolysis , Paclitaxel/pharmacology , Rats, Sprague-Dawley
9.
Mem. Inst. Oswaldo Cruz ; 108(1): 84-90, Feb. 2013. ilus, graf, tab
Article in English | LILACS | ID: lil-666049

ABSTRACT

The trypanosomatid cytoskeleton is responsible for the parasite's shape and it is modulated throughout the different stages of the parasite's life cycle. When parasites are exposed to media with reduced osmolarity, they initially swell, but subsequently undergo compensatory shrinking referred to as regulatory volume decrease (RVD). We studied the effects of anti-microtubule (Mt) drugs on the proliferation of Leishmania mexicana promastigotes and their capacity to undergo RVD. All of the drugs tested exerted antiproliferative effects of varying magnitudes [ansamitocin P3 (AP3)> trifluoperazine > taxol > rhizoxin > chlorpromazine]. No direct relationship was found between antiproliferative drug treatment and RVD. Similarly, Mt stability was not affected by drug treatment. Ansamitocin P3, which is effective at nanomolar concentrations, blocked amastigote-promastigote differentiation and was the only drug that impeded RVD, as measured by light dispersion. AP3 induced 2 kinetoplasts (Kt) 1 nucleus cells that had numerous flagella-associated Kts throughout the cell. These results suggest that the dramatic morphological changes induced by AP3 alter the spatial organisation and directionality of the Mts that are necessary for the parasite's hypotonic stress-induced shape change, as well as its recovery.


Subject(s)
Animals , Mice , Cytoskeleton/drug effects , Leishmania mexicana/drug effects , Tubulin Modulators/pharmacology , Chlorpromazine/pharmacology , Leishmania mexicana/growth & development , Macrolides/pharmacology , Maytansine/analogs & derivatives , Maytansine/pharmacology , Paclitaxel/pharmacology , Trifluoperazine/pharmacology
10.
Yonsei Medical Journal ; : 374-380, 2013.
Article in English | WPRIM | ID: wpr-89573

ABSTRACT

PURPOSE: Hypoxia-inducible factor-1alpha (HIF-1alpha) increases transcription of the vascular endothelial growth factor (VEGF) gene. Inhibition of VEGF abolishes VEGF mediated induction of HIF-1alpha. Recent reports suggested that HIF-1alpha also mediated the induction of class III beta-tubulin (TUBB3) in hypoxia. TUBB3 confers resistance to taxanes. Inhibition of VEGF may decrease the expression of HIF-1alpha and TUBB3. This study was undertaken to investigate the roles of vascular endothelial growth factor receptor (VEGFR) in gastric cancer cell behavior and to identify methods to overcome paclitaxel resistance in vitro. MATERIALS AND METHODS: The protein expression levels of HIF-1alpha and TUBB3 were measured in human gastric cancer cell lines (AGS) under normoxic and hypoxic conditions. The relationship between TUBB3 and paclitaxel resistance was assessed with small interfering TUBB3 RNA. AGS cells were treated with anti-VEGFR-1, anti-VEGFR-2, placental growth factor (PlGF), bevacizuamb, and paclitaxel. RESULTS: Hypoxia induced paclitaxel resistance was decreased by knockdown of TUBB3. Induction of HIF-1alpha and TUBB3 in AGS is VEGFR-1 mediated and PlGF dependent. Hypoxia-dependent upregulation of HIF-1alpha and TUBB3 was reduced in response to paclitaxel treatment. Expressions of HIF-1alpha and TUBB3 were most decreased when AGS cells were treated with a combination of paclitaxel and anti-VEGFR-1. AGS cell cytotoxicity was most increased in response to paclitaxel, anti-VEGFR-1, and anti-VEGFR-2. CONCLUSION: We suggest that blockade of VEGFR-1 and VEGFR-2 enhances paclitaxel sensitivity in TUBB3-expressing gastric cancer cells.


Subject(s)
Humans , Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Hypoxia , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Paclitaxel/pharmacology , Pregnancy Proteins/pharmacology , Stomach Neoplasms/drug therapy , Tubulin/genetics , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
11.
Experimental & Molecular Medicine ; : 633-641, 2012.
Article in English | WPRIM | ID: wpr-149765

ABSTRACT

Triptolide, a compound extracted from the traditional Chinese medicine preparation of Tripterygium wilfordii Hook F., has been reported to have anti-inflammatory and anti-cancer activities. However, its effect on ovarian cancer invasion is unknown. We observed that MMP7 and MMP19 expression increased in ovarian cancer tissue. Triptolide treatment inhibited the migration and invasion of ovarian cancer cells SKOV3 and A2780 at the concentration of 15 nM. We also observed that triptolide suppressed MMP7 and MMP19 promoter activity in a dose-dependent manner, down-regulating the expressions of these promoters on mRNA and protein level. Moreover, triptolide enhanced E-cadherin expression in ovarian cancer cells. In vivo, triptolide inhibited tumor formation and metastasis in nude mice, and suppressed MMP7 and MMP19 expression; it also enhanced E-cadherin expression in tumor in a dose-dependent manner. Over expression of MMP7 and MMP19, or suppression of E-cadherin expression partially abolished the inhibitory effect of triptolide on invasion of ovarian cancer cells. To summarize, triptolide significantly inhibited the migration and invasion of ovarian cancer cells by suppression of MMP7 and MMP19 and up-regulation of E-cadherin expression. This study shows that triptolide is a good candidate for the treatment of ovarian cancer and reduction of metastasis.


Subject(s)
Animals , Female , Humans , Mice , Antineoplastic Agents, Alkylating/pharmacology , Cadherins/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cystadenocarcinoma, Serous/drug therapy , Diterpenes/pharmacology , Epoxy Compounds/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinases, Secreted/genetics , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , Phenanthrenes/pharmacology , Promoter Regions, Genetic , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
12.
São Paulo; s.n; s.n; dez. 2011. 130 p. tab, graf, ilus.
Thesis in Portuguese | LILACS | ID: biblio-837173

ABSTRACT

Uma nova alternativa para o tratamento do câncer foi proposta em estudos anteriores, consistindo no uso de uma nanoemulsão lipídica como transportadora de agentes quimioterápicos às células neoplásicas. A redução da toxicidade da quimioterapia promovida pelo direcionamento específico de quimioterápicos às células tumorais nos levou a testar o potencial de aplicação do sistema de nanopartículas lipídicas na terapêutica combinada do paclitaxel com a sinvastatina, um agente hipolipemiante que pode ser empregado como coadjuvante no tratamento do câncer. Nos dias 11, 14 e 19 após a inoculação de células de melanoma B16F10, camundongos C57BL/6J receberam pela via intraperitoneal soluções de oleato de paclitaxel associado à nanoemulsão lipídica 17,5µmol/kg (Nano-paclitaxel), formulação comercial do paclitaxel 17,5µmol/kg, nanoemulsão lipídica (Nanoemulsão) e solução salina (Controle). A sinvastatina 50mg/kg/dia foi administrada por gavagem do 11° ao 19° dia após a inoculação do tumor em um dos grupos de animais tratados com o Nano-paclitaxel (Nano-paclitaxel + Sinva), no grupo tratado com a formulação comercial do paclitaxel (Paclitaxel + Sinva) e como monoterapia (Sinva). Camundongos Balb-c saudáveis receberam os mesmos tratamentos para avaliação dos possíveis efeitos tóxicos dos diferentes tratamentos. A terapia combinada Nano-paclitaxel + Sinva apresentou toxicidade negligível em comparação com a terapia combinada Paclitaxel + Sinva que provocou perda de peso e mielossupressão nos animais. Nos animais portadores de tumor, o tratamento Nano-paclitaxel + Sinva inibiu 95% do crescimento tumoral, comparado à inibição de 44% promovida pelo tratamento Paclitaxel + Sinva. Além disso, apenas 37% dos animais portadores de melanoma submetidos ao tratamento com Nano-paclitaxel + Sinva apresentaram metástases, em contraste com 90% dos tratados com Paclitaxel + Sinva. A probabilidade de sobrevida também foi maior nos camundongos tratados com o Nano-paclitaxel + Sinva em comparação aos tratados com Paclitaxel + Sinva. A análise de amostras de tumores por citometria de fluxo mostrou que somente nos grupos de animais tratados com Sinva, Nano-paclitaxel ou com a combinação Nano-paclitaxel + Sinva houve aumento na expressão de p21 em comparação ao grupo Controle. Da mesma forma, apenas nos grupos Sinva e Nano-paclitaxel + Sinva houve redução na expressão de ciclina D1 em comparação ao grupo Controle. O teste de viabilidade celular com rodamina 123 mostrou despolarização da membrana mitocondrial com redução no número de células tumorais viáveis em todos os grupos de tratamentos em comparação aos grupos Nanoemulsão e Controle. A avaliação histológica dos tumores demonstrou que os grupos Nanoemulsão e Controle apresentaram alta densidade de células tumorais, diferentemente dos demais grupos de tratamento e que apenas os tumores do grupo Nano-paclitaxel + Sinva apresentaram aumento na presença de fibras de colágeno tipo I e III. Em comparação ao grupo Controle, os tumores dos grupos Sinva, Paclitaxel + Sinva, Nano-paclitaxel e Nano-paclitaxel + Sinva apresentaram redução na expressão imunohistoquímica de ICAM, MCP-1 e MMP-9 sendo que o grupo Nano-paclitaxel + Sinva apresentou a menor porcentagem de área marcada positivamente para a MMP-9. A terapia combinada com Nano-paclitaxel + Sinva é menos tóxica e mais efetiva na inibição do crescimento tumoral do que a mesma terapia com a formulação comercial do paclitaxel


In previous studies we have proposed a novel approach for cancer treatment consisting of the use of a lipid nanoemulsion as a vehicle to direct chemotherapeutic agents to neoplastic cells. Reduction of chemotherapy toxicity promoted by specific targeting of antineoplastic agents to tumor cells led us to test the application of the lipidic nanoparticle system in combined treatment with paclitaxel and simvastatin, a cholesterol-lowering drug that can be used as coadjuvant in cancer treatment. On days 11, 14 and 19 after B16F10 melanoma cells inoculation, C57BL/6J mice were intraperitoneally injected with paclitaxel oleate associated to the lipidic nanoemulsion 17.5 µmol/kg (Nano-paclitaxel), commercial formulation of paclitaxel 17.5 µmol/kg, lipidic nanoemulsion (Nanoemulsion) or saline solution (Control). Simvastatin 50 mg/kg/day was administered by gavage from days 11 to 19 after tumor inoculation in one group of animals treated with Nano-paclitaxel (Nano-paclitaxel + Simva), in the group treated with commercial formulation of paclitaxel (Paclitaxel + Simva) and as monotherapy (Simva). Evaluation of possible toxic effects of the treatments was accessed in healthy Balb-c mice. Combined therapy with Nano-paclitaxel + Simva showed negligible toxicity as compared with the combination of Paclitaxel + Simva which resulted in animal weight loss and myelosuppression. In tumor-bearing animals, treatment with Nano-paclitaxel + Simva resulted in a remarkable tumor growth inhibition rate of 95%, compared to a 44% inhibition rate promoted by treatment with Paclitaxel + Simva. Moreover, only 37% of melanoma bearing animals treated with Nano-paclitaxel + Simva developed metastasis, in contrast to 90% of those treated with Paclitaxel + Simva. Survival rates were also higher in mice treated with Nano-paclitaxel + Simva in comparison to Paclitaxel + Simva treated animals. Analysis of tumor samples by flow cytometry showed that only animals treated with Simva, Nano-paclitaxel or Nano-paclitaxel + Simva increased the expression of p21 in comparison to Control group. Also, tumors from animals treated with Simva or Nano-paclitaxel + Simva presented a decrease in the expression of cyclin D1 in comparison to Control group. Cell viability test with rhodamine 123 showed mitochondrial membrane depolarization with reduction of tumor viable cells in all treatment groups in comparison to Nanoemulsion and Control groups. The histological study revealed that in contrast to drugs treated groups, tumors from Nanoemulsion and Control groups presented high tumor cell density and only Nano-paclitaxel + Simva treated animals presented tumors with increased presence of collagen fibers I and III. In comparison to Control group, tumors from groups Simva, Paclitaxel + Simva, Nano-paclitaxel and Nano-paclitaxel + Simva showed a reduction in immunohistochemical expression of ICAM, MCP-1 and MMP-9 and the group Nano-paclitaxel + Simva presented the lowest percentage of area positively stained for MMP-9. Combined therapy with Nano-paclitaxel + Simva was less toxic and more effective in promoting tumor growth inhibiton than the same combined therapy with the commercial formulation of paclitaxel


Subject(s)
Animals , Male , Mice , Voluntary Health Agencies , Paclitaxel/pharmacology , Simvastatin/pharmacology , Pharmaceutical Preparations/analysis , Drug Therapy/methods , Molecular Targeted Therapy , Neoplasms
13.
Braz. j. biol ; 70(3): 659-664, Aug. 2010. ilus, tab
Article in English | LILACS | ID: lil-555287

ABSTRACT

Usnic acid, a lichen metabolite, is known to exert antimitotic and antiproliferative activities against normal and malignant human cells. Many chemotherapy agents exert their activities by blocking cell cycle progression, inducing cell death through apoptosis. Microtubules, protein structure involved in the segregation of chromosomes during mitosis, serve as chemotherapeutical targets due to their key role in cellular division as well as apoptosis. The aim of this work was to investigate whether usnic acid affects the formation and/or stabilisation of microtubules by visualising microtubules and determining mitotic indices after treatment. The breast cancer cell line MCF7 and the lung cancer cell line H1299 were treated with usnic acid 29 µM for 24 hours and two positive controls: vincristine (which prevents the formation of microtubules) or taxol (which stabilizes microtubules). Treatment of MCF7 and H1299 cells with usnic acid did not result in any morphological changes in microtubules or increase in the mitotic index. These results suggest that the antineoplastic activity of usnic acid is not related to alterations in the formation and/or stabilisation of microtubules.


O ácido úsnico, um metabólito de liquens, é conhecido por sua atividade antimitótica e antiproliferativa em células humanas normais e malignas. Muitos quimioterápicos exercem suas atividades bloqueando a progressão do ciclo celular e induzindo morte celular por apoptose. Os microtúbulos, estruturas protéicas envolvidas na segregação dos cromossomos durante a mitose, servem como alvo quimioterapêutico devido ao seu importante papel tanto na divisão celular quanto nos mecanismos de morte celular por apoptose. O objetivo deste trabalho foi investigar se o ácido úsnico afeta a formação e/ou estabilização dos microtúbulos, a partir da visualização de microtúbulos e determinação de índices mitóticos após o tratamento. Células de câncer de mama MCF7 e de câncer de pulmão H1299 foram tratadas por 24 horas com 29 µM de ácido úsnico e dois controles positivos: vincristina (que impede a formação de microtúbulos) e taxol (que estabiliza microtúbulos). O tratamento das células MCF7 e H1299 com o ácido úsnico não resultou em aumento do índice mitótico. Os resultados sugerem que a atividade antineoplásica do ácido úsnico não está relacionada a alterações na formação e/ou estabilização de microtúbulos.


Subject(s)
Female , Humans , Antimitotic Agents/pharmacology , Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Microtubules/drug effects , Paclitaxel/pharmacology , Vincristine/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor/drug effects , Lung Neoplasms/pathology
15.
Rev. venez. oncol ; 21(2): 85-91, abr.-jun. 2009. tab
Article in Spanish | LILACS | ID: lil-549466

ABSTRACT

Demostrar que la cirugía de intervalo constituye una opción terapéutica efectiva en el aumento de la resecabilidad tumoral, mejorando la sobrevida de aquellas pacientes con tumores epiteliales de ovario clasificados como estadio III y catalogados como "no resecables". Estudio retrospectivo y descriptivo de las pacientes pertenecientes al servicio de ginecología del Hospital Oncológico "Padre Machado" con el diagnóstico de tumor epitelial de ovario clasificados como estadio III, incluidos en un protocolo de cirugía de intervalo, en el período septiembre 2002- septiembre 2007. Fueron identificadas 13 pacientes. El esquema de quimioterapia utilizado en todos los casos fue el de paclitaxel a dosis de 175mg/m² y carboblatino 5ABC, con una media de 4,46 ciclos. Se estableció una respuesta posterior a la quimioterapia primaria mayor al 50 por ciento 76,92 por ciento de los pacientes. La cirugía de intervalo aumenta el porcentaje de resecabilidad y disminuye el porcentaje de complicaciones posoperatorias, no evidenciándose beneficios estadísticamente significativos en términos de aumento de la sobrevida libre de enfermedad.


To demonstrate that interval debulking surgery constitutes an effective therapeutic option in the increase of the tumor like resected, improving the over life of those patients with epithelial tumors of ovary classified III stage, catalogued like “none resected”. Retrospective and descriptive study of the patients that assisted the service of gynecology of the Oncology Hospital “Padre Machado” with the diagnosis of epithelial tumor of ovary III stage, included in protocol interval debulking surgery, during the period September 2002-September 2007. 13 patients were identified. The scheme of chemotherapy used in all the cases was paclitaxel at dose of 175mg/m² and carboblatin 5ABC, with a media of 4.46 cycles. An answer greater than 50 % was settled down subsequent to the primary chemotherapy in a 76.92 % of the patients. The interval debulking surgery increases the percentage of respectability and diminishes the percentage of postoperative complications, not demonstrating itself significant benefits in terms of increase in the free disease period.


Subject(s)
Humans , Adult , Female , Middle Aged , Ovarian Neoplasms/surgery , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Paclitaxel/administration & dosage , Carcinoma/pathology , Medical Oncology , Paclitaxel/pharmacology
16.
Journal of Veterinary Science ; : 99-103, 2009.
Article in English | WPRIM | ID: wpr-221148

ABSTRACT

Taxol has been used effectively in cancer therapies. Our previous study demonstrated that taxol induced altered maturation and improved viability of dendritic cells (DCs). However, the effects of taxol on DC viability have not been fully elucidated. In the present study, flow cytometric analyses revealed that taxol treatment significantly increased the number of viable DCs and the expression levels of a representative anti-apoptotic protein Bcl-xL. Furthermore, mobilization of the p65 subunit of nuclear factor-kappaB (NF-kappaB) from the cytosol to the nucleus in DCs was observed by confocal microscopy. An inhibition assay using N-p-tosyl-L-phenylalanine chloromethyl ketone confirmed that NF-kappaB was intimately involved in the effects of taxol on DC viability. In addition, we investigated the mechanisms of taxol enhancement of DC viability. Since taxol is a popular anticancer agent used in clinic, this study may provide a rationale for the use of taxol in DC immunotherapy to treat cancer patients. Taken together, these results confirm that taxol increases DC viability, and this information may provide new insights for new clinical applications of both taxol and DCs.


Subject(s)
Animals , Female , Mice , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Cell Survival/drug effects , Dendritic Cells/cytology , Flow Cytometry , Interleukin-12/physiology , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Confocal , Paclitaxel/pharmacology , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , Transcription Factor RelA/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , bcl-X Protein/physiology
17.
Experimental & Molecular Medicine ; : 737-745, 2009.
Article in English | WPRIM | ID: wpr-71509

ABSTRACT

Paclitaxel is one of the chemotheraputic drugs widely used for the treatment of nonsmall cell lung cancer (NSCLC) patients. Here, we tested the ability of alpha-tocopheryl succinate (TOS), another promising anticancer agent, to enhance the paclitaxel response in NSCLC cells. We found that sub-apoptotic doses of TOS greatly enhanced paclitaxel-induced growth suppression and apoptosis in the human H460 NSCLC cell lines. Our data revealed that this was accounted for primarily by an augmented cleavage of poly(ADP-ribose) polymerase (PARP) and enhanced activation of caspase-8. Pretreatment with z-VAD-FMK (a pan-caspase inhibitor) or z-IETD-FMK (a caspase-8 inhibitor) blocked TOS/paclitaxel cotreatment-induced PARP cleavage and apoptosis, suggesting that TOS potentiates the paclitaxel-induced apoptosis through enforced caspase 8 activation in H460 cells. Furthermore, the growth suppression effect of TOS/paclitaxel combination on human H460, A549 and H358 NSCLC cell lines were synergistic. Our observations indicate that combination of paclitaxel and TOS may offer a novel therapeutic strategy for improving paclitaxel drug efficacy in NSCLC patient therapy as well as for potentially lowering the toxic side effects of paclitaxel through reduced drug dosage.


Subject(s)
Humans , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Caspase 8/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , Drug Synergism , Drug Therapy, Combination , Neoplastic Stem Cells , Paclitaxel/pharmacology , alpha-Tocopherol/pharmacology
19.
Experimental & Molecular Medicine ; : 109-117, 2008.
Article in English | WPRIM | ID: wpr-77108

ABSTRACT

The mutation and reduction of mitochondrial DNA (mtDNA) have been suggested as factors in the carcinogenesis. However, whether the depletion of mtDNA induces multidrug resistance in cancer cells has not been fully investigated. To elucidate the association of cellular mtDNA content and drug resistance, we generated HCT-8 colon cancer cells which revealed a marked decrease in cellular mtDNA and ATP content, concomitant with a lack of mRNAs encoded by mtDNA. The mtDNA-depleted cells showed a decreased sensitivity and accumulation of anti-cancer drugs, suggesting that mtDNA depletion could develop multidrug resistance (MDR) phenotype in HCT-8 cells. We found that the expression level of MDR1 mRNA and its translated product P-glycoprotein was increased in the mtDNA- depleted cells, indicating that the decrease of sensitivity and accumulation of anti-cancer drug in the mtDNA-depleted cells might be due to a substantial increase in the expression of P-glycoprotein. Furthermore, increased expression of MDR1 mRNA and P-glycoprotein was due to an increase of mRNA stability rather than transcriptional activation. Taken together, these results indicate that mtDNA depletion can induce an increased P-glycoprotein expression via an increase of mRNA stability and suggest that the mtDNA depletion in cancer cells plays an important role in the induction of MDR phenotype.


Subject(s)
Humans , Cell Line, Tumor , DNA, Mitochondrial/metabolism , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Paclitaxel/pharmacology , Promoter Regions, Genetic/genetics , RNA Stability/drug effects , RNA, Messenger/genetics , Up-Regulation/drug effects
20.
Rev. bras. cardiol. invasiva ; 14(4): 358-363, out.-dez. 2006. ilus, graf, tab
Article in English | LILACS | ID: lil-441115

ABSTRACT

Introdução: Os stents farmacológicos liberadores de sirolimus (SES) e de paclitaxel (PES) reduzem de maneira significativa os índices de reestenose se comparados aos stents de metal. Há muita controvérsia com relação aos possíveis efeitos indesejáveis do SES e do PES, como, por exemplo, a trombosesubaguda intra-stent. Este estudo teve como objetivo comparar o efeito arterial local do SES e do PES. Métodos: A dilatação coronariana foi induzida em 16 suínos pela insuflação de um balão superdimensionado na proporção 1.2:1.0 em 43 artérias. Foram olocados aleatoriamente 21 SES e 22 PES na descendente anterior esquerda e na circunflexa esquerda no local da lesão anterior por balão. Os animais foramenviados para necropsia 30 dias após o procedimento. Vinte artérias foram enviadas para análise pelo métodoWestern Blot (16 segmentos com stent + 4 controles normais). Vinte e sete segmentos com stent foram submetidos à análise histológica e à microscopia eletrônica (ME) a baixo vácuo. Resultados: Não foram observadas diferenças com referência às características morfométricas entre os dois grupos. A espessura neointimal se mostrou semelhante nos segmentos com stent SES e PES (0,23±0,05mm vs 0,21±0,08mm, respectivamente – p=0,08). A cicatrização arterial localavaliada pelo método WB demonstrou níveis significantemente mais altos do fator Von Willebrand e CD 31 em SESvs PES (p=0,005 e 0,03, respectivamente). PES demonstrou,ainda, inflamação local mais intensa, expressa pelos níveis mais altos de PDGF (p=0,0007). Este resultado foi corroborado pelos achados de reação inflamatória local mais intensa pela ME, expressa por dados inflamatórios mais elevados no grupo PES. Conclusão: PES demonstrou maior grau de inflamação e menor expressão de CD31 e do fator VonWillebrand, sugerindo, portanto, cicatrização endotelial comprometidaapós a colocação do stent se comparado ao SES.


Background: Sirolimus eluting stents (SES) and Paclitaxel eluting stents (PES) significantly reduce restenosis rates as compared with bare metal stents. There is much controversyregarding possible untoward effects of SES and PES such as subacute stent thrombosis. The present study aimed tocompare the local arterial effect of SES versus PES. Methods: In 16 pigs coronary overstretch was induced by inflating an oversized angioplasty balloon at 1.2:1.0 ratio in 43 arteries. Twenty one SES and 22 PES were randomly deployed in LAD and LCx in the site of previous balloon injury. Animals were sent to necropsy 30 days after the procedure. Twenty arteries were sent to Western Blot (WB) analysis (16 stented segments plus 4 normal controls). Twenty seven stented segments were submitted to histologyanalysis and low vacuum electron microscopy (EM). Results: There were no differences regarding arterial morphometric characteristics between the two groups. Neo intimal thicknesswere similar in SES and PES stented segments (0.23±0.05mm vs 0.21±0.08mm, respectively – p=0.08). Local arterial healing assessed by WB showed significantly higher local levels of Von Willebrand factor and CD 31 in SES vs PES (p values of 0.005 and 0.03, respectively). Also, PES showed higher local inflammation as expressed byhigher PDGF levels (p= 0.0007). This result was corroborated by the EM findings of higher local inflammatory reaction, expressed by higher inflammation scores in the PES group.Conclusion: PES showed higher inflammation and lower expression of CD31 and Von Willebrand factor, suggesting an impaired endothelial healing after stenting when comparedwith SES.


Subject(s)
Animals , Stents , Paclitaxel/pharmacology , Coronary Restenosis/pathology , Sirolimus/pharmacology , Coronary Vessels , Models, Animal , Case-Control Studies , Blood Vessel Prosthesis Implantation/methods , Follow-Up Studies , Swine , Coronary Vessels/pathology , Blotting, Western
SELECTION OF CITATIONS
SEARCH DETAIL