Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Intervalo de año de publicación
1.
Zhongguo Zhong Yao Za Zhi ; 43(1): 160-167, 2018 Jan.
Artículo en Chino | MEDLINE | ID: mdl-29552827

RESUMEN

It was aimed at exploring the potential pharmacological effects of alkaloids in Sophora alopecuroides by means of network pharmacology in this study. The main alkaloids in S. alopecuroides were collected for analysis of drug properties, prediction of potential targets and screening of signaling pathways. DAVID analysis tool combined with KEGG database was used to annotate and analyze the signaling pathway. The alkaloids-targets-signaling pathways network was built through Cytoscape software. Results showed that 17 alkaloids in S. alopecuroides involved 49 targets (170 times in all) and 22 important signaling pathways. Three nodes in model of network pharmacology were cross-linked, and the metabolic pathways were coordinated and regulated by each other. It indicated that alkaloids in S. alopecuroides may have therapeutic effect on diseases of cancer, metabolic disorder, endocrine system, digestive system, nervous system and so on.


Asunto(s)
Alcaloides/farmacología , Transducción de Señal/efectos de los fármacos , Sophora/química , Fitoquímicos/farmacología
2.
Oncotarget ; 8(9): 14748-14758, 2017 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-28107195

RESUMEN

Previous studies have shown that intensive macropinocytosis occurs in cancer cells and neutral red (NR) is noted for its capability to enter into the cell massively through a process mimetic to macropinocytosis. In addition, trans-cinnamic acid (tCA) has been found to be an inhibitor of histone deacetylase (HDAC). In the present study, cinnamoylphenazine (CA-PZ) that consists of NR and tCA moieties was synthesized and evaluated. As shown, CA-PZ massively entered into colon carcinoma HT-29 cells and pancreatic carcinoma MIA PaCa-2 cells and this entry was blocked by 5-(N-ethyl-N-isopropyl) amiloride (EIPA, an inhibitor of macropinocytosis), indicating a macropinocytosis-mediated uptake. Furthermore, CA-PZ markedly increased the protein expression levels of acetyl-H3, acetyl-H4 and p21 in HT-29 cells and MIA PaCa-2 cells. CA-PZ significantly inhibited the growth of colon carcinoma HT-29 and pancreatic carcinoma MIA PaCa-2 xenografts. By in vivo imaging, CA-PZ displayed prominent accumulation in the tumor xenografts. The study indicates that the newly synthesized CA-PZ acts as an HDAC inhibitor in association with intensive macropinocytosis-mediated intracellular delivery in cancer cells. The use of neutral red for preparation of chimeric molecules with the attribute of macropinocytosis-mediated intracellular delivery might open an alternative way for development of HDAC inhibitors.


Asunto(s)
Cinamatos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Fenazinas/farmacología , Pinocitosis/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Células A549 , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Western Blotting , Línea Celular Tumoral , Cinamatos/síntesis química , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Bloqueadores del Canal de Sodio Epitelial/farmacología , Femenino , Células HCT116 , Células HT29 , Inhibidores de Histona Desacetilasas/síntesis química , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Confocal , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fenazinas/síntesis química
3.
Sci Rep ; 6: 31472, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27510727

RESUMEN

Multidrug resistance is a major limitation for microtubule-binding agents in cancer treatment. Here we report a novel microtubule inhibitor (2-morpholin-4-yl-5-nitro-benzoic acid 4-methylsulfanyl-benzyl ester, IMB5046), its cytotoxicity against multidrug-resistant cell lines and its antitumor efficacy in animal models. IMB5046 disrupted microtubule structures in cells and inhibited purified tubulin polymerization in vitro. It bound to the colchicine pocket of tubulin. IMB5046 displayed potent cytotoxicity against multiple tumor cell lines with an IC50 range of 0.037-0.426 µM. Notably, several multidrug-resistant cell lines which were resistant to colchicine, vincristine and paclitaxel remained sensitive to IMB5046. IMB5046 was not a P-glycoprotein substrate. IMB5046 blocked cell cycle at G2/M phase and induced cell apoptosis. Microarray assay indicated that the differentially expressed genes after IMB5046 treatment were highly related to immune system, cell death and cancer. In a mouse xenograft model IMB5046 inhibited the growth of human lung tumor xenograft by 83% at a well-tolerated dose. It is concluded that IMB5046 is a tubulin polymerization inhibitor with novel chemical structure and can overcome multidrug resistance. It is a promising lead compound for cancer chemotherapy, especially for treatment of multidrug-resistant tumors.


Asunto(s)
Benzoatos/administración & dosificación , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Morfolinas/administración & dosificación , Neoplasias/tratamiento farmacológico , Nitrobenzoatos/administración & dosificación , Moduladores de Tubulina/administración & dosificación , Células A549 , Animales , Benzoatos/química , Benzoatos/farmacología , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Células HT29 , Humanos , Ratones , Morfolinas/química , Morfolinas/farmacología , Células 3T3 NIH , Neoplasias/genética , Nitrobenzoatos/química , Nitrobenzoatos/farmacología , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Oncotarget ; 7(36): 58418-58434, 2016 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-27517152

RESUMEN

K-Ras mutant pancreatic cancer cells display intensive macropinocytosis, indicating that this process may be exploited in the design of anticancer targeted therapies. In this study, we constructed a macropinocytosis-oriented recombinantly tailored defensin (DF-HSA) which consists of human ß-defensin-2 (DF) and human serum albumin (HSA). The macropinocytosis intensity and cytotoxicity of DF-HSA were investigated in K-Ras mutant MIA PaCa-2 cells and wild-type BxPC-3 cells. As found, the DF-HSA uptake in MIA PaCa-2 cells was much higher than that in wild-type BxPC-3 cells. Correspondingly, the cytotoxicity of DF-HSA to MIA PaCa-2 cells was more potent than that to BxPC-3 cells. In addition, the cytotoxicity of DF-HSA was much stronger than that of ß-defensin HBD2. DF-HSA suppressed cancer cell proliferation and induced mitochondrial pathway apoptosis. Notably, DF-HSA significantly inhibited the growth of human pancreatic carcinoma MIA PaCa-2 xenograft in athymic mice at well tolerated dose. By in vivo imaging, DF-HSA displayed a prominent accumulation in the tumor. The study indicates that the recombinantly tailored ß-defensin can intensively enter into the K-Ras mutant pancreatic cancer cells through macropinocytosis-mediated process and exert potent therapeutic efficacy against the pancreatic carcinoma xenograft. The novel format of ß-defensin may play an active role in macropinocytosis-mediated targeting therapy.


Asunto(s)
Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Pinocitosis , Proteínas Proto-Oncogénicas p21(ras)/genética , Albúmina Sérica Humana/metabolismo , beta-Defensinas/metabolismo , Células A549 , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Femenino , Genes ras , Vectores Genéticos , Humanos , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Trasplante de Neoplasias , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo
5.
J Cancer Res Ther ; 12(1): 182-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27072234

RESUMEN

AIMS: The aim of this study is to explore the antitumor efficacy of lidamycin (LDM) against human multiple myelomas (MM). MATERIALS AND METHODS: Human MM RPMI 8226 cells and the xenograft model in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice were used to examine the antitumor activity of LDM. RESULTS: Notably, LDM markedly suppressed the growth of human MM RPMI 8226 xenograft in NOD/SCID mice. In vitro, there was a significant reduction in cell proliferation after treatment with LDM. The overall growth inhibition correlated with the increase of apoptotic cells. The apoptosis-related proteins including caspase-3, 7, and 9 were activated, and poly adenosine diphosphate-ribose polymerase was cleaved. Further investigation revealed that cellular Bcl-2 and survivin decreased, whereas the level of Bax increased in the LDM-treated cells. CONCLUSIONS: LDM is highly effective against the growth of MM xenograft in NOD/SCID mice. The potent apoptosis.inducing effect of LDM may be mediated through caspase. and mitochondria.dependent pathway.


Asunto(s)
Aminoglicósidos/administración & dosificación , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Enediinos/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Animales , Caspasas/biosíntesis , Caspasas/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos NOD , Mieloma Múltiple/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Med Rep ; 13(5): 4159-66, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27035417

RESUMEN

Previous studies have shown that trans-cinnamic acid (tCA) has a broad spectrum of biological activities, and exhibits antioxidant, anti-inflammatory and anticancer properties. In addition, tCA and a variety of its analogs have been detected as gut microbe­derived metabolites exerting various biological effects in the colon. The aim of this study was to assess the antitumor activity of tCA in vitro and in vivo, in particular its therapeutic efficacy against colon cancer xenografts in athymic mice. Furthermore, it aimed to examine the effects of tCA on histone deacetylases (HDACs) and to identify the underlying molecular mechanisms. Using an MTT assay, tCA was observed to inhibit the proliferation of several cancer cell lines, and the half maximal inhibitory concentration (IC50) in HT29 colon carcinoma cells was ~1 mM. Western blot analysis demonstrated that tCA upregulated the expression of acetyl­H3 and acetyl­H4 proteins, which was consistent with the effects of the HDAC inhibitor, trichostatin A (TSA). Furthermore, expression of Bcl­2 (a marker of cell proliferation) was reduced, and apoptosis was induced. Apoptosis was shown by the activation of cleavage of poly ADP ribose polymerase and the increased expression of Bax. Apoptosis was also confirmed using APC Annexin V and SYTOX Green Nucleic Acid Stain. In addition, the tCA­induced inhibition of the expression of HDAC markers and activation of apoptosis in tumor tissues were further confirmed by immunohistochemistry. Intragastric administration of tCA at doses of 1.0 and 1.5 mmol/kg body weight suppressed the growth of HT29 human colon carcinoma xenografts in athymic mice at well­tolerated doses. No toxic changes were found in the heart, lung, liver, kidney, colon or bone marrow following histopathological examination. This study indicated that tCA is effective against colon cancer xenograft in nude mice. The antitumor mechanism of tCA was mediated, at least in part, by inhibition of HDACs in cancer cells. As an endogenous microbial metabolite predominantly produced in the colon, tCA is an agent of interest for further evaluation.


Asunto(s)
Antineoplásicos/farmacología , Cinamatos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Anticancer Drugs ; 27(1): 24-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26457548

RESUMEN

Neocarzinostatin (NCS) consists of an enediyne chromophore and an apoprotein (NCP). Lidamycin (LDM) is composed of another active enediyne chromophore (AE) and an acidic protein (LDP). Although the structures of NCP and LDP are very similar, LDM has been shown to have an increased tumor-suppressive activity than that of NCS. The aim of this study was to construct a chimeric protein (CMP) that consists of both the terminus residue of NCP and an LDP pocket-forming residue that can bind AE. This CMP will have a structure similar to NCS and an antitumor activity similar to LDM. The assembling efficiency of LDP, CMP, and NCP was 73.9, 1.5, and 1.1%, respectively. The cytotoxicity was consistent with their assembling efficiency of AE in proteins. When CMP-AE and NCP-AE were administered at equivalent AE doses of LDM, the inhibition rate of CMP-AE was the same as LDM and significantly higher than that of NCP-AE. Our study implied that the binding activity between LDP and AE was very specific. The terminus residue of LDP could affect the specifically binding activity. The pocket-forming residue could confer a protective function to the chromophore. Further investigation of its bioactivity might serve as a new drug design strategy and drug-delivery carrier in targeted cancer therapy.


Asunto(s)
Aminoglicósidos/química , Antineoplásicos/química , Apoproteínas/química , Enediinos/química , Proteínas Recombinantes de Fusión/química , Cinostatina/química , Aminoglicósidos/genética , Animales , Antineoplásicos/farmacología , Apoproteínas/genética , Apoproteínas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología
8.
Acta Pharm Sin B ; 5(3): 264-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26579455

RESUMEN

In this study, N-terminal site-specific mono-PEGylation of the recombinant lidamycin apoprotein (rLDP) of lidamycin (LDM) was prepared using a polyethyleneglycol (PEG) derivative (M w 20 kDa) through a reactive terminal aldehyde group under weak acidic conditions (pH 5.5). The biochemical properties of mPEG-rLDP-AE, an enediyne-integrated conjugate, were analyzed by SDS-PAGE, RP-HPLC, SEC-HPLC and MALDI-TOF. Meanwhile, in vitro and in vivo antitumor activity of mPEG-rLDP-AE was evaluated by MTT assays and in xenograft model. The results indicated that mPEG-rLDP-AE showed significant antitumor activity both in vitro and in vivo. After PEGylation, mPEG-rLDP still retained the binding capability to the enediyne AE and presented the physicochemical characteristics similar to that of native LDP. It is of interest that the PEGylation did not diminish the antitumor efficacy of LDM, implying the possibility that this derivative may function as a payload to deliver novel tumor-targeted drugs.

9.
Basic Clin Pharmacol Toxicol ; 117(2): 105-16, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25615234

RESUMEN

Gelatinases play important roles in tumour invasion and metastasis and are thus considered promising targets for cancer therapy. In this study, a new single-chain variable fragment (scFv)-based fusion protein Fv-LDP, composed of the anti-gelatinases scFv and lidamycin apoprotein (LDP), was prepared, and its combination with angiogenesis inhibitor Endostar was then investigated. The fusion protein Fv-LDP specifically bound to various tumour cells, and its binding capability to human pulmonary giant cell carcinoma (PG) cells was higher than that of LDP. Fv-LDP inhibited the expression and secretion of gelatinases and could be internalized into tumour cells via endocytosis. Fv-LDP also suppressed the growth of human hepatoma cells and murine hepatoma 22 transplanted in Kunming mice in various degrees. In addition, Endostar could enhance the synergistic or additive inhibition of Fv-LDP on the growth, migration or invasion of human hepatoma cells shown by a colony formation assay and a transwell-based migration or invasion assay, respectively. In vivo, Fv-LDP/Endostar combination showed a significantly synergistic effect on the growth of a human hepatoma xenograft, with an inhibition rate of 80.8% compared with the Fv-LDP (44.1%) or Endostar (8.9%)-treated group. The above-mentioned results indicate that the fusion protein Fv-LDP is effective against transplantable hepatoma in mice and human hepatoma xenografts in athymic mice. Moreover, Endostar can potentiate the inhibition effect of Fv-LDP on the growth of human hepatoma cells and xenografts. These data will provide a new combined strategy for improving the therapeutic efficacy of treatments for hepatoma or other gelatinase-overexpressing tumours.


Asunto(s)
Aminoglicósidos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Endostatinas/farmacología , Enediinos/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Anticuerpos de Cadena Única/farmacología , Aminoglicósidos/administración & dosificación , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacología , Animales , Apoproteínas/administración & dosificación , Apoproteínas/farmacología , Carcinoma de Células Gigantes/tratamiento farmacológico , Carcinoma de Células Gigantes/enzimología , Carcinoma de Células Gigantes/patología , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Sinergismo Farmacológico , Endostatinas/administración & dosificación , Enediinos/administración & dosificación , Femenino , Gelatinasas/metabolismo , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Recombinantes , Anticuerpos de Cadena Única/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Carcinog ; 54(1): 1-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23857500

RESUMEN

Recently, tumor initiating cells are considered as the central role of tumorigenicity in hepatocellular carcinoma. Enediyne anticancer antibiotic lidamycin with great potential antitumor activity is currently evaluated in Phase II clinical trials. In this study, we evaluated the effect of lidamycin on tumor initiating cells of hepatocellular carcinoma Huh7 and identified the potential mechanism. Flow cytometry analysis and sorting assay, surface marker assay, sphere formation assay, and aldefluor assay were used to evaluate the effect of lidamycin on Huh7 tumor initiating cells in vitro. To investigate the potential mechanism, the activity of GSK3ß/ß-catenin pathway was detected by Western blot and T cell factors transcriptional activity assay. Subcutaneous tumor model in nude mice was used to observe in vivo effect of lidamycin on Huh7 cells. Lidamycin decreased the proportion of EpCAM+ cells and the expression of EpCAM protein. Lidamycin inhibited sphere formation of sorted EpCAM+ cells in 7 d, and of parental cells in three serial passages. The population of aldehyde dehydrogenase-positive cells was reduced by lidamycin. In addition, lidamycin restrained tumor volume and incidence in vivo. Lidamycin activated GSK3ß, and degraded the activity of ß-catenin. Consequently, transcriptional activity of ß-catenin/T cell factors was decreased. In brief, these results suggest that lidamycin suppressed Huh7 tumor initiating cells via GSK3ß/ß-catenin pathway. These findings reveal the potential mechanism of lidamycin on tumor initiating cells and the benefit for further clinical evaluation.


Asunto(s)
Aminoglicósidos/farmacología , Antibióticos Antineoplásicos/farmacología , Carcinoma Hepatocelular/patología , Enediinos/farmacología , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas Experimentales , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo
11.
Asian Pac J Cancer Prev ; 15(17): 7043-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25227788

RESUMEN

Inhibition of heat shock protein 90 (Hsp90) leads to inappropriate processing of proteins involved in DNA damage repair pathways after DNA damage and may enhance tumor cell radio- and chemo-therapy sensitivity. To investigate the potentiation of antitumor efficacy of lidamycin (LDM), an enediyne agent by the Hsp90 inhibitor geldanamycin (GDM), and possible mechanisms, we have determined effects on ovarian cancer SKOV- 3, hepatoma Bel-7402 and HepG2 cells by MTT assay, apoptosis assay, and cell cycle analysis. DNA damage was investigated with H2AX C-terminal phosphorylation (γH2AX) assays. We found that GDM synergistically sensitized SKOV-3 and Bel-7402 cells to the enediyne LDM, and this was accompanied by increased apoptosis. GDM pretreatment resulted in a greater LDM-induced DNA damage and reduced DNA repair as compared with LDM alone. However, in HepG2 cells GDM did not show significant sensitizing effects both in MTT assay and in DNA damage repair. Abrogation of LDM-induced G2/M arrest by GDM was found in SKOV-3 but not in HepG2 cells. Furthermore, the expression of ATM, related to DNA damage repair responses, was also decreased by GDM in SKOV-3 and Bel-7402 cells but not in HepG2 cells. These results demonstrate that Hsp90 inhibitors may potentiate the antitumor efficacy of LDM, possibly by reducing the repair of LDM-induced DNA damage.


Asunto(s)
Aminoglicósidos/farmacología , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Benzoquinonas/farmacología , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Enediinos/farmacología , Lactamas Macrocíclicas/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Células Hep G2 , Histonas/efectos de los fármacos , Histonas/metabolismo , Humanos , Fosforilación
12.
BMC Cancer ; 13: 479, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24128285

RESUMEN

BACKGROUND: Endostatin (ES) inhibits endothelial cell proliferation, migration, invasion, and tube formation. It also shows antiangiogenesis and antitumor activities in several animal models. Endostatin specifically targets tumor vasculature to block tumor growth. Lidamycin (LDM), which consists of an active enediyne chromophore (AE) and a non-covalently bound apo-protein (LDP), is a member of chromoprotein family of antitumor antibiotics with extremely potent cytotoxicity to cancer cells. Therefore, we reasoned that endostatin-lidamycin (ES-LDM) fusion proteins upon energizing with enediyne chromophore may obtain the combined capability targeting tumor vasculature and tumor cell by respective ES and LDM moiety. METHODS: In this study, we designed and obtained two new endostatin-based fusion proteins, endostatin-LDP (ES-LDP) and LDP-endostatin (LDP-ES). In vitro, the antiangiogenic effect of fusion proteins was determined by the wound healing assay and tube formation assay and the cytotoxicity of their enediyne-energized analogs was evaluated by CCK-8 assay. Tissue microarray was used to analyze the binding affinity of LDP, ES or ES-LDP with specimens of human lung tissue and lung tumor. The in vivo efficacy of the fusion proteins was evaluated with human lung carcinoma PG-BE1 xenograft and the experimental metastasis model of 4T1-luc breast cancer. RESULTS: ES-LDP and LDP-ES disrupted the formation of endothelial tube structures and inhibited endothelial cell migration. Evidently, ES-LDP accumulated in the tumor and suppressed tumor growth and metastasis. ES-LDP and ES show higher binding capability than LDP to lung carcinoma; in addition, ES-LDP and ES share similar binding capability. Furthermore, the enediyne-energized fusion protein ES-LDP-AE demonstrated significant efficacy against lung carcinoma xenograft in athymic mice. CONCLUSIONS: The ES-based fusion protein therapy provides some fundamental information for further drug development. Targeting both tumor vasculature and tumor cells by endostatin-based fusion proteins and their enediyne-energized analogs probably provides a promising modality in cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Endostatinas/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Endostatinas/farmacocinética , Endostatinas/uso terapéutico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Femenino , Humanos , Pulmón/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/uso terapéutico , Análisis de Matrices Tisulares , Distribución Tisular , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Anticancer Drugs ; 24(6): 609-16, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23698252

RESUMEN

The proteasome inhibitor bortezomib has been applied successfully to treat multiple myeloma (MM). Its synergistic effects with other anticancer drugs have been studied widely. In the present study, it was found that lidamycin (LDM), a member of the enediyne antibiotic family, showed much more potent cytotoxicity than bortezomib to MM cell lines: U266 and SKO-007. Here, we investigated the potential synergy of bortezomib and LDM on MM cells. The results showed that cotreatment of bortezomib and LDM synergistically induced cytotoxicity and apoptosis in MM cell lines, followed by enhanced caspase-3 cleavage and degradation of poly-ADP-ribose polymerase together with the decreased nuclear factor-κB protein. These two drugs synergistically induced apoptosis, which was associated with enhanced activation of two mitogen-activated protein kinases: p38 mitogen-activated protein kinase and c-Jun NH(2)-terminal kinase. Moreover, bortezomib plus LDM synergistically induced apoptosis was also associated with downregulation of extracellular signal-regulated kinase, and induction of endoplasmic reticulum stress response. Overall, our results indicate that the combined regimen of bortezomib and LDM might be a potential therapeutic remedy for the treatment of MM.


Asunto(s)
Aminoglicósidos/farmacología , Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Enediinos/farmacología , Mieloma Múltiple/tratamiento farmacológico , Pirazinas/farmacología , Aminoglicósidos/administración & dosificación , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Ácidos Borónicos/administración & dosificación , Bortezomib , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Enediinos/administración & dosificación , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Pirazinas/administración & dosificación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Biomed Pharmacother ; 67(2): 164-71, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23206754

RESUMEN

Targeting and inhibiting angiogenesis is a promising strategy for treatment of cancer. NGR peptide motif is a tumor-homing peptide, which could bind with CD13 expressed on tumor blood vessels. Lidamycin is a highly potent antitumor antibiotic, which is composed of an apoprotein (LDP) and an active enediyne chromophore (AE). Here, an NGR-integrated and enediyne-energized apoprotein composed of cyclic NGR peptide and lidamycin was developed by a two-step procedure. Firstly, we prepared the fusion protein composed of NGR peptide and LDP by recombinant DNA technology. Then, AE was reloaded to the fusion protein to get NGR-LDP-AE. Our experiments showed that NGR-LDP could bind to CD13-expressing HT-1080 cells, whereas the recombinant LDP (rLDP) showed weak binding. NGR-LDP-AE exerted highly potent cytotoxicity to cultured tumor cells in vitro. In vivo antitumor activity was evaluated in murine hepatoma 22 (H22) model and human fibrosarcoma HT-1080 model. At the tolerable dose, NGR-LDP-AE and lidamycin inhibited H22 tumor growth by 94.8 and 66.9%, and the median survival time of the mice was 62 and 37 days, respectively. In the HT-1080 model, NGR-LDP-AE inhibited tumor growth by 88.6%, which was statistically different from that of lidamycin (74.5%). Immunohistochemical study showed that NGR-LDP could bind to tumor blood vessels. Conclusively, these results demonstrate that fusion of LDP with CNGRC peptide delivers AE to tumor blood vessels and improves its antitumor activity.


Asunto(s)
Apoproteínas/farmacología , Antígenos CD13/metabolismo , Enediinos/farmacología , Oligopéptidos/farmacología , Aminoglicósidos/farmacología , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Antibióticos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Apoproteínas/genética , Apoproteínas/metabolismo , Línea Celular Tumoral , Femenino , Fibrosarcoma/tratamiento farmacológico , Células Hep G2 , Humanos , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oligopéptidos/genética , Oligopéptidos/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología
15.
Eur J Pharm Sci ; 47(4): 781-9, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-22982402

RESUMEN

Arginine-rich peptides belong to a subclass of cell penetrating peptides that are taken up by living cells and can be detected freely diffusing inside the cytoplasm and nucleoplasm. This phenomenon has been attributed to either an endocytotic mode of uptake and a subsequent release from vesicles or a direct membrane penetration. Lidamycin is an antitumor antibiotic, which consists of an active enediyne chromophore (AE) and a noncovalently bound apoprotein (LDP). In the present study, a fusion protein (Arg)(9)-LDP composed of cell penetrating peptide (Arg)(9) and LDP was prepared by DNA recombination, and the enediyne-energized fusion protein (Arg)(9)-LDP-AE was prepared by molecular reconstitution. The data in fixed cells demonstrated that (Arg)(9)-LDP could rapidly enter cells, and the results based on fluorescence activated cell sorting indicated that the major route for (Arg)(9)-mediated cellular uptake of protein molecules was endocytosis. (Arg)(9)-LDP-AE demonstrated more potent cytotoxicity against different carcinoma cell lines than lidamycin in vitro. In the mouse hepatoma 22 model, (Arg)(9)-LDP-AE (0.3mg/kg) suppressed the tumor growth by 89.2%, whereas lidamycin (0.05 mg/kg) by 74.6%. Furthermore, in the glioma U87 xenograft model in nude mice, (Arg)(9)-LDP-AE at 0.2mg/kg suppressed tumor growth by 88.8%, compared with that of lidamycin by 62.9% at 0.05 mg/kg. No obvious toxic effects were observed in all groups during treatments. The results showed that energized fusion protein (Arg)(9)-LDP-AE was more effective than lidamycin and would be a promising candidate for glioma therapy. In addition, this approach to manufacturing fusion proteins might serve as a technology platform for the development of new cell penetrating peptides-based drugs.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Apoproteínas/farmacología , Péptidos de Penetración Celular/farmacología , Enediinos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Aminoglicósidos/farmacología , Animales , Antibióticos Antineoplásicos/metabolismo , Apoproteínas/genética , Apoproteínas/metabolismo , Arginina/genética , Arginina/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Péptidos de Penetración Celular/genética , Péptidos de Penetración Celular/metabolismo , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Enediinos/metabolismo , Femenino , Células Hep G2 , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
16.
Cancer Lett ; 301(2): 212-20, 2011 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-21216091

RESUMEN

We evaluated the efficacy of a combination strategy, Endostar, a modified recombinant human endostatin, plus dexamethasone, against angiogenesis and hepatoma growth. By colony formation assay, synergistic effects of the combination of Endostar and dexamethasone were observed on the proliferations of human umbilical endothelial cells and hepatoma Bel-7402 cells. Endostar plus dexamethasone inhibited angiogenesis events in vitro. Examined with transwell assay, HUVECs invasion was more efficiently suppressed by combination of Endostar and dexamethasone than by the respective single drug. But the combination treatment of Endostar and dexamethasone to Bel-7402 cells did not alter the migration of HUVECs. The migration of HUVEC tended to coincident with VEGF secretion as determined by ELISA. Tube formation assay, rat aortic ring assay and chick embryo chorioallantoic membrane (CAM) assay indicated that the anti-angiogenesis effects of Endostar were significantly enhanced by dexamethasone. In mouse hepatoma H22 and human hepatoma Bel-7402 subcutaneous xenograft, Endostar plus dexamethasone presented more potent efficacy in tumor growth suppression than the single drug treatments. The above confirms the synergism of Endostar and dexamethasone on anti-angiogenesis and suppression of hepatoma growth. The potentiation effects of the combination indicate that Endostar plus dexamethasone might be a positive strategy for cancer therapy.


Asunto(s)
Dexametasona/farmacología , Endostatinas/farmacología , Neoplasias Hepáticas Experimentales/prevención & control , Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de la Angiogénesis/farmacología , Animales , Antiinflamatorios/farmacología , Aorta/efectos de los fármacos , Aorta/fisiología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Sinergismo Farmacológico , Endostatinas/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Humanos , Técnicas In Vitro , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Desnudos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Anticancer Drugs ; 22(2): 166-75, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20948431

RESUMEN

Boningmycin, a new antibiotic of the bleomycin family, is isolated from the fermentation broth of Streptomyces verticillus var. pingyangensis n.sp. This study aimed to evaluate its antitumor actions and mechanism. The results showed that boningmycin exhibited potent inhibitory effects on several human solid tumor cells and that it was stronger than bleomycin. The administration of boningmycin inhibited the growth of human hepatoma HepG2 xenografts in nude mice, with more efficacy than that of bleomycin. Boningmycin led to an increase of the reactive oxygen species involving iron and caused G2/M phase accumulation in the HepG2 and human breast cancer MCF-7 cells. Two types of cell death, apoptosis and senescence, were detected after exposure to boningmycin. The accumulation of sub-G1 phase cells, an index of apoptosis, and the activation of caspase apoptotic pathways were detected after treatment with higher concentrations of boningmycin. Low concentrations of boningmycin led to a senescent phenotype with an increase in senescence-associated ß-galactosidase activity and the time-dependent increase of p21, p27, and p53 expressions from 48 to120 h. Taken together, the results showed that boningmycin exhibits potent antitumor actions through the induction of apoptosis and cellular senescence.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Bleomicina/análogos & derivados , Senescencia Celular/efectos de los fármacos , Animales , Bleomicina/farmacología , Caspasas/metabolismo , Caspasas/farmacología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Fase G2/efectos de los fármacos , Células HCT116 , Células Hep G2 , Humanos , Hierro/metabolismo , Células KB , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Especies Reactivas de Oxígeno/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Yao Xue Xue Bao ; 46(11): 1321-5, 2011 Nov.
Artículo en Chino | MEDLINE | ID: mdl-22260022

RESUMEN

This study is to investigate inhibitory effects of lidamycin (LDM) on the proliferation of HERG K+ channel highly expressing cancer cells and its synergy with anticancer drugs. MTT assay was used to examine the inhibitory effects of lidamycin combined with various anticancer drugs on the proliferation of human lung cancer A549 cells, human colon cancer HT-29 cells and herg-stably-transfected A549 cells. Using the xenograft model of subcutaneously transplanted HT-29 in nude mice, inhibitory effect was appraised in vivo. The coefficient of drug interaction (CDI) was used to evaluate the synergistic effect of drug combination. LDM significantly inhibited the proliferation ofA549 cells and HT-29 cells with IC50 values of 2.14 and 4.64 ng mL(-1), respectively. The efficacy in HT-29 cells with high HERG potassium expression level is less potent than that in A549 cells with low expression level. In terms of IC50 values, LDM suppressed the growth of herg-stably-transfected A549 cells less potently than pCDNA3.1-stably-transfected A549 cells. There existed synergistic effects in the combinations of fluorouracil (5-FU) and LDM, doxorubicin (DOX) and LDM, or hydroxycamptothecine (HCPT) and LDM. CDI values of the combinations of 5-FU and LDM were more than 0.75. CDI values of LDM and DOX were more than 0.70, but some CDI values of LDM and HCPT were less than 0.70. As for the CDI values, synergistic effects of the combination of LDM and HCPT were the most potent of the three groups. There is no relationship between the inhibitory effect of the growth of cancer cells by 5-FU and HERG potassium expression level. HERG expression level negatively correlated with inhibitory effect on the proliferation of cancer cells by DOX. HERG expression levels and chemosensitivity were positively correlated for HCPT. In the model of subcutaneously xenograft transplanted HT-29 in vivo, LDM and/or HCPT effectively inhibited the growth of HT-29 in nude mice, and the optimum CDI of the combination of LDM and HCPT was less than 1. HERG expression level negatively correlates the chemosensitivity of cancer cells to LDM. There exist synergistic effects in vitro and in vivo in the combination of LDM and HCPT, which inhibitory effects of the proliferation of cancer cells positively modulated by HERG potassium expression level. HERG K+ channel may become a target of combined therapy for choosing anticancer drugs.


Asunto(s)
Aminoglicósidos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proliferación Celular/efectos de los fármacos , Enediinos/farmacología , Canales de Potasio Éter-A-Go-Go/metabolismo , Neoplasias Pulmonares/patología , Aminoglicósidos/administración & dosificación , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/administración & dosificación , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Sinergismo Farmacológico , Canal de Potasio ERG1 , Enediinos/administración & dosificación , Fluorouracilo/administración & dosificación , Células HT29 , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Oncol Rep ; 23(6): 1747-56, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20428834

RESUMEN

Potassium channels are essential for the regulation of cell proliferation. As reported, HERG protein is overexpressed in a wide range of human tumors, including colon carcinoma. The aim of this study was to investigate the effects of antibacterial agents sparfloxacin (SPFX), a blocker of HERG channel, on HERG K+ channel highly expressing colon cancer cells. Expression of HERG and apoptosis correlative proteins was examined by Western blotting. The MTT assay was used to detect the cytotoxicity of drugs and drug combination in vitro. Gene transfection was used to examine the changes in herg-related chemosensitivity. Cell apoptosis was analyzed by flow cytometry. The migration and invasion capacity of tumor cells by SPFX was determined by gelatin zymography assay and Boyden chamber. The in vivo efficacy of SPFX was assessed in murine colon carcinoma C26 in BALB/c mice and human colon carcinoma HCT116 xenografts in nude mice. High expression of HERG protein was detected in colon cancer C26, HCT116 and HT-29 cells. The cell viability of the colon cancer cells was inhibited by SPFX in a dose-dependent manner. SPFX induced apoptosis and inhibited migration and invasion of colon cancer HCT116 cells. The increase in apoptosis was associated with a decrease in procaspase-3 and Bcl-2 protein expression. Study with herg-transfected HEK293 cells and siRNA-knock down HCT116 cells confirmed that the cell viability inhibition by SPFX was correlated with HERG expression. When combined with 5-fluorouracil, SPFX showed synergistic anti-proliferation activity in HCT116 and HT-29 cells. Furthermore, SPFX inhibited the growth of human colon carcinoma HCT116 xenografts and showed synergistic effect with 5-fluorouracil in vivo. Our finding suggested that SPFX could be a biochemical modulator in treatment of colon cancer with chemotherapeutic drugs.


Asunto(s)
Antituberculosos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Canales de Potasio Éter-A-Go-Go/metabolismo , Fluoroquinolonas/farmacología , Animales , Western Blotting , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Canal de Potasio ERG1 , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Cicatrización de Heridas/efectos de los fármacos
20.
Am J Chin Med ; 37(5): 923-31, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19885952

RESUMEN

In previous studies, rhein, one of the major bioactive constituents in the rhizome of rhubarb, inhibited the proliferation of various human cancer cells. However, because of its water insolubility, the anti-tumor efficacy of rhein was limited in vivo. In this study, we observed the anti-tumor activity of rhein lysinate (the salt of rhein and lysine easily dissolves in water) in vivo and investigated its mechanism. Inhibition of ovarian cancer SKOV-3 cell proliferation was determined by MTT assay and the mechanism of action of rhein lysinate was investigated by Western blot analysis. The therapeutic efficacy of rhein lysinate was evaluated by intragastric and intraperitoneal administrations in H22 hepatocellular carcinoma mice. Rhein lysinate inhibited the proliferation of SKOV-3 cells and the IC50 value was 80 microM. Rhein lysinate inhibited the phosphorylation of MEK and ERK and increased the anti-tumor activity of Taxol in vitro. It inhibited tumor growth by both intragastric and intraperitoneal administrations and improved the therapeutic effect of Taxol in H22 hepatocellular carcinoma mice. In conclusion, rhein lysinate offers an anti-tumor activity in vivo and is hopeful to be a chemotherapeutic drug.


Asunto(s)
Antraquinonas/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Paclitaxel/farmacología , Animales , Antraquinonas/administración & dosificación , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Humanos , Concentración 50 Inhibidora , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Paclitaxel/administración & dosificación , Fitoterapia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Rheum/química , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA