Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Ann Med ; 54(1): 743-753, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35243950

RESUMO

Background: Previous studies have proven that Purinostat Mesylate (PM) is a new HDAC inhibitor and exhibits significant antitumor efficacy. However, the clinical application of PM was greatly limited by its poor solubility in water and low bioavailability.Objective:To increase the solubility of PM through pharmaceutical research, and prepare it into an injection that meets the needs of intravenous use to promote its clinical application.MethodsThe prepared PM/HP-ß-CD inclusion complex was studied by computer simulation, fourier transform infrared spectroscopy (FT-IR), nuclear magnetic resonance (1H-NMR spectroscopy), and scanning electron microscopy (SEM). Then, the antitumor effects of PM/HP-ß-CD inclusion complex were studied by in vitro cytotoxicity assay, apoptosis assay, pharmacokinetic study and in vivo antitumor assay.Results:Phase Solubility Analysis revealed that PM and HP-ß-CD were compatible and the solubility of PM increased almost 220 times, to 2.02 mg/mL. The interaction mechanism studies revealed that PM could be embedded into the cavity of HP-ß-CD through the side of the aminobenzene ring. Cell viability and apoptosis assays showed that PM/HP-ß-CD complex maintained the good anti-cancer activity of PM, and PM/HP-ß-CD complex has a better anti-tumor effect and lower toxicity than LBH589 and Hyper-CVAD/RTX in vivo. All the results suggest that HP-ß-CD can solve the problem of PM administration and provide a way for clinical application of PM.Conclusions: In this study, an injectable formulation of PM in HP-ß-CD (10% w/v) was prepared to improve its water solubility. Our research provides a way for clinical administration of PM, which has been under phase I clinical trial for the treatment of relapsed or refractory B-cell-related hematologic malignancies in China and the USA.KEY MESSAGESWe developed a preparation of Purinostat Mesylate that can be administered intravenously, reducing the toxicity associated with oral administration.This preparation has an outstanding therapeutic effect on SU-DHL-6 xenograft tumour, indicating its clinical value, which has been under phase I clinical trial for the treatment of relapsed or refractory B-cell-related haematologic malignancies in China and the USA.


Assuntos
Mesilatos , 2-Hidroxipropil-beta-Ciclodextrina/química , Varredura Diferencial de Calorimetria , Simulação por Computador , Humanos , Espectroscopia de Infravermelho com Transformada de Fourier
2.
Drug Deliv ; 28(1): 1055-1066, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34078185

RESUMO

Given the maturation of small-interfering RNA (siRNA) techniques with nanotechnology, and because overexpression of human programmed death-ligand 1 (PD-L1) is crucial for T cell inactivation and immunosuppression of the tumor microenvironment, application of siRNA-PD-L1 has demonstrated positive progress in preclinical studies; however, the limited penetration of this compound into solid tumors remains a challenge. To decrease PD-L1 expression and increase the penetration efficacy of solid tumors, we synthesized a novel tumor-microenvironment-sensitive delivery polymer by conjugating hyaluronic acid (HA) to polyethyleneimine (PEI), with a matrix metalloproteinase-2 (MMP-2)-sensitive peptide acting as the linker (HA-P-PEI), for use in delivery of PD-L1-siRNA. Concurrent synthesis of a linker-less HA-PEI compound allowed confirmation that negatively charged siRNA can be complexed onto the positively charged HA-PEI and HA-P-PEI compounds to form nanoparticles with the same particle size and uniform distribution with serum stability. We found that the size of the HA-P-PEI/siRNA nanoparticles decreased to <10 nm upon addition of MMP-2, and that H1975 cells overexpressing CD44, PD-L1, and MMP-2 aided confirmation of the delivery efficacy of the HA-P-PEI/siRNA nanocomplexes. Additionally, the use of HA-P-PEI caused less cytotoxicity than PEI alone, demonstrating its high cellular uptake. Moreover, pretreatment with MMP-2 increased nanocomplex tumor permeability, and western blot showed that HA-P-PEI/PD-L1-siRNA efficiently downregulated the PD-L1 expression in H1975 cells. These results demonstrated a novel approach for siRNA delivery and tumor penetration for future clinical applications in cancer treatment.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Pulmão/efeitos dos fármacos , Metaloproteinase 2 da Matriz/metabolismo , Nanopartículas/química , RNA Interferente Pequeno/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Química Farmacêutica , Relação Dose-Resposta a Droga , Portadores de Fármacos , Estabilidade de Medicamentos , Inativação Gênica , Humanos , Receptores de Hialuronatos/biossíntese , Ácido Hialurônico/química , Micelas , Tamanho da Partícula , Polietilenoimina/química , RNA Interferente Pequeno/administração & dosagem , Propriedades de Superfície , Microambiente Tumoral/efeitos dos fármacos
3.
Sci Adv ; 7(21)2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34138737

RESUMO

Microtubules, composed of αß-tubulin heterodimers, have remained popular anticancer targets for decades. Six known binding sites on tubulin dimers have been identified thus far, with five sites on ß-tubulin and only one site on α-tubulin, hinting that compounds binding to α-tubulin are less well characterized. Cevipabulin, a microtubule-active antitumor clinical candidate, is widely accepted as a microtubule-stabilizing agent by binding to the vinblastine site. Our x-ray crystallography study reveals that, in addition to binding to the vinblastine site, cevipabulin also binds to a new site on α-tubulin. We find that cevipabulin at this site pushes the αT5 loop outward, making the nonexchangeable GTP exchangeable, which reduces the stability of tubulin, leading to its destabilization and degradation. Our results confirm the existence of a new agent binding site on α-tubulin and shed light on the development of tubulin degraders as a new generation of antimicrotubule drugs targeting this novel site.

4.
Curr Pharm Des ; 26(34): 4338-4348, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32324508

RESUMO

BACKGROUND: PEGylation of stealth liposomes elevates their stability and prolongs plasma half-life. Stealth liposomes modified with targeting ligands are expected to be ideal drug delivery carriers. OBJECTIVE: To encapsulate docetaxel in tbFGF (truncated basic fibroblast growth factor)-functionalized liposomes with mPEG2000-VE (d-κ-tocopheryl polyethylene glycol succinate, TPGS2K) and measure their antitumor effects in vitro and in vivo. METHODS: TPGS2K and COOH-PEG2000-VE were synthesized, and tbFGF was conjugated to COOH-PEG2000-VE to prepare tbFGF-PEG2000-VE. Then, tbFGF-functionalized liposomes (DTX-tbFGF-LPs) were prepared by inserting tbFGF-PEG2000-VE into docetaxel liposomes comprising TPGS2K (DTX-PEG-LPs). The stabilities and drug release profiles of the formulation were evaluated. P-glycoprotein (P-gp) inhibition was measured by ATPase assay. MTT and cell uptake were measured with B16 cells. A B16 C57BL/6 mouse model was used to evaluate in vivo antitumor efficacy. RESULTS: Both DTX-PEG-LPs and DTX-tbFGF-LPs exhibited good stability and sustained drug release. MTT, flow cytometry, and fluorescence microscopy of B16 cells revealed higher antitumor activity and more efficient cell uptake for DTX-tbFGF-LPs compared with DTX-PEG-LPs and DTX-LPs. The P-gp ATPase assay showed that both PEG-LP and tbFGF-PEG-LP formulations inhibited P-gp pump activity in vitro. DTX-tbFGF-LPs had the highest antitumor efficacy and lowest toxicity in vivo. CONCLUSION: Truncated basic fibroblast growth factor-functionalized liposomes with TPGS2K as drug delivery nanocarriers were effective chemotherapy agents targeting FGFR-overexpressing tumors.


Assuntos
Antineoplásicos , Lipossomos , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Docetaxel/farmacologia , Fator 2 de Crescimento de Fibroblastos , Camundongos , Camundongos Endogâmicos C57BL , Polietilenoglicóis , Succinatos , Vitamina E
5.
Colloids Surf B Biointerfaces ; 182: 110356, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31319226

RESUMO

A novel triblock amphiphilic copolymer (PAL-PEG-Birinapant) was designed and synthesized as a dual-functional micellar carrier utilizing birinapant (an inhibitor of inhibitor-of-apoptosis proteins) as a pH-sensitive segment and inhibitor-of-apoptosis proteins-targeting ligand. The mixed micelles comprised of PAL-PEG-Birinapant (PPB) and mPEG2k-PDLLA2k (MPP), named as PPB/MPP (2/1,w/w) micelles were developed for enhanced solubility and antitumor potency of hydrophobic drugs as paclitaxel (PTX). In vitro cell viability and cytotoxicity studies revealed that the PTX-loaded PPB/MPP micelles were more potent than the commercial PTX formulation (Taxol®), as well as the in vitro cell apoptosis study. Clear differences in the intracellular uptake of free coumarin-6 (C6) solution and C6-loaded PPB/MPP micelles were observed and indicated that the PPB/MPP micelles could efficiently deliver chemical compound into tumor cells. PPB copolymer and PTX-loaded PPB/MPP micelles demonstrated an excellent safety profile with a maximum tolerated dose (MTD) of above 1.2 g copolymer/kg and above 100 mg PTX/kg in mice respectively in contrast to 20˜24 mg/kg of Taxol®. The near infrared (NIR) fluorescence imaging showed that PPB/MPP micelles persisted for a relatively long time in the circulation and accumulated preferentially in tumor tissue. Moreover, PTX loaded PPB/MPP micelles significantly inhibited the tumor growth both in MDA-MB-231 and Ramos cancer xenograft mice models without obvious toxicity. Collectively, our study presents a new dual-functional micelles that improve the therapeutic efficacy of PTX in vitro and in vivo.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Dipeptídeos/química , Portadores de Fármacos , Indóis/química , Paclitaxel/farmacologia , Polietilenoglicóis/química , Células A549 , Animais , Antineoplásicos Fitogênicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cumarínicos/metabolismo , Feminino , Corantes Fluorescentes/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Micelas , Neoplasias/patologia , Paclitaxel/química , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Biomed Nanotechnol ; 15(7): 1515-1531, 2019 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-31196355

RESUMO

In our previous study, we identified a class of 4-substituted coumarins as a powerful microtubule inhibitors binding to the colchicine site of ß-tubulin. H6 showed potent anti-proliferative ability with IC50 values from 7 to 47 nM, and remarkable ability to reduce tumor growth in several xenograft models including taxol resistant tumor models. However, the extremely hydrophobicity limited its clinical application. In this study, to improve the anticancer activity and reduce the toxicity of H6, we successfully prepared MPEG-PCL with different proportions and H6-loaded polymeric micelles (H6/MPEG2kPCL2k micelles) by a simple thin-film hydration method. The prepared H6/MPEG-PCL micelles had a drug loading of 3.79 ± 0.001%, an encapsulation efficiency of 98.00 ± 0.41%, a mean particle size of 30.45 ± 0.18nm and a polydispersity index (PDI) of 0.096 ± 0.009. Computer simulation results revealed a good compatibility of H6 and MPEG2k-PCL2k copolymer. In in vitro release study and pharmacokinetic study showed H6 micelles can release H6 over an extended period. Furthermore, H6 micelles possessed comparative effect as free H6 in inhibiting cell growth, preventing cell migration, and inducing apoptosis. Mechanism study identified that H6 is a novel reversible microtubule inhibitor. In in vivo studies, H6 micelles exhibited tumor growth inhibition on two pulmonary metastatic tumor models (B16/F10 and 4T1). Importantly, H6 micelles significantly improved the solubility, reduced the toxicity, extended the half-life of drugs, and augmented the therapeutic window. All these results imply that H6 micelles have great potential for suppression of tumor metastasis.


Assuntos
Cumarínicos/química , Linhagem Celular Tumoral , Simulação por Computador , Portadores de Fármacos , Humanos , Micelas , Paclitaxel , Tamanho da Partícula , Poliésteres , Polietilenoglicóis
7.
J Biomed Nanotechnol ; 14(7): 1225-1238, 2018 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-29944097

RESUMO

To achieve higher therapeutic efficiency with catabatic side effects, desirable nanocarriers should be designed to retain the loaded drug tightly during the systemic circulation, but release the drug rapidly and efficiently upon endocytosis by tumor cells. Herein, we synthesized a novel folate conjugated poly(ethylene glycol)-poly(L-glutamic acid)-poly(L-phenylalanine) (folate-PEG-PLG(HS)-PPhe) copolymer to achieve a desired controlled delivery of doxorubicin (DOX). The copolymer could self-assemble into interlayer-crosslinked micelles with reduction sensitivity, and DOX was successfully loaded into the interior of copolymer. The interlayer-crosslinked disulfide bond at the intermediate region of between PEG and poly(L-phenylalanine) led to significant improvement of the system stability through the introduction of an additional mechanism of carrier/carrier interaction. The crosslinked interlayer could be cleaved at the desired target site under tumor-relevant reductive conditions and DOX were rapidly released from the DOX loaded folate-PEG-PLG (HS)-PPhe micelles (DOX-fPGPM), and significantly lowered the drug leakage without glutathione (GSH). Importantly, the DOX-fPGPM exhibited significantly higher antitumor efficiency both in vitro and in vivo in comparison with free DOX, and Doxil (commercial doxorubicin-loaded liposomes). Biodistribution studies showed that DOX more effectively accumulated in tumor tissue after iv injection of DOXfPGPM. The DOX-fPGPM designed in this work potentially resolved the dilemma between systemic stability and rapid intracellular drug release, and would provide a promising nanomedicine platform for cancer therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Doxorrubicina , Portadores de Fármacos , Humanos , Micelas , Polietilenoglicóis , Distribuição Tecidual
8.
J Biol Chem ; 293(24): 9461-9472, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29691282

RESUMO

Inhibitors that bind to the paclitaxel- or vinblastine-binding sites of tubulin have been part of the pharmacopoeia of anticancer therapy for decades. However, tubulin inhibitors that bind to the colchicine-binding site are not used in clinical cancer therapy, because of their low therapeutic index. To address multidrug resistance to many conventional tubulin-binding agents, numerous efforts have attempted to clinically develop inhibitors that bind the colchicine-binding site. Previously, we have found that millepachine (MIL), a natural chalcone-type small molecule extracted from the plant Millettia pachycarpa, and its two derivatives (MDs) SKLB028 and SKLB050 have potential antitumor activities both in vitro and in vivo However, their cellular targets and mechanisms are unclear. Here, biochemical and cellular experiments revealed that the MDs directly and irreversibly bind ß-tubulin. X-ray crystallography of the tubulin-MD structures disclosed that the MDs bind at the tubulin intradimer interface and to the same site as colchicine and that their binding mode is similar to that of colchicine. Of note, MDs inhibited tubulin polymerization and caused G2/M cell-cycle arrest. Comprehensive analysis further revealed that free MIL exhibits an s-cis conformation, whereas MIL in the colchicine-binding site in tubulin adopts an s-trans conformation. Moreover, introducing an α-methyl to MDs to increase the proportion of s-trans conformations augmented MDs' tubulin inhibition activity. Our study uncovers a new class of chalcone-type tubulin inhibitors that bind the colchicine-binding site in ß-tubulin and suggests that the s-trans conformation of these compounds may make them more active anticancer agents.


Assuntos
Chalconas/química , Chalconas/farmacologia , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia , Tubulina (Proteína)/metabolismo , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Sítios de Ligação/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Colchicina/farmacologia , Cristalografia por Raios X , Células Hep G2 , Humanos , Simulação de Acoplamento Molecular , Polimerização/efeitos dos fármacos , Tubulina (Proteína)/química
9.
Biomaterials ; 141: 188-198, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28689115

RESUMO

Honokiol (HK), a natural chemical isolated from Mangnolia officinalis, has shown antitumorigenic activities when used to treat a variety of tumor cell lines. The mechanism of honokiol activity when used to treat gefitinib-sensitive and gefitinib-resistant non-small cell lung cancer (NSCLC) requires elucidation. Here, the presence of liposomal honokiol (LHK) induced apoptotic and antitumor activities in four xenograft models generated using NSCLC cell lines such as HCC827 (gefitinib-sensitive) and H1975 (gefitinib-resistant). Mechanistic studies revealed that LHK inhibited the Akt and Erk1/2, both EGFR signaling cascades effectors, by promoting degradation of HSP90 client proteins (HCP), including wild-type or mutant EGFR, Akt and C-Raf. Molecular biology assays showed that LHK induced HCP degradation through a lysosomal pathway, rather than the canonical proteasome protein degradation pathway. As a result of misfolded protein accumulation, LHK induced endoplasmic reticulum (ER) stress and autophagy. Inhibition of ER stress (with 4-phenylbutyrate) or autophagy (with small interfering RNA) reduced LHK-induced HCP degradations. Additionally, LHK induced autophagy showed a protective role for cancer cell as inhibition of autophagy in vitro and in vivo by autophagosome degradation inhibitors could promote the anticancer activity of LHK. LHK has been approved by the China Food and Drug Administration for first-in-human clinical trials in NSCLC. The current study will guide the design of future LHK clinical trials.


Assuntos
Antineoplásicos/uso terapêutico , Autofagia/efeitos dos fármacos , Compostos de Bifenilo/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Proteínas de Choque Térmico HSP90/metabolismo , Lignanas/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Quinazolinas/uso terapêutico , Animais , Antineoplásicos/farmacologia , Compostos de Bifenilo/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Gefitinibe , Humanos , Lignanas/farmacologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos
10.
J Pharm Sci ; 104(11): 3934-3942, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26306026

RESUMO

This work aims to improve the solubility of 7-ethyl-14-aminocamptothecin (EACPT) by encapsulating it into monomethoxy poly(ethylene glycol)2000 -poly(lactic acid)2000 (MPEG-PLA) polymeric micelles. Micelles were prepared by a solid dispersion method; the properties including particle size distribution, morphology, drug loading, entrapment efficiency, crystallography property, solubility, and stability were characterized in detail. The results demonstrated that the EACPT-loaded MPEG-PLA polymeric micelle (EACPT-M) was successfully developed with a small particle size of 20.7 ± 0.2 nm, and the solubility was increased approximately 800-fold compared with free drug. In vitro release study showed sustained-release behavior of EACPT-M. Cytotoxicity assay suggested the incorporated EACPT maintained the potent antitumor effect of free drug. Furthermore, the obtained EACPT-M (1 mg/mL) did not induce hemolysis in vitro. Additionally, EACPT-M exhibited significant tumor growth inhibition in H460 human tumor xenograft model. The results indicated that the EACPT-M was a water-soluble, safe, and effective delivery system for human cancer chemotherapy.


Assuntos
Antineoplásicos/administração & dosagem , Camptotecina/análogos & derivados , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Preparações de Ação Retardada/química , Neoplasias Pulmonares/tratamento farmacológico , Micelas , Poliésteres/química , Polietilenoglicóis/química , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Camptotecina/administração & dosagem , Camptotecina/farmacologia , Camptotecina/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Células Hep G2 , Humanos , Ácido Láctico/química , Pulmão/efeitos dos fármacos , Pulmão/patologia , Neoplasias Pulmonares/patologia , Camundongos Nus , Polímeros/química
11.
J Pharmacol Sci ; 128(3): 116-24, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26220468

RESUMO

Honokiol (HK), a biphenolic neolignan isolated from Magnolia officinalis, has been reported to possess anti-inflammatory and anti-angiogenic activaties. In this study, our aim was to investigate anti-psoriatic activities of HK and the involved mechanisms. In vitro, the effects of HK on the regulation of Th1/Th2 and TNF-α-induced NF-κB (p65) activation were analyzed by respective FCS and immunofluorescence. Additionally, the K14-VEGF transgenic model was used for the in vivo study. ELISA and Q-PCR were performed to evaluate serum levels of Th1/Th2 cytokines and their corresponding mRNA expressions. Effects on VEGFR-2 and p65 activation, as well as other angiogenic and inflammatory parameters were studied by immunostainings. Importantly, we found that HK significantly decreased the ratio of Th1/Th2-expression CD4(+) T cells and inhibited TNF-α-induced activation of NF-κB. The morphology and histological features of psoriasis were effectively improved by HK treatment. The expression of TNF-α and IFN-γ, and their corresponding mRNA levels were down-regulated and the expression of nuclear p65, VEGFR-2, as well as related phosphorylated proteins (p-VEGFR-2, p-ERK1/2, p-AKT and p-p38) were also suppressed. Overall, these results in our study suggested that HK exhibits anti-psoriatic effects through the inhibition of NF-κB and VEGFR-2.


Assuntos
Compostos de Bifenilo/farmacologia , Compostos de Bifenilo/uso terapêutico , Lignanas/farmacologia , Lignanas/uso terapêutico , Terapia de Alvo Molecular , NF-kappa B/antagonistas & inibidores , Fitoterapia , Psoríase/tratamento farmacológico , Psoríase/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese , Animais , Anti-Inflamatórios , Compostos de Bifenilo/isolamento & purificação , Linfócitos T CD4-Positivos , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Lignanas/isolamento & purificação , Magnolia , Camundongos Transgênicos , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Psoríase/patologia , Equilíbrio Th1-Th2/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
J Pharmacol Sci ; 126(3): 198-207, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25341684

RESUMO

SKLB-M8, a derivative of millepachine, showed significant anti-proliferative effects in melanoma cell lines. In this study, we investigated the anti-melanoma and anti-angiogenic activity of SKLB-M8 on three melanoma cell lines (A2058, CHL-1, and B16F10) and human umbilical vein endothelial cells (HUVECs). In vitro, SKLB-M8 showed anti-proliferative activity with IC50 values of 0.07, 0.25, and 0.88 µM in A2058, CHL-1, and B16F10 cell lines, respectively. Flow cytometory analysis showed that SKLB-M8 induced G2/M arrest in three melanoma cell lines, and western blotting demonstrated that SKLB-M8 down-regulated the expression of cdc2, up-regulated p53 in A2058 and CHL-1 cells, and triggered cell apoptosis through down-regulating AKT and phosphorylated mTOR (p-mTOR). SKLB-M8 also inhibited HUVEC proliferation, migration, invasion, and tube formation in vitro with the inhibition of phosphorylated ERK1/2 (p-ERK1/2). In vivo, alginate-encapsulated tumor cell assay revealed that SKLB-M8 suppressed B16F10 tumor angiogenesis. In CHL-1- and B16F10-tumor-bearing mouse models, SKLB-M8 inhibited tumor growth by oral treatment with less toxicity. CD31 immunofluoresence staining and caspase-3 immunohistochemistry indicated that SKLB-M8 inhibited melanoma tumor growth by targeting angiogenesis and inducing caspase3-dependent apoptosis. SKLB-M8 might be a potential anti-melanoma drug candidate.


Assuntos
Inibidores da Angiogênese/farmacologia , Apoptose/efeitos dos fármacos , Chalconas/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Melanoma Experimental/tratamento farmacológico , Melanoma/tratamento farmacológico , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , Serina-Treonina Quinases TOR/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Concentração Inibidora 50 , Melanoma/enzimologia , Melanoma/patologia , Melanoma Experimental/enzimologia , Melanoma Experimental/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Neovascularização Patológica , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/patologia , Fatores de Tempo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Med Chem ; 57(19): 7977-89, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25208345

RESUMO

Twenty-one novel derivatives of millepachine were synthesized and evaluated for their in vitro antiproliferative activity. Among them, 8 exhibited the most potent activity, with IC50 values of 8-27 nM against panel of cancer cell lines and retained full activity in multidrug resistant cancer cells. Treated cells were arrested in G2/M phase and resulted in cellular apoptosis. Microtubule dynamics confirmed 8 was a novel tubulin polymerization inhibitor by binding at the colchicine site. 8 also exhibited antivascular activity because it concentration dependently reduced the cell migration and disrupted capillary like tube formation in HUVEC cells. Furthermore, the hydrochloride salt of 8 (8·HCl) significantly improved the bioavailability up to 47% while retaining the antiproliferative activity. Importantly, 8·HCl significantly inhibited tumor growths in four xenograft models including resistance tumor-cell-bearing mice models without causing significant loss of body weight, suggesting that 8 is a promising new orally anticancer agent to be developed.


Assuntos
Antineoplásicos/síntese química , Chalconas/farmacologia , Moduladores de Tubulina/síntese química , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Chalconas/química , Resistência a Múltiplos Medicamentos , Imunofluorescência , Células Hep G2 , Humanos , Camundongos Endogâmicos BALB C , Relação Estrutura-Atividade , Moduladores de Tubulina/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Dermatol Sci ; 74(3): 242-50, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24685902

RESUMO

BACKGROUND: Psoriasis is a chronic T cell-mediated inflammatory skin disease. Studies have shown that angiogenesis plays an important role in the pathogenesis of psoriasis. Studies have also indicated that Gambogic acid (GA) inhibits angiogenesis and may be a viable drug candidate in anti-angiogenesis therapies. OBJECTIVE: The aim of this study was to investigate the anti-psoriatic effect of GA and the possible mechanisms. METHODS: MTT test on HaCaT cells and immunofluorescence on HUVEC cells were processed. An O/W cream of GA was prepared and topically applied to the ears of K14-VEGF transgenic mice and psoriasis-like guinea-pigs, and the tail skin of Balb/C mice independently. Furthermore, hematoxylin-eosin staining of tissues from three models and immunohistochemistry staining of ear samples from K14-VEGF mice were performed. RESULTS: In vitro, GA inhibited proliferation of HaCaTs and TNF-α-induced activation of NF-κB in HUVECs. In vivo, animals treated with GA showed significant morphological and histological improvements. Immunohistochemical analysis of K14-VEGF transgenic mice revealed that hyperplastic and inflamed vessels were suppressed with GA treatment. The expression of adhesion molecules such as ICAM-1 and E-selectin was significantly decreased. GA inhibited angiogenesis and the expression of VEGFR2 and p-VEGFR2. T lymphocyte infiltration and the expression of IL-17 and IL-22 were also reduced by GA treatment. CONCLUSION: Our results suggest that GA has anti-psoriatic efficacy through inhibition of angiogenesis and inflammation. Therefore, GA is attractive and offers future potential for application in patients with psoriasis.


Assuntos
Garcinia , Fitoterapia , Psoríase/tratamento farmacológico , Xantonas/uso terapêutico , Animais , Linhagem Celular , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Cobaias , Células Endoteliais da Veia Umbilical Humana , Humanos , Hiperplasia , Inflamação/tratamento farmacológico , Camundongos Transgênicos , NF-kappa B/metabolismo , Neovascularização Patológica/tratamento farmacológico , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Plantas Medicinais , Distribuição Aleatória , Pele/efeitos dos fármacos , Pele/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Xantonas/farmacologia
15.
Int J Nanomedicine ; 9: 243-55, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24403830

RESUMO

The clinical application of gambogic acid, a natural component with promising antitumor activity, is limited due to its extremely poor aqueous solubility, short half-life in blood, and severe systemic toxicity. To solve these problems, an amphiphilic polymer-drug conjugate was prepared by attachment of low molecular weight (ie, 2 kDa) methoxy poly(ethylene glycol) methyl ether (mPEG) to gambogic acid (GA-mPEG2000) through an ester linkage and characterized by (1)H nuclear magnetic resonance. The GA-mPEG2000 conjugates self-assembled to form nanosized micelles, with mean diameters of less than 50 nm, and a very narrow particle size distribution. The properties of the GA-mPEG2000 micelles, including morphology, stability, molecular modeling, and drug release profile, were evaluated. MTT (3-(4,5-dimethylthiazo l-2-yl)-2,5 diphenyl tetrazolium bromide) tests demonstrated that the GA-mPEG2000 micelle formulation had obvious cytotoxicity to tumor cells and human umbilical vein endothelial cells. Further, GA-mPEG2000 micelles were effective in inhibiting tumor growth and prolonged survival in subcutaneous B16-F10 and C26 tumor models. Our findings suggest that GA-mPEG2000 micelles may have promising applications in tumor therapy.


Assuntos
Água Corporal/química , Nanocápsulas/química , Neoplasias Experimentais/química , Neoplasias Experimentais/tratamento farmacológico , Polietilenoglicóis/química , Xantonas/administração & dosagem , Xantonas/química , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Sinergismo Farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Micelas , Nanocápsulas/administração & dosagem , Nanocápsulas/ultraestrutura , Solubilidade , Resultado do Tratamento
16.
Carbohydr Polym ; 101: 623-30, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24299819

RESUMO

The aim of this study was to improve the water solubility of barbigerone by complexing it with hydroxypropyl-ß-cyclodextrin (HP-ß-CD). The inclusion complexation behavior, characterization and interactions of barbigerone with HP-ß-CD were investigated in both solution and the solid state by means of UV/VIS, (1)H NMR, FT-IR, PXRD, SEM. All the characterization information demonstrated the formation of barbigerone-HP-ß-CD (bar-HP-ß-CD) inclusion complex, and the bar-HP-ß-CD inclusion compounds exhibited different spectroscopic features and properties from barbigerone. The results demonstrated that the water solubility of barbigerone was notably increased in the presence of HP-ß-CD. Furthermore, preliminary in vitro cytotoxicity assay showed that bar-HP-ß-CD still maintain the anticancer activity of barbigerone. These results suggest that HP-ß-CD will be potentially useful in the delivery of water-insoluble anticancer agents such as barbigerone.


Assuntos
Isoflavonas/química , Isoflavonas/farmacologia , beta-Ciclodextrinas/química , 2-Hidroxipropil-beta-Ciclodextrina , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Solubilidade , Água/química
17.
PLoS One ; 8(4): e60266, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23560085

RESUMO

The Hedgehog (Hh) pathway inhibitors have shown great promise in cancer therapeutics. SANT75, a novel compound we previously designed to specially inhibit the Smoothened (SMO) protein in the Hh pathway, has greater inhibitory potency than many of commonly used Hh inhibitors. However, preclinical studies of SANT75 revealed water insolubility and acute toxicity. To overcome these limitations, we developed a liposomal formulation of SANT75 and investigated its antitumor efficacy in vitro and in vivo. We encapsulated SANT75 into PEGylated liposome and the mean particle size distribution and zeta-potential (ZP) of liposomes were optimized. Using the Shh-light2 cell and Gli-GFP or Flk-GFP transgenic reporter zebrafish, we confirmed that liposome-encapsulated SANT75 inhibited Hh activity with similar potency as the original SANT75. SANT75 encapsulated into liposome exerted strong tumor growth-inhibiting effects in vitro and in vivo. In addition, the liposomal SANT75 therapy efficiently improved the survival time of tumor-bearing mice without obvious systemic toxicity. The pathological morphology and immunohistochemistry staining revealed that liposomal SANT75 induced tumor cell apoptosis, inhibited tumor angiogenesis as assessed by CD31 and down-regulated the expression of Hh target protein Gli-1 in tumor tissues. Our findings suggest that liposomal formulated SANT75 has improved solubility and bioavailability and should be further developed as a drug candidate for treating tumors with abnormally high Hh activity.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Lipossomos/química , Proteínas Oncogênicas/antagonistas & inibidores , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transativadores/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Composição de Medicamentos , Embrião não Mamífero , Feminino , Genes Reporter , Proteínas de Fluorescência Verde , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Polietilenoglicóis/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Transativadores/genética , Transativadores/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra , Proteína GLI1 em Dedos de Zinco
18.
Eur J Med Chem ; 62: 579-89, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23425970

RESUMO

Twenty-five novel pyranochalcone derivatives were synthesized and evaluated for their in vitro and in vivo antiproliferative activities. Among them, compound 10i exhibited superior potent activity against 21 tumor cell lines including multidrug resistant phenotype with the IC50 values ranged from 0.09 to 1.30 µM. In addition, 10i significantly induced cell cycle arrest in G2/M phase, promoted tubulin polymerization into microtubules and caused microtubule stabilization. Further studies confirmed that 10i significantly suppressed the growth of tumor volume in HepG2 xenograft tumor model. Our study demonstrated that 10i could have beneficial antitumor activity as a novel microtubule stabilizing agent.


Assuntos
Antineoplásicos/farmacologia , Benzopiranos/farmacologia , Calônios/farmacologia , Microtúbulos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Moduladores de Tubulina/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Benzopiranos/síntese química , Benzopiranos/química , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Calônios/síntese química , Calônios/química , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Células Hep G2 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Estrutura Molecular , Neoplasias/patologia , Relação Estrutura-Atividade , Moduladores de Tubulina/síntese química , Moduladores de Tubulina/química
19.
Int J Nanomedicine ; 7: 4499-510, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22923988

RESUMO

BACKGROUND AND METHODS: Paclitaxel, a widely used antitumor agent, has limited clinical application due to its hydrophobicity and systemic toxicity. To achieve sustained and targeted delivery of paclitaxel to tumor sites, liposomes composed of egg phosphatidylcholine, cholesterol, and distearolyphosphatidyl ethanolamine-N-poly(ethylene glycol) (PEG(2000)) were prepared by a lipid film method. In addition, the liposomes also contained truncated fibroblast growth factor fragment-PEG-cholesterol as a ligand targeting the tumor marker fibroblast growth factor receptor. Physicochemical characteristics, such as particle size, zeta potential, entrapment efficiency, and release profiles were investigated. Pharmacokinetics and biodistribution were evaluated in C57BL/6 J mice bearing B16 melanoma after intravenous injection of paclitaxel formulated in Cremophor EL (free paclitaxel), conventional liposomes (CL-PTX), or in targeted PEGylated liposomes (TL-PTX). RESULTS: Compared with CL-PTX and free paclitaxel, TL-PTX prolonged the half-life of paclitaxel by 2.01-fold and 3.40-fold, respectively, in plasma and improved the AUC(0→t) values of paclitaxel by 1.56-fold and 2.31-fold, respectively, in blood. Biodistribution studies showed high accumulation of TL-PTX in tumor tissue and organs containing the mononuclear phagocyte system (liver and spleen), but a considerable decrease in other organs (heart, lung, and kidney) compared with CL-PTX and free paclitaxel. CONCLUSION: The truncated fibroblast growth factor fragment-conjugated PEGylated liposome has promising potential as a long-circulating and tumor-targeting carrier system.


Assuntos
Antineoplásicos/farmacocinética , Lipossomos/farmacocinética , Paclitaxel/farmacocinética , Peptídeos/metabolismo , Polietilenoglicóis/química , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Antineoplásicos/sangue , Antineoplásicos/química , Área Sob a Curva , Sítios de Ligação , Linhagem Celular Tumoral , Colesterol/química , Estabilidade de Medicamentos , Feminino , Humanos , Ligantes , Lipossomos/sangue , Lipossomos/química , Masculino , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/enzimologia , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Paclitaxel/sangue , Paclitaxel/química , Peptídeos/química , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Distribuição Aleatória , Receptores de Fatores de Crescimento de Fibroblastos/biossíntese , Distribuição Tecidual
20.
PLoS One ; 6(4): e18490, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21559510

RESUMO

BACKGROUND: Gliosarcoma is one of the most common malignant brain tumors, and anti-angiogenesis is a promising approach for the treatment of gliosarcoma. However, chemotherapy is obstructed by the physical obstacle formed by the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB). Honokiol has been known to possess potent activities in the central nervous system diseases, and anti-angiogenic and anti-tumor properties. Here, we hypothesized that honokiol could cross the BBB and BCSFB for the treatment of gliosarcoma. METHODOLOGIES: We first evaluated the abilities of honokiol to cross the BBB and BCSFB by measuring the penetration of honokiol into brain and blood-cerebrospinal fluid, and compared the honokiol amount taken up by brain with that by other tissues. Then we investigated the effect of honokiol on the growth inhibition of rat 9L gliosarcoma cells and human U251 glioma cells in vitro. Finally we established rat 9L intracerebral gliosarcoma model in Fisher 344 rats and human U251 xenograft glioma model in nude mice to investigate the anti-tumor activity. PRINCIPAL FINDINGS: We showed for the first time that honokiol could effectively cross BBB and BCSFB. The ratios of brain/plasma concentration were respectively 1.29, 2.54, 2.56 and 2.72 at 5, 30, 60 and 120 min. And about 10% of honokiol in plasma crossed BCSFB into cerebrospinal fluid (CSF). In vitro, honokiol produced dose-dependent inhibition of the growth of rat 9L gliosarcoma cells and human U251 glioma cells with IC(50) of 15.61 µg/mL and 16.38 µg/mL, respectively. In vivo, treatment with 20 mg/kg body weight of honokiol (honokiol was given twice per week for 3 weeks by intravenous injection) resulted in significant reduction of tumor volume (112.70±10.16 mm(3)) compared with vehicle group (238.63±19.69 mm(3), P = 0.000), with 52.77% inhibiting rate in rat 9L intracerebral gliosarcoma model, and (1450.83±348.36 mm(3)) compared with vehicle group (2914.17±780.52 mm(3), P = 0.002), with 50.21% inhibiting rate in human U251 xenograft glioma model. Honokiol also significantly improved the survival over vehicle group in the two models (P<0.05). CONCLUSIONS/SIGNIFICANCE: This study provided the first evidence that honokiol could effectively cross BBB and BCSFB and inhibit brain tumor growth in rat 9L intracerebral gliosarcoma model and human U251 xenograft glioma model. It suggested a significant strategy for offering a potential new therapy for the treatment of gliosarcoma.


Assuntos
Antineoplásicos/farmacologia , Compostos de Bifenilo/farmacocinética , Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Gliossarcoma/tratamento farmacológico , Lignanas/farmacocinética , Animais , Apoptose , Linhagem Celular Tumoral , Líquido Cefalorraquidiano/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...