Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Technol Cancer Res Treat ; 20: 15330338211064434, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34931914

RESUMO

Objective: This study aimed to investigate the tolerance and pharmacokinetic characteristics of recombinant human endostatin (rh-endostatin) administered as single-dose or multiple-dose infusions in patients with advanced solid tumors. Methods: This phase I trial was designed as a single-center, single-arm, nonrandomized, open-label, dose-escalation study. The trial consisted of 2 parts: a single-dose part and a multiple-dose part, each with 3 dose comparison groups. Rh-endostatin was administered as an intravenous injection only once at a dose of 5 mg/m2, 7.5 mg/m2, or 10 mg/m2 in the single-dose part and as a daily intravenous injection for 14 days at the same doses in the multiple-dose part. The serum pharmacokinetics, toxicity and immunogenicity of rh-endostatin were evaluated. Results: Dose-limiting toxicity (DLT) was not observed in any group. A few patients developed cardiotoxicity, such as QT prolongation or narrow arrhythmia. Other adverse events were slight coagulation abnormalities and haematological abnormalities. For rh-endostatin doses of 5 mg/m2, 7.5 mg/m2, and 10 mg/m2, the mean Cmax values in the single-dose part were 344 ± 38.7 ng/mL, 524 ± 157 ng/mL, and 800 ± 201 ng/mL, respectively, and the average AUC0-t values were 3290 ± 3790 ng•h/mL, 4940 ± 4380 ng•h/mL, and 5050 ± 3980 ng•h/mL, respectively. The Cmax ss values of the 3 doses in the multiple-dose part were 575 ± 270 ng/mL, 531 ± 106 ng/mL, and 864 ± 166 ng/mL, respectively, and the AUC0-τ values were 3610 ± 1040 ng•h/mL, 3290 ± 1090 ng•h/mL, and 5180 ± 1210 ng•h/mL, respectively. The Cmax of a single-dose regimen showed linear kinetic characteristics. The patients in the single-dose group were negative for serum antibodies against rh-endostatin, while one patient in the multiple-dose group was positive. Conclusions: Rh-endostatin as a daily intravenous injection for 14 days in patients with advanced solid tumors is safe and well tolerated, without DLT, at doses of 5 mg/m2, 7.5 mg/m2, and 10 mg/m2. Serum antibodies against rh-endostatin were very low after multiple infusions. For phase II trials, the recommended rh-endostatin dose is 10 mg/m2 as a daily intravenous injection for 14 days.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Endostatinas/administração & dosagem , Endostatinas/farmacocinética , Neoplasias/tratamento farmacológico , Proteínas Recombinantes , Adulto , Idoso , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/farmacocinética , Antineoplásicos/efeitos adversos , Gerenciamento Clínico , Relação Dose-Resposta a Droga , Monitoramento de Medicamentos , Endostatinas/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/diagnóstico , Neoplasias/mortalidade , Prognóstico , Resultado do Tratamento
2.
Life Sci ; 238: 116967, 2019 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-31626794

RESUMO

To investigate the pharmacokinetics of PEGylated recombinant human endostatin (M2ES) in rhesus monkey. M2ES was administered to rhesus monkeys by intravenous bolus injection, and serum M2ES concentration was determined by a self-developed ELISA assay. Pharmacokinetic parameters were calculated using a non-compartmental model of WinNonlin V2.1A software. The standard curve of self-developed ELISA assay was fitted by four-parameter method. The limit of detection (LOD) and LOQ were 0.3050 ng/mL and 0.9140 ng/mL, respectively. Following IV infusions of M2ES at 0.3, 1, and 3 mg/kg in rhesus monkeys, the serum M2ES concentration-time curve was fitted with a non-compartment model. The pharmacokinetic parameters were evaluated as follows: Terminal elimination half-life (T1/2) of M2ES were 3.30 ±â€¯0.70, 29.50 ±â€¯18.80 and 24.60 ±â€¯8.90 h. Systemic clearance (CLsys) of M2ES were 339.60 ±â€¯66.30, 161.40 ±â€¯18.20 and 260.10 ±â€¯43.70 mL/h/kg. AUC(0-∞) values of M2ES were 909.30 ±â€¯199.60, 6251.00 ±â€¯739.60 and 11758.00 ±â€¯2010.10 ng∙h/mL. The dosage was positively correlated with AUC, and the correlation coefficient r2 = 0.9327. Self-developed ELISA assay could meet the requirements of serum M2ES concentration detection. PEGylation modification substantially expands the circulation time of recombinant human endostatin and effectively improves its pharmacokinetic behavior.


Assuntos
Antineoplásicos/farmacocinética , Endostatinas/farmacocinética , Polietilenoglicóis/química , Proteínas Recombinantes/farmacocinética , Animais , Antineoplásicos/administração & dosagem , Área Sob a Curva , Endostatinas/administração & dosagem , Feminino , Humanos , Injeções Intravenosas , Macaca mulatta , Masculino , Proteínas Recombinantes/administração & dosagem , Distribuição Tecidual
3.
Carbohydr Polym ; 207: 79-90, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30600066

RESUMO

Endostatin (ES) has attracted considerable attention for the treatment of anti-angiogenesis-related disorders. An 11-amino-acid peptide (ES2, IVRRADRAAVP) from the amino terminal of ES is of interest because it is the main active fragment of ES. However, both ES and ES2 have a poor stability and a short half-life, and other disadvantages need to be further resolved. Thus, we conjugated ES2 to glycol-split heparin derivatives (GSHPs) to yield the polymer-peptide conjugate, GSHP-ES2. This study showed that GSHP-ES2 exhibited increased stability, a wider pH activity range, better inhibition of endothelial cell proliferation, migration and tube formation in vitro, better anti-angiogenic activity and a longer half-life in vivo compared with ES2. These results also indicate that GSHP-ES2 has good potential for the treatment of angiogenesis-related diseases, either alone or in combination with other chemicals.


Assuntos
Inibidores da Angiogênese/farmacologia , Endostatinas/farmacologia , Glicopeptídeos/farmacologia , Heparina/química , Oligopeptídeos/farmacologia , Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/farmacocinética , Inibidores da Angiogênese/toxicidade , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Galinhas , Estabilidade de Medicamentos , Endostatinas/síntese química , Endostatinas/farmacocinética , Endostatinas/toxicidade , Feminino , Glicopeptídeos/síntese química , Glicopeptídeos/farmacocinética , Glicopeptídeos/toxicidade , Meia-Vida , Humanos , Camundongos Endogâmicos BALB C , Oligopeptídeos/síntese química , Oligopeptídeos/farmacocinética , Oligopeptídeos/toxicidade , Peixe-Zebra
4.
Artigo em Inglês | MEDLINE | ID: mdl-29935477

RESUMO

HYD-PEP06 is a novel RGD-modified Endostar mimetic peptide with 30 amino acids that is intended to suppress the formation of neoplasm vessels. This assay was developed and validated to monitor the level of the peptide HYD-PEP06 in rat blood, using liquid chromatography tandem mass spectrometry (LC-MS/MS). HYD-PEP10, another peptide similar to the analyte, was used as an internal standard (IS). A triple quadrupole mass spectrometry in Multiple Reaction Monitoring (MRM) mode and an electrospray interface (ESI) in the positive mode were used for MS analysis. The analysis was optimized with addition of 0.3% formic acid (FA) into the mobile phase as well as with a needle washing solution to overcome the carryover effect. In addition, the carryover was reduced by optimizing the mobile phase gradient. Methanol was used as a diluent of working solutions to avoid any adsorption. Methanol:acetonitrile (1:1, v:v) containing 0.3% FA was employed to precipitate the blood samples. Unknown blood samples must be placed in ice bath immediately, and precipitating agents should be added within 30 min to ensure the stability of blood samples. The assay was established and validated. This method showed a good linear relationship for the HYD-PEP06 in the range of 10 ng·mL-1 to 2000 ng·mL-1, with R > 0.99. HYD-PEP06 was determined with accuracy values (RE%) of -5.06%-8.54%, intra- and inter-day precisions (RSD%) of 3.13%-4.87% and 4.81%-9.42%. The method was successfully in monitoring the concentration of HYD-PEP06 in rat blood.


Assuntos
Inibidores da Angiogênese/sangue , Cromatografia Líquida/métodos , Endostatinas/sangue , Oligopeptídeos/sangue , Espectrometria de Massas em Tandem/métodos , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacocinética , Animais , Estabilidade de Medicamentos , Endostatinas/química , Endostatinas/farmacocinética , Feminino , Modelos Lineares , Masculino , Oligopeptídeos/química , Oligopeptídeos/farmacocinética , Ratos , Ratos Wistar , Proteínas Recombinantes , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
5.
Curr Pharm Biotechnol ; 19(12): 996-1004, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30761959

RESUMO

BACKGROUND: Endostatin (ES) is a promising anti-angiogenesis protein and has been approved for the treatment of non-small cell lung cancer, but short half-life, poor stability and nonspecific delivery caused great pain to patients and produced unsatisfactory treatment effectiveness. OBJECTIVE: In this work, in order to overcome these disadvantages, ES was covalently modified by polysulfated heparin (PSH) with the expectancy of longer half-life, higher anti-angiogenesis activity and better cellular uptake. METHODS: To characterize the cellular uptake, flow cytometry and confocal laser scanning microscopy were used to study the intracellular localization of fluorescein isothiocyanate-labeled ES and PSH-ES in EAhy926 endothelial cells. Zebrafish model was used to study the anti-angiogenesis activities of ES and its derivatives in vivo. The 125I-radiolabeled ES and PSH-ES were administered to healthy BALC/c mice for the pharmacokinetics study. RESULTS: Compared with ES, better cellular uptake effects were detected in PSH-ES group. Both ES and PSH-ES showed inhibition on the intersegmental vessels formation, while PSH-ES displayed a higher one. The half-life of PSH-ES was lengthened and area under the curve (AUC) was increased. At the same time, ES and PSH-ES were both widely and rapidly distributed in the lungs, livers, kidneys and hearts with little difference. CONCLUSION: The results indicated that PSH displayed good properties as a novel glyco-modifier for protein and peptide. The results also showed that PSH-ES displayed better cellular uptake, higher antiangiogenesis activity and prolonged half-life, which would lead to better anti-tumour effects.


Assuntos
Inibidores da Angiogênese/farmacologia , Endostatinas/farmacologia , Células Endoteliais/metabolismo , Heparina de Baixo Peso Molecular/química , Neovascularização Fisiológica/efeitos dos fármacos , Peixe-Zebra/sangue , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacocinética , Animais , Transporte Biológico , Endostatinas/química , Endostatinas/farmacocinética , Feminino , Meia-Vida , Humanos , Camundongos
6.
Nucl Med Commun ; 38(11): 919-926, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28863122

RESUMO

BACKGROUND: Gliomas are rich in blood vessels and are the most primary and malignant type of brain tumor affecting the central nervous system. A few fluorine-18 (F)-labeled imaging agents can be used for imaging of tumor angiogenesis. In the current study, F-labeled recombinant human endostatin (rh-endostatin) was developed and evaluated as a probe for PET imaging of tumor angiogenesis. MATERIALS AND METHODS: F-fluorobenzoyl-endostatin (F-FB-endostatin) was synthesized from radiolabeling of rh-endostatin with N-succinimidyl-4-F-fluorobenzoate produced by a facile module-assisted radiosynthesis procedure. Blocking studies were used to measure the relative affinities of F-FB-endostatin to human glioblastoma U87MG cells in tumor tissues rich with vessels. In addition, biodistribution, metabolic stability, and small-animal PET imaging studies were carried out with F-FB-endostatin using Institute of Cancer Research and U87MG tumor-bearing mice. RESULTS: Noninvasive small-animal PET imaging indicated that F-FB-endostatin showed rapid and good tumor uptake. The probe was rapidly cleared from the blood and most organs, resulting in excellent tumor-to-normal tissue contrasts. Tumor uptake and rapid clearance were further confirmed with biodistribution studies. Metabolite assays showed that the probe was highly stable, making it suitable for in-vivo applications. CONCLUSION: F-FB-endostatin shows promising in-vivo properties. Therefore, the promising properties of F-FB-endostatin indicate that this probe can be a powerful tool to evaluate the antiangiogenic therapy for gliomas and thus warrants further investigation as a novel PET probe for imaging of tumor angiogenesis.


Assuntos
Endostatinas , Glioma/irrigação sanguínea , Neovascularização Patológica/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Animais , Linhagem Celular Tumoral , Endostatinas/farmacocinética , Radioisótopos de Flúor , Glioma/diagnóstico por imagem , Humanos , Marcação por Isótopo , Camundongos , Distribuição Tecidual
7.
J Pathol ; 243(2): 255-266, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28771739

RESUMO

Endostatin was discovered as an endogenous angiogenesis inhibitor with broad-spectrum antitumour activities. Although clinical efficacy was observed when endostatin was combined with standard chemotherapy for non-small cell lung cancer (NSCLC), as well as other cancer types, the specific mechanisms underlying the benefit of endostatin are not completely understood. Extensive investigations suggest that endostatin is a multifunctional protein possessing more than anti-angiogenic activity. Here, we found that endostatin exerts a direct chemosensitizing effect on p53-deficient tumour cells. Concomitant treatment with endostatin and genotoxic drugs resulted in therapeutic synergy in both cellular and animal models of p53-deficient NSCLC. Mechanistically, endostatin specifically interacts with DNA-dependent protein kinase catalytic subunit (DNA-PKcs) in tumour cells and suppresses its DNA repair activity. Using isogenic NSCLC cells with different p53 statuses, we discovered that p53-deficient tumour cells show chemoresistance to genotoxic drugs, creating a synthetic dependence on DNA-PKcs-mediated DNA repair. In this setting, endostatin exerted inhibitory effects on DNA-PKcs activity, leading to accumulation of DNA lesions and promotion of the therapeutic effect of genotoxic chemotherapy. In contrast, p53-proficient tumour cells were more sensitive to genotoxic drugs so that DNA-PKcs could be cleaved by drug-activated caspase-3, making DNA-PKcs inhibition less effective during this ongoing apoptotic process. Therefore, our data demonstrate a novel mechanism for endostatin as a DNA-PKcs suppressor, and indicate that combination therapy of endostatin with genotoxic drugs could be a promising treatment strategy for cancer patients with p53-deficient tumours. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Endostatinas/farmacocinética , Neoplasias Pulmonares/tratamento farmacológico , Proteína Supressora de Tumor p53/deficiência , Animais , Antineoplásicos/uso terapêutico , Apoptose/fisiologia , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Linhagem Celular Tumoral , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Proteína Quinase Ativada por DNA/metabolismo , Xenoenxertos , Humanos , Neoplasias Pulmonares/mortalidade , Camundongos Nus , Transplante de Neoplasias
8.
Biotechnol Lett ; 38(5): 801-7, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26830094

RESUMO

OBJECTIVE: To produce a therapeutic protein (endostatin) by fusion with two fragments of the carboxyl-terminal peptide (CTP) of the human chorionic gonadotropin ß-subunit in Pichia pastoris. RESULTS: Two CTP sequences were fused to the C-terminal of human endostatin, and the fusion protein (endo-CTP) was expressed by P. pastoris. Endo-CTP inhibited proliferation of endothelial cells with an IC50 of 7 µg ml(-1), and 30 % of cells were annexin V-positive after treatment with 20 µg endo-CTP ml(-1) for 48 h. Migration of endothelial cells was inhibited by endo-CTP in a concentration-dependent manner. The half-life of endo-CTP in Sprague-Dawley rats was much longer than that of its commercial counterpart (Endostar). CONCLUSION: A long-acting endostatin can be produced using CTP technology.


Assuntos
Antineoplásicos/metabolismo , Gonadotropina Coriônica/metabolismo , Endostatinas/metabolismo , Subunidades Proteicas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Antineoplásicos/farmacocinética , Proliferação de Células/efeitos dos fármacos , Gonadotropina Coriônica/genética , Endostatinas/genética , Endostatinas/farmacocinética , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Meia-Vida , Concentração Inibidora 50 , Pichia/genética , Pichia/metabolismo , Subunidades Proteicas/genética , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética
9.
Acta Pharmacol Sin ; 36(7): 847-54, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26027657

RESUMO

AIM: M2ES is PEGylated recombinant human endostatin. In this study we investigated the pharmacokinetics, tissue distribution, and excretion of M2ES in rats. METHODS: (125)I-radiolabeled M2ES was administered to rats by intravenous bolus injection at 3 mg/kg. The pharmacokinetics, tissue distribution and excretion of M2ES were investigated using the trichloroacetic acid (TCA) precipitation method. RESULTS: The serum M2ES concentration-time curve after a single intravenous dose of 3 mg/kg in rats was fitted with a non-compartment model. The pharmacokinetic parameters were evaluated as follows: Cmax=28.3 µg·equ/mL, t1/2=71.5 h, AUC(0-∞)=174.6 µg·equ·h/mL, Cl=17.2 mL·h(-1)·kg(-1), MRT=57.6 h, and Vss=989.8 mL/kg for the total radioactivity; Cmax=30.3 µg·equ/mL, t1/2=60.1 h, AUC(0-∞)=146.2 µg·equ·h/mL, Cl=20.6 mL·h(-1)·kg(-1), MRT=47.4 h, and Vss=974.6 mL/kg for the TCA precipitate radioactivity. M2ES was rapidly and widely distributed in various tissues and showed substantial deposition in kidney, adrenal gland, lung, spleen, bladder and liver. The radioactivity recovered in the urine and feces by 432 h post-dose was 71.3% and 8.3%, respectively. Only 0.98% of radioactivity was excreted in the bile by 24 h post-dose. CONCLUSION: PEG modification substantially prolongs the circulation time of recombinant human endostatin and effectively improves its pharmacokinetic behavior. M2ES is extensively distributed in most tissues of rats, including kidney, adrenal gland, lung, spleen, bladder and liver. Urinary excretion was the major elimination route for M2ES.


Assuntos
Endostatinas/farmacocinética , Polietilenoglicóis/farmacocinética , Animais , Feminino , Humanos , Masculino , Ligação Proteica/fisiologia , Ratos , Ratos Wistar , Proteínas Recombinantes/farmacocinética , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia
10.
Int J Nanomedicine ; 10: 3791-802, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26060399

RESUMO

Tumor angiogenesis plays a key role in tumor growth and metastasis; thus, targeting tumor-associated angiogenesis is an important goal in cancer therapy. However, the efficient delivery of drugs to tumors remains a key issue in antiangiogenesis therapy. GX1, a peptide identified by phage-display technology, is a novel tumor vasculature endothelium-specific ligand and possesses great potential as a targeted vector and antiangiogenic agent in the diagnosis and treatment of human cancers. Endostar, a novel recombinant human endostatin, has been shown to inhibit tumor angiogenesis. In this study, we developed a theranostic agent composed of GX1-conjugated poly(lactic acid) nanoparticles encapsulating Endostar (GPENs) and labeled with the near-infrared dye IRDye 800CW to improve colorectal tumor targeting and treatment efficacy in vivo. The in vivo fluorescence molecular imaging data showed that GPENs (IRDye 800CW) more specifically targeted tumors than free IRDye 800CW in colorectal tumor-bearing mice. Moreover, the antitumor efficacy was evaluated by bioluminescence imaging and immunohistology, revealing that GPENs possessed improved antitumor efficacy on subcutaneous colorectal xenografts compared to other treatment groups. Thus, our study showed that GPENs, a novel GX1 peptide guided form of nanoscale Endostar, can be used as a theranostic agent to facilitate more efficient targeted therapy and enable real-time monitoring of therapeutic efficacy in vivo.


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Endostatinas/farmacocinética , Nanopartículas , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/química , Animais , Benzenossulfonatos/química , Linhagem Celular Tumoral/efeitos dos fármacos , Neoplasias Colorretais/irrigação sanguínea , Endostatinas/administração & dosagem , Endostatinas/farmacologia , Fluorescência , Humanos , Indóis/química , Ácido Láctico/química , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Imagem Molecular/métodos , Nanopartículas/administração & dosagem , Nanopartículas/química , Neovascularização Patológica/tratamento farmacológico , Peptídeos/química , Poliésteres , Polímeros/química , Proteínas Recombinantes , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Regul Toxicol Pharmacol ; 69(3): 512-23, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24878240

RESUMO

PEGylated recombinant human endostatin (M2ES) exhibited prolonged serum half-life and enhanced antitumor activity when compared with endostatin. A non-clinical study was performed to evaluate the toxicokinetics and safety of M2ES in rhesus monkeys. After intravenous (IV) infusions of M2ES at a dose level of 3, 10, and 30mg/kg in rhesus monkeys, the concentration-time curves of M2ES were best fitted to a non-compartment model, and area under the curve (AUC) was positively correlated with the dosage. M2ES had a tendency to accumulate in vivo following successive IV infusions. Serum anti-M2ES IgG antibodies were generated quickly during IV administration, and the antibody level in serum did not significantly decrease after four-week recovery period. Animals administered IV infusions twice weekly (M2ES at 10 or 30mg/kg body weight per day) for 3months developed mild or moderate vacuolation of proximal tubule epithelial cell in proximal convoluted tubule of kidney, but this adverse-effect was reversible. In summary, M2ES was well tolerated and did not cause any serious toxicity. These pre-clinical safety data contribute to the initiation of the ongoing clinical study.


Assuntos
Endostatinas/farmacocinética , Macaca mulatta/metabolismo , Proteínas Recombinantes/farmacocinética , Animais , Área Sob a Curva , Células Epiteliais/efeitos dos fármacos , Feminino , Meia-Vida , Humanos , Infusões Intravenosas , Túbulos Renais Proximais/efeitos dos fármacos , Masculino , Segurança , Toxicocinética
12.
J Nanosci Nanotechnol ; 14(7): 5075-87, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24757983

RESUMO

The present research work describes the formulation of arginine conjugated 3.0G Poly(propylene) imine (PPI) dendrimers, mimicking the surface structure of an endogenous angiogenesis-inhibitor endostatin; for tumor specific delivery of a model anticancer drug, doxorubicin hydrochloride (Dox). Synthesis of PPI dendrimers and conjugation of arginine to surface groups was confirmed by FTIR, NMR, TEM and mass spectrometry. Drug was loaded by equilibrium dialysis method and developed formulation was evaluated for entrapment efficiency, hemolytic toxicity, in vitro drug release, stability, anti-angiogenic activity via in vivo chick embryo chorioallantoic membrane (CAM) assay, and anticancer activity and cell uptake using MCF-7 cancer cell lines. The system exhibited the initial rapid release followed by sustained release of Dox with significant antiangiogenic activity in the CAM assay. Further, the arginine conjugated dendrimers was found to inhibit growth of cancer cells in ex vivo studies with MCF-7 cell lines. Cell uptake studies suggested that in comparison to free drug the formulation was preferably taken up by the tumor cells. Thus the two pronged attack on cancerous tissue i.e., inhibition of angiogenesis and killing of cancer cells by anticancer drug, might prove to be a promising approach in the treatment of fatal disease, cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Dendrímeros/síntese química , Doxorrubicina/administração & dosagem , Endostatinas/administração & dosagem , Nanocápsulas/administração & dosagem , Nanocápsulas/química , Neoplasias Experimentais/tratamento farmacológico , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Linhagem Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacocinética , Endostatinas/química , Endostatinas/farmacocinética , Humanos , Células MCF-7 , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Resultado do Tratamento
13.
Theranostics ; 4(3): 256-66, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24505234

RESUMO

PURPOSE: Targeted radiotherapy (TRT) is an emerging approach for tumor treatment. Previously, 3PRGD2 (a dimeric RGD peptide with 3 PEG4 linkers) has been demonstrated to be of advantage for integrin αvß3 targeting. Given the promising results of (99m)Tc-3PRGD2 for lung cancer detection in human beings, we are encouraged to investigate the radiotherapeutic efficacy of radiolabeled 3PRGD2. The goal of this study was to investigate and optimize the integrin αvß3 mediated therapeutic effect of (177)Lu-3PRGD2 in the animal model. EXPERIMENTAL DESIGN: Biodistribution, gamma imaging and maximum tolerated dose (MTD) studies of (177)Lu-3PRGD2 were performed. The targeted radiotherapy (TRT) with single dose and repeated doses as well as the combined therapy of TRT and the anti-angiogenic therapy (AAT) with Endostar were conducted in U87MG tumor model. The hematoxylin and eosin (H&E) staining and immunochemistry (IHC) were performed post-treatment to evaluate the therapeutic effect. RESULTS: The U87MG tumor uptake of (177)Lu-3PRGD2 was relatively high (6.03 ± 0.65 %ID/g, 4.62 ± 1.44 %ID/g, 3.55 ± 1.08 %ID/g, and 1.22 ± 0.18 %ID/g at 1 h, 4 h, 24 h, and 72 h postinjection, respectively), and the gamma imaging could visualize the tumors clearly. The MTD of (177)Lu-3PRGD2 in nude mice (>111 MBq) was twice to that of (90)Y-3PRGD2 (55.5 MBq). U87MG tumor growth was significantly delayed by (177)Lu-3PRGD2 TRT. Significantly increased anti-tumor effects were observed in the two doses or combined treatment groups. CONCLUSION: The two-dose TRT and combined therapy with Endostar potently enhanced the tumor growth inhibition, but the former does not need to inject daily for weeks, avoiding a lot of unnecessary inconvenience and suffering for patients, which could potentially be rapidly translated into clinical practice in the future.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Complexos de Coordenação/uso terapêutico , Endostatinas/uso terapêutico , Integrina alfaVbeta3/metabolismo , Peptídeos Cíclicos/uso terapêutico , Compostos Radiofarmacêuticos/uso terapêutico , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacocinética , Animais , Linhagem Celular Tumoral , Terapia Combinada , Complexos de Coordenação/farmacocinética , Endostatinas/administração & dosagem , Endostatinas/farmacocinética , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Peptídeos Cíclicos/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Proteínas Recombinantes , Proteína Tumoral 1 Controlada por Tradução , Ensaios Antitumorais Modelo de Xenoenxerto
14.
BMC Cancer ; 13: 479, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24128285

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Endostatinas/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Endostatinas/farmacocinética , Endostatinas/uso terapêutico , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Feminino , Humanos , Pulmão/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico , Análise Serial de Tecidos , Distribuição Tecidual , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Biomaterials ; 34(26): 6261-71, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23714245

RESUMO

Endostatin (ES), a 20 kDa protein derived from the carboxy-terminus of collagen XVIII is a potent angiogenesis inhibitor, but clinical development has been hindered by poor clinical efficacy and insufficient functional information from which to design agents with improved activity. The present study investigated protein uptake by cells as a determinant of ES activity. We developed a cell-permeable ES protein (HM73ES) with enhanced capacity to enter cells by adding a macromolecule transduction domain (MTD). HM73ES inhibited angiogenesis-associated phenotypes in cultured endothelial cells [as assessed by tube formation, wound-healing, cell proliferation and survival assays]. These effects were accompanied by reductions in MAPK signaling (ERK phosphorylation), and in ß-Catenin, c-Myc, STAT3, and VEGF protein expression. The cell-permeable ES displayed greater tissue penetration in mice and suppressed the growth of human tumor xenografts to a significantly greater extent than ES protein without the MTD sequence. Our results suggest that anti-angiogenic activities of native ES are limited at the level of protein uptake and/or subcellular localization, and that much of the activity of ES against tumors depends on one or more intracellular functions. This study will inform future efforts to understand ES function(s) and suggest strategies for improving ES-based cancer therapeutics.


Assuntos
Inibidores da Angiogênese/farmacocinética , Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Endostatinas/farmacocinética , Endostatinas/uso terapêutico , Neoplasias/tratamento farmacológico , Sequência de Aminoácidos , Inibidores da Angiogênese/química , Inibidores da Angiogênese/genética , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Endostatinas/química , Endostatinas/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Células NIH 3T3 , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico
16.
J Pharm Biomed Anal ; 70: 505-11, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22858310

RESUMO

LC-MS/MS is a promising analytical platform for the quantification of recombinant therapeutic proteins in biological fluids for pharmacokinetic (PK) studies. Herein, an absolute quantification method based on LC-MS/MS technique was developed to quantify endostar, which is modified from the recombinant human endostatin by adding a nine-amino acid sequence (MGGSHHHHH) at the N-terminal. A reproducible three-step analytical procedure was adopted: (1) Ni(2+) Sepharose was used to selectively extract endostar; (2) the signature peptide "TEAPSATGQASSLLGGR" (m/z 802.3(2+)-651.8(2+)) of endostar and a synthetic peptide "TEAPSATGQVSSLLGGR" (m/z 816.9(2+)-666.4(2+)) as internal standard (IS) were selected and analyzed in the multiple reaction monitoring (MRM) mode; (3) the proposed method was validated and applied to the pharmacokinetic study of endostar. The lower limit of quantification (LLOQ) for quantifying endostar was 50 ng/ml and this method is linear over 50-10,000 ng/ml. The accuracy was between 85% and 115%, and the intra-batch and inter-batch analytic precision and accuracy were below 15%. This LC-MS/MS approach was validated for the application to the pharmacokinetic study of endostar in rats.


Assuntos
Inibidores da Angiogênese/sangue , Inibidores da Angiogênese/farmacocinética , Cromatografia Líquida , Endostatinas/sangue , Endostatinas/farmacocinética , Espectrometria de Massas em Tandem , Inibidores da Angiogênese/administração & dosagem , Animais , Calibragem , Cromatografia Líquida/normas , Estabilidade de Medicamentos , Endostatinas/administração & dosagem , Feminino , Humanos , Injeções Intravenosas , Limite de Detecção , Modelos Lineares , Masculino , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/sangue , Proteínas Recombinantes/farmacocinética , Padrões de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Sefarose/análogos & derivados , Sefarose/química , Espectrometria de Massas em Tandem/normas
17.
Int J Nanomedicine ; 6: 1535-41, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21845043

RESUMO

BACKGROUND: Particulate carriers are necessary to control the release of endostar and prolong its circulation in vivo. The purpose of this study was to identify a suitable carrier for the capsulation and delivery of endostar. METHODS: We prepared a series of poly (DL-lactide-co-glycolide) (PLGA) and poly (ethylene glycol) (PEG)-modified PLGA (PEG-PLGA) particulate carriers, and then characterized them according to their ability to prolong the circulation of endostar, their physicochemical properties, endostar-loading content, and in vitro and in vivo particulate carrier release profiles. RESULTS: All the particulate carriers had spherical core shell structures. The PEG-PLGA material and nanosize range appeared to enable the carriers to encapsulate more endostar, release endostar faster in vitro, and accumulate more endostar in vivo. The drug loading capacity of PEG-PLGA and PLGA nanoparticles was 8.03% ± 3.41% and 3.27% ± 5.26%, respectively, and for PEG-PLGA and PLGA microspheres was 15.32% ± 5.61% and 9.21% ± 4.73%. The cumulative amount of endostar released from the carriers in phosphate-buffered saline over 21 days was 23.79%, 20.45%, 15.13%, and 10.41%, respectively. Moreover, the terminal elimination half-life of endostar in the rabbit was 26.91 ± 7.93 hours and 9.32 ± 5.53 hours in the PEG-PLGA group and the PLGA nanoparticle group. Peak endostar concentration was reached at day 7 in the group treated with subcutaneous injection of PEG-PLGA microspheres and at day 14 in the group receiving subcutaneous injection of PLGA microspheres. Endostar was detectable in vivo in both groups after injection of the particulate carriers. CONCLUSION: PEG-PLGA nanoparticles might be better than other nanoparticulate carriers for encapsulation and distribution of endostar.


Assuntos
Portadores de Fármacos/química , Endostatinas/administração & dosagem , Nanopartículas/química , Animais , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/farmacocinética , Endostatinas/sangue , Endostatinas/química , Endostatinas/farmacocinética , Ácido Láctico/administração & dosagem , Ácido Láctico/química , Masculino , Microesferas , Nanopartículas/administração & dosagem , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Ácido Poliglicólico/administração & dosagem , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Coelhos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/sangue , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética
18.
Blood ; 117(23): 6392-403, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-21482707

RESUMO

Specific internalization of endostatin into endothelial cells has been proved to be important for its biologic functions. However, the mechanism of endostatin internalization still remains elusive. In this study, we report for the first time that both caveolae/lipid rafts and clathrin-coated pits are involved in endostatin internalization. Inhibition of either the caveolae pathway or the clathrin pathway with the use of chemical inhibitors, small interfering RNAs, or dominant-negative mutants alters endostatin internalization in vitro. Intriguingly, cholesterol sequestration by nystatin, a polyene antifungal drug, significantly enhances endostatin uptake by endothelial cells through switching endostatin internalization predominantly to the clathrin-mediated pathway. Nystatin-enhanced internalization of endostatin also increases its inhibitory effects on endothelial cell tube formation and migration. More importantly, combined treatment with nystatin and endostatin selectively enhances endostatin uptake and biodistribution in tumor blood vessels and tumor tissues but not in normal tissues of tumor-bearing mice, ultimately resulting in elevated antiangiogenic and antitumor efficacies of endostatin in vivo. Taken together, our data show a novel mechanism of endostatin internalization and support the potential application of enhancing the uptake and therapeutic efficacy of endostatin via regulating distinct endocytic pathways with cholesterol-sequestering agents.


Assuntos
Antifúngicos/farmacocinética , Antineoplásicos/farmacocinética , Colesterol/metabolismo , Endocitose/efeitos dos fármacos , Endostatinas/farmacocinética , Endotélio Vascular/metabolismo , Nistatina/farmacocinética , Animais , Antifúngicos/agonistas , Antifúngicos/farmacologia , Antineoplásicos/agonistas , Antineoplásicos/farmacologia , Cavéolas/metabolismo , Linhagem Celular Tumoral , Vesículas Revestidas por Clatrina/metabolismo , Sinergismo Farmacológico , Endostatinas/agonistas , Endostatinas/farmacologia , Endotélio Vascular/patologia , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Nistatina/agonistas , Nistatina/farmacologia
19.
Anticancer Drugs ; 22(5): 409-15, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21427563

RESUMO

Many antitumor drugs have a limited ability to penetrate more than a few cell diameters from blood vessels into solid tumors, which limits their effectiveness. In this study, we investigated whether the biological activity of endostatin can be enhanced by the addition of an integrin-targeting and permeability-enhancing sequence. The internalization RGD (CRGDKGPDC; iRGD) sequence was added at the carboxyl terminus of endostatin. Modification of endostatin with the iRGD motif showed specific and increased binding to endothelial cells; the increased binding correlated with an improved antiangiogenic property. iRGD-modified endostatin was more effective than human endostatin in inhibiting liver cancer growth in athymic mice. The finding indicates that addition of a vascular targeting and permeability sequence can enhance the biological activity of an antiangiogenic molecule and tumor targeting.


Assuntos
Endostatinas/administração & dosagem , Endostatinas/farmacocinética , Oligopeptídeos/administração & dosagem , Oligopeptídeos/farmacocinética , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacocinética , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Sequência de Bases , Permeabilidade Capilar , Bovinos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Peptídeos Penetradores de Células/administração & dosagem , Peptídeos Penetradores de Células/genética , Peptídeos Penetradores de Células/farmacocinética , Embrião de Galinha , Endostatinas/genética , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Humanos , Integrinas/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Oligopeptídeos/genética , Ligação Proteica , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética
20.
Int J Nanomedicine ; 5: 1039-48, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21170352

RESUMO

Endostar, a novel recombinant human endostatin, which was approved by the Chinese State Food and Drug Administration in 2005, has a broad spectrum of activity against solid tumors. In this study, we aimed to determine whether the anticancer effect of Endostar is increased by using a nanocarrier system. It is expected that the prolonged circulation of endostar will improve its anticancer activity. Endostar-loaded nanoparticles were prepared to improve controlled release of the drug in mice and rabbits, as well as its anticancer effects in mice with colon cancer. A protein release system could be exploited to act as a drug carrier. Nanoparticles were formulated from poly (ethylene glycol) modified poly (DL-lactide-co-glycolide) (PEG-PLGA) by a double emulsion technique. Physical and release characteristics of endostar-loaded nanoparticles in vitro were evaluated by transmission electron microscopy (TEM), photon correlation spectroscopy (PCS), and micro bicinchoninic acid protein assay. The pharmacokinetic parameters of endostar nanoparticles in rabbit and mice plasma were measured by enzyme-linked immunosorbent assay. Western blot was used to detect endostatin in different tissues. To study the effects of endostar-loaded nanoparticles in vivo, nude mice in which tumor cells HT-29 were implanted, were subsequently treated with endostar or endostar-loaded PEG-PLGA nanoparticles. Using TEM and PCS, endostar-loaded PEG-PLGA nanoparticles were found to have a spherical core-shell structure with a diameter of 169.56 ± 35.03 nm. Drug-loading capacity was 8.22% ± 2.35% and drug encapsulation was 80.17% ± 7.83%. Compared with endostar, endostar-loaded PEG-PLGA nanoparticles had a longer elimination half-life and lower peak concentration, caused slower growth of tumor cell xenografts, and prolonged tumor doubling times. The nanoparticles changed the pharmacokinetic characteristics of endostar in mice and rabbits, thereby reinforcing anticancer activity. In conclusion, PEG-PLGA nanoparticles are a feasible carrier for endostar. Endostar-loaded PEG-PLGA nanoparticles seem to have a better anticancer effect than conventional endostar. We believe that PEG-PLGA nanoparticles are an effective carrier for protein medicines.


Assuntos
Sistemas de Liberação de Medicamentos , Endostatinas/química , Endostatinas/farmacologia , Nanopartículas/química , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Poliglactina 910/química , Poliglactina 910/farmacologia , Análise de Variância , Animais , Proliferação de Células/efeitos dos fármacos , Endostatinas/sangue , Endostatinas/farmacocinética , Ensaio de Imunoadsorção Enzimática , Células HT29 , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Eletrônica de Transmissão , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Tamanho da Partícula , Polietilenoglicóis/farmacocinética , Poliglactina 910/farmacocinética , Coelhos , Proteínas Recombinantes/sangue , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...