Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Mater Chem B ; 9(42): 8768-8778, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34585713

RESUMO

HER2+ breast cancer is highly aggressive and proliferative even after multiple chemotherapy regimens. At present, the available clinical treatment duration of chemotherapeutic agents is limited by severe toxicity to noncancerous tissues, which are attributed to insufficient targeting. Here, we designed an active-targeted and pH-responsive liposome to improve the treatment. The ideas were as follows: (1) using liposome as a nano-delivery system for HER2 inhibitor (lapatinib; LAP) to reduce the toxicity; (2) modifying the capsule with T7 peptide for specific targeted delivery to the tumor cells, and (3) enabling the capsule with the pH-sensitive ability and triggering sustained drug release at extracellular weakly acidic microenvironment to emerge toxicity in tumors and to improve curative effects. It was found that T7 peptide-modified pH-sensitive liposome (T7-LP) was more effective and safer than free drug and unmodified liposome, and reduced drug-induced side effects and noncancerous toxicity. These results support the application potential of T7-LP in improving the efficacy of LAP in HER2+ breast cancer treatment. It might be a novel LAP formulation as a clinical agent.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Colágeno Tipo IV/química , Lapatinib/farmacologia , Fragmentos de Peptídeos/química , Inibidores de Proteínas Quinases/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Animais , Antineoplásicos/química , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Lapatinib/química , Lipossomos/química , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Estrutura Molecular , Imagem Óptica , Inibidores de Proteínas Quinases/química , Ratos , Ratos Sprague-Dawley , Receptor ErbB-2/metabolismo
2.
Int J Biol Macromol ; 184: 101-108, 2021 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-34119545

RESUMO

Lentinan is a natural ß-glucan with various bioactivities and is combined with chemotherapy drugs for cancer treatment. Regorafenib is an oral multi-kinase inhibitor approved by FDA for treatment of metastatic colorectal cancer, advanced hepatocellular carcinoma, and metastatic gastrointestinal stromal tumors. Regorafenib has poor water solubility and multiple toxicities. We report drug-drug nanosuspensions of regorafenib and lentinan. Results of dynamic light scattering and scanning electron microscopy showed that the mean particle size of the regorafenib-lentinan nanosuspensions was approximately 200 nm and was uniformly distributed. Transmission electron microscopy findings indicated that lentinan stabilized the nanosuspensions by steric manner. Hydrogen bonds and hydrophobic interactions were found between regorafenib and lentinan by molecular dynamics simulation. The results of cytotoxicity assay and pharmacokinetics study in rats showed that the regorafenib-lentinan nanosuspensions reduced the toxicity and enhanced the in vitro anticancer activity and oral bioavailability of regorafenib. Lentinan as a natural stabilizer has the potential using for drug nanosuspensions. Drug-drug nanosuspensions are a new form of combination therapies that can reduce the number of drugs taken by patients and improve their compliance.


Assuntos
Antineoplásicos/administração & dosagem , Lentinano/administração & dosagem , Compostos de Fenilureia/administração & dosagem , Piridinas/administração & dosagem , Administração Oral , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Disponibilidade Biológica , Composição de Medicamentos , Células HCT116 , Células HEK293 , Humanos , Lentinano/química , Lentinano/farmacocinética , Simulação de Dinâmica Molecular , Nanopartículas , Tamanho da Partícula , Compostos de Fenilureia/química , Compostos de Fenilureia/farmacocinética , Piridinas/química , Piridinas/farmacocinética , Ratos , Ratos Sprague-Dawley , Solubilidade , Suspensões
3.
Carbohydr Polym ; 229: 115476, 2020 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-31826488

RESUMO

The objective of present study is to explore whether polysaccharide could be a crystal growth inhibitor in poorly soluble antitumor drug Ibrutinib (IBR) formulation. In this work, small molecular ligands (amino or organic acids) in co-amorphous system (CAS) were preliminarily screened. A polysaccharide, microcrystalline cellulose (MCC) was selected to stabilize amorphous drug and enhance pharmacokinetic properties. Fourier-transform infrared, Raman, and X-ray photoelectron spectroscopy confirmed the ionic interaction of the ternary IBR formulation. Moreover, the biosafety of the ternary formulation was the same as that of IBR while the in vitro performance advantage of the ternary formulation was converted into higher bioavailability in vivo experiments. Overall, MCC as an effective crystal growth inhibitor in the novel ternary IBR formulation is a promising approach to improve the dissolution rate of crystalline drugs and the stability of amorphous drugs, as well as providing a theoretical basis for clinical applications.


Assuntos
Celulose/química , Pirazóis/química , Pirimidinas/química , Adenina/análogos & derivados , Animais , Disponibilidade Biológica , Linhagem Celular Tumoral , Fenômenos Químicos , Cristalização , Composição de Medicamentos , Humanos , Masculino , Piperidinas , Pirazóis/farmacocinética , Pirazóis/farmacologia , Pirimidinas/farmacocinética , Pirimidinas/farmacologia , Ratos
4.
Mater Sci Eng C Mater Biol Appl ; 105: 110012, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31546459

RESUMO

Telaprevir (TVR) is typically a poorly soluble drug with an extremely low bioavailability of 1.7%. Polymorph modifications cannot improve the solubility of TVR because it only has a single unsolvated crystalline form. Co-crystals also provide limited bioavailability enhancement for TVR. Thus, in this study, we increased the solubility and dissolution rate of TVR through formulations of TVR-polymer solid dispersions. Three solid dispersions of TVR were successfully prepared by co-milling with polyvinylpyrrolidone K30 (PVP), polyethylene glycol 6000, and hydroxypropyl methylcellulose (HPMC), which were characterized by different techniques. According to X-ray powder diffraction and differential scanning calorimetry results, TVR presented in amorphous form in all solid dispersions. The fourier transform infrared spectra results indicated that TVR may connect with polymers through the N-H···O or O-H···O hydrogen bonds, which were verified by molecular docking. TVR-PVP and TVR-HPMC displayed a good stability at conventional RH levels, and their thermostabilities were better than those of milled TVR. Among the three solid dispersions, TVR-HPMC showed significant solubility and dissolution rate advantages in different media. Moreover, TVR-HPMC displayed the same anticancer efficacy with crystalline TVR and presented no toxic side effects to normal liver cells. Thus, TVR-HPMC showed potential application value.


Assuntos
Fenômenos Químicos , Composição de Medicamentos/métodos , Oligopeptídeos/farmacologia , Varredura Diferencial de Calorimetria , Morte Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Conformação Molecular , Simulação de Acoplamento Molecular , Oligopeptídeos/química , Polímeros/química , Solubilidade , Espectroscopia de Infravermelho com Transformada de Fourier , Difração de Raios X
5.
Int J Biol Macromol ; 140: 709-718, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31445155

RESUMO

Nanoscale metal-organic frameworks (NMOFs) have attracted considerable attention for controlled drug delivery. However, the interaction between nanoparticles and the biological macromolecules of physiological system must be valued because the formed protein corona will endow NMOFs with new biorecognition properties. In this study, we carried out detailed protein adsorption studies in vitro and cell uptake tests of HeLa cells for nanospherical Uio66 and nanooctahedral Uio67. Uio67 with higher binding constants to human serum albumin needed to combine more protein molecules to achieve colloidal stability state than that needed by Uio66, and this phenomenon led Uio67 to aggregate under the same incubation condition due to the formation of a single-layer protein. Uio67 also induced an evident conformation change in protein to stabilize the combination. In particular, the cell uptake efficiencies of the two systems showed a significant thickness dependence on the protein corona. When samples incubated in 10% fetal bovine serum (FBS), the intracellular rate was the highest for both systems, but the rate was not proportional to the FBS concentration. Results of this work are important to the development of the considerable potential NMOFs-based medicals and also provide additional insight into protein corona.


Assuntos
Comunicação Celular/fisiologia , Estruturas Metalorgânicas/química , Nanopartículas/química , Coroa de Proteína/química , Coroa de Proteína/metabolismo , Adsorção , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Células HeLa , Humanos , Tamanho da Partícula , Ligação Proteica , Albumina Sérica Humana/química , Albumina Sérica Humana/metabolismo
6.
Mol Pharm ; 15(12): 5637-5645, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30422663

RESUMO

Dabrafenib is a novel targeted antimelanoma drug. The present work explored the binding mechanism of dabrafenib-human serum albumin (HSA) and the effect on the esterase-like activity and antioxidant activity of HSA by using 19F NMR, spectroscopy methods, and molecular dynamics simulation. The results of 19F NMR, fluorescence, and time-resolved fluorescence spectroscopy revealed that dabrafenib spontaneously binds to the subdomain IIIA of the HSA by hydrophobic action and forms a static complex. The binding affects the esterase-like activity of HSA but not its antioxidant activity. According to the results of molecular dynamics simulation, dabrafenib interacts with Arg410 and Tyr411, which are the key residue associated with the esterase-like activity of HSA. Meanwhile, dabrafenib does not interact with Cys34, the key residue associated with the antioxidant activity of HSA. The results of circular dichroism spectroscopy and molecular dynamics simulation show that the conformation of HSA is not affected by the binding of dabrafenib. This study can provide useful information for understanding the pharmacokinetic properties of dabrafenib.


Assuntos
Antineoplásicos/farmacocinética , Esterases/metabolismo , Imidazóis/farmacocinética , Oximas/farmacocinética , Albumina Sérica Humana/metabolismo , Antineoplásicos/química , Sítios de Ligação , Dicroísmo Circular , Cisteína/metabolismo , Esterases/química , Interações Hidrofóbicas e Hidrofílicas , Imidazóis/química , Simulação de Dinâmica Molecular , Oximas/química , Ligação Proteica , Albumina Sérica Humana/química , Espectrometria de Fluorescência
7.
Cancer Biol Ther ; 19(11): 1015-1022, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29799306

RESUMO

ABSTRACT The PI3K/mTOR pathway is one of the most frequently aberrantly activated pathways in human malignancies, such as renal cell carcinoma (RCC), and contributes to resistance to antitumor therapies. Thus, PI3K/mTOR is an attractive target for the development of antitumor agents. In this study, we evaluated the preclinical effects of a novel inhibitor SN202. We examined Akt/mTOR activities in renal cancer cells after SN202 treatment. The preclinical effects of SN202 on tumor growth were evaluated in renal cancer cells in vitro and in murine xenografts in vivo. SN202 inhibits PI3Kα, PI3Kγ, and mTOR, the corresponding IC50 values were 3.2, 3.3, and 1.2 nM, respectively. In A498, 786-0, and ACHN renal cancer cell lines, SN202 inhibits cell proliferation in a dose-dependent manner and significantly inhibits 786-0 cell growth. Western blot analysis revealed that SN202 decreases the phosphorylation of PI3K downstream signaling molecules, Akt and S6K, in 786-0 renal cancer cells. Furthermore, oral administration of SN202 results in significant inhibition in human renal carcinoma xenografts in nude mice and favourable pharmacokinetic properties in rats. These results suggest that SN202 might be a promising therapeutic agent against RCC as a dual PI3K/mTOR inhibitor.

8.
RSC Adv ; 8(9): 4742-4749, 2018 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-35539509

RESUMO

Trametinib is a novel anticancer drug for treating metastatic cutaneous melanoma. The present study probed into the binding of trametinib to human serum albumin (HSA) through spectroscopy methods and molecular simulations. Trametinib could quench the fluorescence of HSA through static quenching which could be probed via fluorescence spectroscopy and time-resolved fluorescence. Thermodynamic parameters and docking results indicated that hydrogen bonds and van der Waals forces play crucial roles in this binding process, which exerts almost no effect on the HSA conformation under synchronous fluorescence, three-dimensional fluorescence, circular dichroism spectra, and molecular dynamics simulations. Site marker displacement experiments and molecular docking reveal that trametinib primarily binds to Sudlow site I of HSA. In addition, the trametinib-HSA interaction was hardly influenced by varying amino acid (glutamine, alanine, glycine, and valine) concentrations. This study can provide useful information for the pharmacokinetic properties of trametinib.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA