Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Nanobiotechnology ; 22(1): 90, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38439048

RESUMO

Immune checkpoint inhibitor (ICI)-derived evolution offers a versatile means of developing novel immunotherapies that targets programmed death-ligand 1 (PD-L1)/programmed death-1 (PD-1) axis. However, one major challenge is T cell exhaustion, which contributes to low response rates in "cold" tumors. Herein, we introduce a fluorinated assembly system of LFNPs/siTOX complexes consisting of fluorinated EGCG (FEGCG), fluorinated aminolauric acid (LA), and fluorinated polyethylene glycol (PEG) to efficiently deliver small interfering RNA anti-TOX (thymus high mobility group box protein, TOX) for synergistic tumor cells and exhausted T cells regulation. Using a microfluidic approach, a library of LFNPs/siTOX complexes were prepared by altering the placement of the hydrophobe (LA), the surface PEGylation density, and the siTOX ratio. Among the different formulations tested, the lead formulation, LFNPs3-3/siTOX complexes, demonstrated enhanced siRNA complexation, sensitive drug release, improved stability and delivery efficacy, and acceptable biosafety. Upon administration by the intravenous injection, this formulation was able to evoke a robust immune response by inhibiting PD-L1 expression and mitigating T cell exhaustion. Overall, this study provides valuable insights into the fluorinated assembly and concomitant optimization of the EGCG-based delivery system. Furthermore, it offers a promising strategy for cancer immunotherapy, highlighting its potential in improving response rates in ''cold'' tumors.


Assuntos
Nanopartículas , Neoplasias , Linfócitos T , Antígeno B7-H1 , Ligantes , Microfluídica , Imunoterapia , Neoplasias/tratamento farmacológico
2.
ACS Nano ; 17(8): 7498-7510, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37011376

RESUMO

Classical chemotherapeutic drugs may cause immunogenic cell death (ICD), followed by activating CD8+ T cells to promote cell-mediated antitumor immune responses. However, CD8+ T cells become exhausted due to tumor antigens' continuous stimulation, creating a major obstacle to effectively suppressing tumor growth and metastasis. Here, we develop an approach of chemo-gene combinational nanomedicine to bridge and reprogram chemotherapy and immunotherapy. The dually loaded nanomedicine induces ICD in tumor cells through doxorubicin and reverses the antitumor effects of exhausted CD8+ T cells through the small interfering RNA. The synergistic chemo-gene and fluorine assembly nanomedicine enriched in reactive oxygen species and acid-sensitive bonds results in enhanced cancer immunotherapy to inhibit tumor growth and the lung metastasis of breast cancer in a mouse model of breast cancer and melanoma. This study provides an efficient strategy and insights into chemoimmunological cascade therapy for combating malignant metastatic tumors.


Assuntos
Flúor , Neoplasias , Camundongos , Animais , Nanomedicina/métodos , Linfócitos T CD8-Positivos , Neoplasias/tratamento farmacológico , Doxorrubicina/química , Imunoterapia/métodos , Linhagem Celular Tumoral , Microambiente Tumoral
3.
Adv Mater ; 34(38): e2203019, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35918816

RESUMO

Chemoimmunotherapy has shown great potential to activate an immune response, but the immunosuppressive microenvironment associated with T cell exhaustion remains a challenge in cancer therapy. The proper immune-modulatory strategy to provoke a robust immune response is to simultaneously regulate T-cell exhaustion and infiltration. Here, a new kind of carrier-free nanoparticle is developed to simultaneously deliver chemotherapeutic drug (doxorubicin, DOX), cytolytic peptide (melittin, MPI), and anti-TOX small interfering RNA (thymocyte selection-associated high mobility group box protein, TOX) using a fluorinated prodrug strategy. In this way, the enhanced immunogenic cell death (ICD) induced by the combination of DOX and MPI can act as "offense" signaling to increase CD8+ T-cell infiltration, while the decreased TOX expression interfered with siTOX can serve as "defense" signaling to mitigate CD8+ T-cell exhaustion. As a result, the integration of DOX, MPI, and siTOX in such a bifunctional system produced a potent antitumor immune response in liver cancer and metastasis, making it a promising delivery platform and effective strategy for converting "cold" tumors into "hot" ones.


Assuntos
Neoplasias Hepáticas , Neoplasias , Pró-Fármacos , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Humanos , Imunoterapia , Meliteno/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Pró-Fármacos/farmacologia , RNA Interferente Pequeno/genética , Microambiente Tumoral
4.
Biomaterials ; 284: 121492, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35395456

RESUMO

Hepatic stellate cell (HSC)-targeted delivery is an attractive strategy for liver fibrosis therapy, but the efficacy is hampered by poor delivery of nanomaterials and complicated microenvironments of the fibrotic liver. Here, we report a versatile CXCR4-inhibiting nanocomplex composed of polymeric CXCR4 antagonism (PAMD, PA), CLD (clodronate) and siPAI-1 (siRNA of plasminogen activator inhibitor-1) that surmounts multiple barriers to improve the outcome by co-regulating Kupffer cells (KCs), extracellular matrix (ECM) and HSCs. Upon encountering biological barriers, the nanocomplex exerted penetrating and targeting functions, efficiently overcoming KCs capture, ECM trapping and nonspecific recognition of HSCs, finally contributing to the enhanced HSCs uptake. Moreover, an enlarged antifibrotic activity is realized through synergetic regulation of KCs apoptosis, ECM degradation and HSCs inactivation. Overall, such a versatile nanocomplex provides a framework for designing HSC-targeted delivery system and has valuable potential as a novel antifibrotic strategy.


Assuntos
Células Estreladas do Fígado , Células de Kupffer , Matriz Extracelular/metabolismo , Fibrose , Células Estreladas do Fígado/patologia , Humanos , Células de Kupffer/metabolismo , Fígado/metabolismo , Cirrose Hepática/patologia , Receptores CXCR4/metabolismo
5.
Adv Sci (Weinh) ; 9(15): e2105894, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35486032

RESUMO

Nano-based immunotherapy of therapeutic biomolecules is attractive but tremendously hampered by the poor delivery efficiency. This study reports a novel delivery system of fluorinated-coordinative-epigallocatechin gallate (EGCG), referring as FEGCG/Zn, through the integration of fluorination and zinc ions (Zn2+ ) into EGCG. The robust therapeutics of FEGCG/Zn are measured in terms of the regulating effect on programmed cell death ligand 1 (PD-L1), the effective delivery of diverse biomolecules, and the hitchhiking ability using living cells. Taking small interfering RNA of PD-L1 (siPD-L1) and erythrocytes as an example, the fabricated biomimetic system achieves excellent siPD-L1 delivery and further improves siPD-L1 accumulation in tumors. Finally, the combination of FEGCG/Zn and siPD-L1 promotes antitumor immunotherapy through alleviation of T cells exhaustion by regulating PD-L1 expression in tumor cells. The results demonstrate that FEGCG/Zn substantially regulates PD-L1 expression and improves immune-biomolecule delivery by forming biomimetic nanoassemblies, offering a versatile platform for cancer immunotherapy.


Assuntos
Antígeno B7-H1 , Neoplasias , Antígeno B7-H1/genética , Biomimética , Catequina/análogos & derivados , Linhagem Celular Tumoral , Fatores Imunológicos , Imunoterapia , Neoplasias/tratamento farmacológico
6.
J Nanobiotechnology ; 19(1): 213, 2021 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-34275480

RESUMO

BACKGROUND: RBC membrane derived nanoparticles (NPs) represent an emerging platform with prolonged circulation capacity for the delivery of active substances. For functionalize derived RBCs NPs, various strategies, such as biomimetic rebuilding of RBCs, chemical modification or inserting ligands, have been carried out to improve their performance. However, one potential adverse effect for these methods is the structural failure of membrane proteins, consequently affecting its original immune escape function. RESULTS: In this study, we reported a green technology of "disassembly-reassembly" to prepare biomimetic reconstituted RBCs membrane (rRBCs) by separating the endogenous proteins and lipids from nature RBC membrane. IR780 iodide was used as a pattern drug to verify the property and feasibility of rRBCs by constructing IR780@rRBC NPs with IR780@RBC NPs and free IR780 as controls. The results demonstrated the superiority of IR780@rRBC NPs in toxicity, stability, pharmacokinetics and pharmacodynamics compared with IR780@rRBC and free IR780. CONCLUSIONS: The reported "disassembly-reassembly" strategy shows great potential to produce controllable and versatile rRBC membrane-inspired delivery platform, which may be used to overcome the deficiency of functionalization in cell membrane coated nanoparticles .


Assuntos
Biomimética/métodos , Membrana Eritrocítica/química , Terapia Fototérmica/métodos , Animais , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Eritrócitos , Indóis , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Células RAW 264.7
7.
Biomaterials ; 261: 120311, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32911091

RESUMO

The treatment options of liver fibrosis remain limited except for liver transplantation due to the complexity and slow development in its progression. Besides, liver fibrosis recurrence and intervention time have not been reported as significant indicators to affect the anti-fibrotic efficacy of tested drugs/strategies. Herein, a novel fluoropolymer is developed to achieve high drug loading of sorafenib and efficient delivery of miR155 inhibitor (anti-miR155) for dual-targeting of hepatic stellate cells (HSCs) and kupffer cells (KCs), and we report a detailed plan on the design of treatment regimen to reveal the relationship between chemogene therapy, intervention time and fibrosis recurrence. Such a combined chemo-gene therapy of sorafenib and anti-miR155 can achieve superior therapeutic efficiency by polarizing the pro-inflammatory M1 to anti-inflammatory M2 of KCs and inhibiting the proliferation of HSCs. Importantly, efficacy and recurrence prevention of chemogene therapy earlier in the liver fibrosis will be more effective than the treatment at later stage. In conclusion, this work proposes a novel strategy to improve the efficacy and prevent recurrence of liver fibrosis by dual-regulating of KCs and HSCs, and emphasizes the importance of therapy earlier in the treatment of liver fibrosis.


Assuntos
Nanopartículas , Preparações Farmacêuticas , Fibrose , Terapia Genética , Humanos , Fígado/patologia , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/patologia
8.
Bioconjug Chem ; 31(3): 698-707, 2020 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-31967460

RESUMO

Polyethylenimine (PEI) is a promising delivery vector of nucleic acids, but cytotoxicity and only moderate transfection efficacy with small RNAs limit its applications. Here we hypothesized that hydrophobization of PEI by combined modification with perfluorinated moieties (F) and cholesterol (Ch) will help in addressing both the cytotoxicity and siRNA delivery efficacy. To test the hypothesis, we synthesized a series of copolymers (F-PEI-Ch) by modifying PEI by reaction with heptafluorobutyric anhydride and cholesteryl chloroformate. We investigated and compared the effect of the modifications on siRNA delivery in vitro and in vivo. We found that the F-PEI-Ch copolymers assembled into micellar structures and that the copolymer with the highest Ch content exhibited the best siRNA delivery performance, including lower cytotoxicity, enhanced cell uptake, improved endosomal escape, and the best siRNA silencing efficacy in vitro and in vivo when compared with control PEI, F-PEI, and PEI-Ch. Overall, hydrophobization of PEI with a combination of cholesterol and superhydrophobic perfluorinated moieties represents a promising approach to the design of siRNA delivery vectors with decreased toxicity and enhanced transfection efficacy.


Assuntos
Colesterol/química , Portadores de Fármacos/química , Fluorocarbonos/química , Interações Hidrofóbicas e Hidrofílicas , Polietilenoimina/química , RNA Interferente Pequeno/química , Animais , Linhagem Celular Tumoral , Inativação Gênica , Camundongos , RNA Interferente Pequeno/genética
9.
ACS Appl Mater Interfaces ; 11(23): 20689-20698, 2019 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-31117451

RESUMO

Inefficient transfection of biocompatible low-molecular-weight (LMW) polycations, such as 1.8k polyethylenimine (PEI), is a major challenge for successful nucleic acid delivery. Current strategies to improve transfection efficiency are bottlenecked by maintaining the balance between efficient gene encapsulation and on-demand cargo release. Here, we developed a new class of Zn(II)-coordinated micelles, which showed tight small interfering RNA (siRNA) binding and pH-switchable release. The dipicolylamine-modified PEI 1.8k (PD) and dopamine-conjugated cholesterol (Chol-Dopa) assemble into coordinative micelles (Zn-PD/Chol-Dopa) via the coordination of 2,2'-dipicolylamine (DPA) and Dopa through Zn(II) as a bridge. The high phosphate-binding affinity of Zn-DPA enhanced the siRNA packaging and the interaction between cholesterol and cell membranes enhanced cellular uptake. Moreover, the coordination effect weakened in the acidic environment of lyso/endosome, triggering the disassembly of micelles and siRNA release. These properties of the micelles resulted in strong siRNA transfection efficiencies in various cell lines. Our strategy of constructing coordinative micelles improves the transfection efficiency of LMW PEI and holds tremendous potential to develop the endogenous responsive gene delivery systems.


Assuntos
Poliaminas/química , Polietilenoimina/química , Transfecção/métodos , Zinco/química , Aminas/química , Animais , Células CHO , Colesterol/química , Cricetulus , Inativação Gênica , Concentração de Íons de Hidrogênio , Micelas , Ácidos Picolínicos/química , Polieletrólitos , RNA Interferente Pequeno/química
10.
Int J Nanomedicine ; 14: 2927-2944, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31118614

RESUMO

Background: Liver fibrosis is a chronic liver disease associated with an excessive accumulation of extracellualr matrix (ECM) proteins which ultimately lead to cirrohosis and hepatocellular carcinoma. Purpose: Liver fibrosis therapies that use combination approaches with the ability to affect multiple disease pathways have proven higher efficacies. This study aimed at optimizing and characterizing the co-encapsulation of pirfenidone (PF) and AMD3100 (AMD) into CXCR4-targeted combination liposomes (CTC liposome) for CXCR4 targeting, and the inhibition of major molecular culprits ie α-SMA, CXCR4, TGFß, and P-p38 involved in liver fibrosis in-vitro. Methods: The CTC liposomes were prepared using the thin-film hydration method. The concentration of encapsulated AMD and PF was measured by HPLC and UV spectrophotometry, respectively. Tramsmission electron microscopy (TEM) was used to determine the liposomal morphology. The CXCR4 targeting ability was determined by CXCR4 redistribution assay. Confocal microscopy and flowcytometry were used to determine the CXCR4 mediated cell uptake. The apoptosis inducing and protein downreguating ability of CTC liposomes were determined by apoptosis assay and western blot analysis, respectively. In-vivo biodistribution and Hoechst staining were used to confirm the feasibility of CTC liposome for the in-vivo applications and drug targeted accumulation, respectively. Results: The TEM studies revealed that CTC liposomes were spherical in shape. The cumulative release of AMD and PF from CTC liposome was 67% and 84%, respectively, at 48 h. Compared to the free drug counterparts, encapsulated drugs displayed higher cell viability. The CXCR4 redistribution assay confirmed the CXCR4 targeting and antagonistic ability of CTC liposomes. The CTC liposomes were internalized more effectively via caveolae-mediated endocytic pathways. CTC liposomes displayed aggressive apoptosis (87.3%) in TGFß-induced activated HSC-T6 cells suggesting a propensity to fibrosis regression. Also, CTC liposomes significantly reduced α-SMA (65%), CXCR4 (77%), TGFß (89%), and P-p38 (66%) expressions, better than free drugs. CTC@IR780 liposomes (CTC liposomes incorporating IR780 dye) were more accumulated in fibrotic livers compared to free IR780, as judged by in-vivo imaging, biodistribution analysis, and Hoechst staining. These findings suggest that this simple and stable CTC liposomal system holds a great promise for the treatment and prevention of liver fibrosis.


Assuntos
Sistemas de Liberação de Medicamentos , Células Estreladas do Fígado/patologia , Compostos Heterocíclicos/administração & dosagem , Cirrose Hepática/tratamento farmacológico , Piridonas/administração & dosagem , Receptores CXCR4/metabolismo , Animais , Apoptose/efeitos dos fármacos , Benzilaminas , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclamos , Endocitose/efeitos dos fármacos , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Compostos Heterocíclicos/farmacologia , Compostos Heterocíclicos/uso terapêutico , Humanos , Lipossomos , Camundongos , Piridonas/farmacologia , Piridonas/uso terapêutico , Ratos , Receptores CXCR4/antagonistas & inibidores , Transdução de Sinais , Distribuição Tecidual/efeitos dos fármacos , Fator de Crescimento Transformador beta , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Macromol Biosci ; 18(11): e1800234, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30259651

RESUMO

Chemokine receptor CXC receptor 4 (CXCR4) plays a crucial role in cell invasion and metastasis of multiple types of cancer. Dual-function polymeric CXCR4 antagonists based on cyclam-modified poly(ethylenimine) (C-PEI) have been shown to have potential as nucleic acid delivery vectors and antimetastatic therapeutics in recent studies. How cholesterol modification of C-PEI affects the ability of the polycation to deliver siRNA and inhibit CXCR4 is tested here. It is shown that the C-PEI with the lower content of cholesterol exhibits the highest siRNA transfection efficiency and demonstrates enhanced CXCR4 antagonism and antimetastatic activity in a breast cancer model in vivo. Overall, the cholesterol modification of C-PEI is a viable strategy to achieve efficient delivery of siRNA and simultaneous CXCR4 inhibition for combined antimetastatic therapies is validated by this study.


Assuntos
Portadores de Fármacos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Proteínas de Neoplasias/antagonistas & inibidores , Polietilenoimina , RNA Interferente Pequeno , Receptores CXCR4/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Feminino , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Polietilenoimina/química , Polietilenoimina/farmacologia , RNA Interferente Pequeno/química , RNA Interferente Pequeno/farmacologia , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
12.
ACS Nano ; 12(7): 6620-6636, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29985577

RESUMO

Metastatic breast cancer is a major cause of cancer-related female mortality worldwide. The signal transducer and activator of transcription 3 (STAT3) and the chemokine receptor CXCR4 are involved in the metastatic spread of breast cancer. The goal of this study was to develop nanomedicine treatment based on combined inhibition of STAT3 and CXCR4. We synthesized a library of CXCR4-inhibiting polymers with a combination of beneficial features that included PEGylation, fluorination, and bioreducibility to achieve systemic delivery of siRNA to silence STAT3 expression in the tumors. An in vivo structure-activity relationship study in an experimental lung metastasis model revealed superior antimetastatic activity of bioreducible fluorinated polyplexes when compared with nonreducible controls despite similar CXCR4 antagonism and the ability to inhibit in vitro cancer cell invasion. When compared with nonreducible and nonfluorinated polyplexes, improved siRNA delivery was observed with the bioreducible fluorinated polyplexes. The improvement was ascribed to a combination of enhanced physical stability, decreased serum destabilization, and improved intracellular trafficking. Pharmacokinetic analysis showed that fluorination decreased the rate of renal clearance of the polyplexes and contributed to enhanced accumulation in the tumors. Therapeutic efficacy of the polyplexes with STAT3 siRNA was assessed in early stage breast cancer and late-stage metastatic breast cancer with primary tumor resection. Strong inhibition of the primary tumor growth and pronounced antimetastatic effects were observed in both models of metastatic breast cancer. Mechanistic studies revealed multifaceted mechanism of action of the combined STAT3 and CXCR4 inhibition by the developed polyplexes relying both on local and systemic effects.


Assuntos
Neoplasias da Mama/terapia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Nanopartículas/uso terapêutico , Poliaminas/uso terapêutico , RNA Interferente Pequeno/uso terapêutico , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Halogenação , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Nanomedicina , Nanopartículas/química , Poliaminas/química , Polieletrólitos , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Terapêutica com RNAi , Receptores CXCR4/antagonistas & inibidores , Fator de Transcrição STAT3/genética
13.
Nanomedicine ; 14(6): 1765-1776, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29777878

RESUMO

This report describes the development of polyplexes based on CXCR4-inhibiting poly(ethylenimine) derivative (PEI-C) for pulmonary delivery of siRNA to silence plasminogen activator inhibitor-1 (siPAI-1) as a new combination treatment of pulmonary fibrosis (PF). Safety and delivery efficacy of the PEI-C/siPAI-1 polyplexes was investigated in vitro in primary lung fibroblasts isolated from mice with bleomycin-induced PF. Biodistribution analysis following intratracheal administration of fluorescently labeled polyplexes showed prolonged retention in the lungs. Treatment of mice with bleomycin-induced PF using the PEI-C/siPAI-1 polyplexes resulted in a significant down-regulation of the PAI-1 expression and decreased collagen deposition in the lung. The results of this study provide first evidence of the potential benefits of combined inhibition of CXCR4 and PAI-1 in the pulmonary treatment of PF.


Assuntos
Sistemas de Liberação de Medicamentos , Polietilenoimina/química , Polímeros/administração & dosagem , Fibrose Pulmonar/prevenção & controle , RNA Interferente Pequeno/genética , Receptores CXCR4/antagonistas & inibidores , Serpina E2/antagonistas & inibidores , Animais , Antibióticos Antineoplásicos/toxicidade , Apoptose , Bleomicina/toxicidade , Proliferação de Células , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patologia , Inativação Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Polímeros/química , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Receptores CXCR4/genética , Serpina E2/genética
14.
Adv Healthc Mater ; 7(5)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29280311

RESUMO

The use of small interfering RNA (siRNA) in cancer treatment has been limited by the lack of effective systemic delivery methods. Although synthetic polycations have been widely explored in siRNA delivery, polycation/siRNA polyplexes often suffer from insufficient stability in vivo. Here, rationally designed siRNA delivery systems that meet the requirements for systemic siRNA delivery to distant tumors are reported. The hypothesis that modular design of delivery systems based on poly(amido amine)s that combine fluorination for systemic stability with bioreducibility for easy intracellular siRNA release, and PEGylation for improved safety and colloidal stability will overcome problems with contradicting siRNA delivery demands is tested. PEGylated, fluorinated, and bioreducible copolymers (PEG-PCD-F) with different degree of fluorination are thus synthesized. The fluorinated copolymers readily formed polyplexes with siRNA and achieved greatly improved gene silencing efficacy in multiple cell lines in vitro when compared with nonfluorinated controls. The results show fluorination-induced enhancement of stability, cellular uptake, and endosomal escape of the polyplexes, while exhibiting efficient siRNA release in reducing intracellular environment. PEG-PCD-F polyplexes with siRNA against Bcl2 inhibit breast tumor growth following systemic intravenous administration. The results provide strong evidence of successful combination of bioreducibility with fluorination and PEGylation to achieve systemic siRNA polyplex delivery.


Assuntos
Hidrocarbonetos Fluorados , Neoplasias Mamárias Experimentais/tratamento farmacológico , Poliaminas , RNA Interferente Pequeno , Animais , Células CHO , Cricetulus , Feminino , Hidrocarbonetos Fluorados/química , Hidrocarbonetos Fluorados/farmacocinética , Hidrocarbonetos Fluorados/farmacologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Poliaminas/química , Poliaminas/farmacocinética , Poliaminas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacocinética , RNA Interferente Pequeno/farmacologia
15.
Drug Dev Ind Pharm ; 42(9): 1424-33, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26984179

RESUMO

Liposomes have successfully been used for decades to encapsulate and protect drugs that are prone to deactivation in the body. The present study aimed to demonstrate the use of liposomes to encapsulate cordycepin, an adenosine analog that quickly loses its activity in vivo. The cordycepin-loaded liposomes were prepared by the ammonium sulfate gradient approach, and its in vitro and in vivo antitumour activities were evaluated using BEL-7402 cells and hepatocellular carcinoma H22 transplanted tumors, respectively. An MTT assay was used to observe the cytotoxicity of cells treated with cordycepin and cordycepin-loaded liposomes in vitro. High-content screening (HSC) was carried out using Hoechst 33342 to detect apoptotic cells and the ratio of cells in different cell cycle stages. The data demonstrated that both the cordycepin and the cordycepin-loaded liposomes resulted in clear cytotoxicity with IC50 values of 18.97 and 29.39 µg/mL, respectively. The latter showed significantly strong inhibitory effects on H22 tumor growth in mice, while the former did not show any inhibitory effects on tumor growth. In addition, the HSC assay showed that the cordycepin-loaded liposomes resulted in a higher rate of apoptosis than the cordycepin alone in BEL-7402 cells. Further data analysis revealed that the cells treated with cordycepin-loaded liposomes were predominately arrested at the G2/M phase (p < 0.05), while those treated with cordycepin alone were arrested in the G0/G1 phase (p < 0.05). In conclusion, these results suggest that liposomes can enhance and maintain the in vivo anti-tumor activity of cordycepin.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Desoxiadenosinas/farmacologia , Lipossomos/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA