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








Base de dados
Intervalo de ano de publicação
1.
J Control Release ; 362: 524-535, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37673307

RESUMO

Chimeric antigen receptor (CAR)-modified natural killer (NK) cells are recognized as promising immunotherapeutic agents for cancer treatment. However, the efficacy and trafficking of CAR-NK cells in solid tumors are hindered by the complex barriers present in the tumor microenvironment (TME). We have developed a novel strategy that utilizes living CAR-NK cells as carriers to deliver anticancer drugs specifically to the tumor site. We also introduce a time-lapse method for evaluating the efficacy and tumor specificity of CAR-NK cells using a two-photon microscope in live mouse models and three-dimensional (3D) tissue slide cultures. Our results demonstrate that CAR-NK cells exhibit enhanced antitumor immunity when combined with photosensitive chemicals in both in vitro and in vivo tumor models. Additionally, we have successfully visualized the trafficking, infiltration, and accumulation of drug-loaded CAR-NK cells in deeply situated TME using non-invasive intravital two-photon microscopy. Our findings highlight that tumor infiltration of CAR-NK cells can be intravitally monitored through the two-photon microscope approach. In conclusion, our study demonstrates the successful integration of CAR-NK cells as drug carriers and paves the way for combined cellular and small-molecule therapies in cancer treatment. Furthermore, our 3D platform offers a valuable tool for assessing the behavior of CAR cells within solid tumors, facilitating the development and optimization of immunotherapeutic strategies with clinical imaging approaches.

2.
Front Immunol ; 14: 1213285, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37503351

RESUMO

HPV (Human papillomavirus) affects 600,000 people worldwide each year. Almost all cervical cancers are associated with a past HPV infection. In particular, the positivity to the high-risk type HPV16 is detected in most of the invasive cervical cancers. FDA has approved prophylactic vaccines that protect against new HPV16 infections, but do not induce immunity in those patients with established infections or neoplasms. To date, no therapeutic vaccine targeting HPV16-associated lesions has been authorized. We have developed an mRNA-based vaccine against the HPV16 late oncoproteins E6 and E7, which are abundantly and exclusively expressed in high-grade squamous intraepithelial lesions (HSILs), a stage of the cervical disease that precedes the progression to carcinoma. Our in vitro and in vivo studies demonstrated that the translated mRNA is functional and elicits an antigen-specific adaptive immune response. Upon immunization with the vaccine, mice with HPV16+ lesions exhibited tumor growth inhibition, extension of lifespan, and development of a protective immune memory. In light of these results and the remarkable clinical success of mRNA vaccines against SARS-CoV2, we believe that our mRNA-based therapeutic vaccine has the potential to offer a non-invasive treatment alternative to the current standard of care for HPV16+ HSILs.


Assuntos
COVID-19 , Infecções por Papillomavirus , Neoplasias do Colo do Útero , Feminino , Humanos , Animais , Camundongos , Neoplasias do Colo do Útero/prevenção & controle , Neoplasias do Colo do Útero/patologia , Papillomavirus Humano 16/genética , RNA Viral , COVID-19/complicações , SARS-CoV-2/genética , Papillomavirus Humano , RNA Mensageiro/genética
4.
Small ; 15(15): e1900300, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30884183

RESUMO

Cells secrete substances that are essential to the understanding of numerous immunological phenomena and are extensively used in clinical diagnoses. Countless techniques for screening of biomarker secretion in living cells have generated valuable information on cell function and physiology, but low volume and real-time analysis is a bottleneck for a range of approaches. Here, a simple, highly sensitive assay using a high-throughput micropillar and microwell array chip (MIMIC) platform is presented for monitoring of biomarkers secreted by cancer cells. The sensing element is a micropillar array that uses the enzyme-linked immunosorbent assay (ELISA) mechanism to detect captured biomolecules. When integrated with a microwell array where few cells are localized, interleukin 8 (IL-8) secretion can be monitored with nanoliter volume using multiple micropillar arrays. The trend of cell secretions measured using MIMICs matches the results from conventional ELISA well while it requires orders of magnitude less cells and volumes. Moreover, the proposed MIMIC is examined to be used as a drug screening platform by delivering drugs using micropillar arrays in combination with a microfluidic system and then detecting biomolecules from cells as exposed to drugs.


Assuntos
Biomarcadores/análise , Ensaios de Triagem em Larga Escala/métodos , Microtecnologia/métodos , Animais , Anticorpos/análise , Contagem de Células , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Humanos , Camundongos
5.
Int J Biol Sci ; 14(7): 799-806, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29910690

RESUMO

The insulin-like growth factors (IGFs), IGF-I and IGF-II, are essential for regulating cell growth, differentiation and metastasis of a broad range of malignancies. The IGF-I/II actions are mediated through the IGF receptor type 1 (IGF-1R) and the insulin receptor (IR), which are overexpressed in multiple types of tumors. Here, we have firstly identified a human engineered antibody domain (eAd) from a phage-displayed VH library. The eAd suppressed the signal transduction of IGF-1R mediated by exogenous IGF-I or IGF-II in breast cancer cell lines through neutralizing both IGF-I and IGF-II. It also significantly inhibited the growth of breast cancer cells. Therefore, the anti-IGF-I/II eAd offers an alternative approach to target both the IGF-1R signaling pathways through the inhibition of IGF-I/II.


Assuntos
Neoplasias da Mama/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Proliferação de Células/fisiologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like II/genética , Fosforilação/genética , Fosforilação/fisiologia , Transdução de Sinais
6.
Adv Mater ; 30(22): e1706245, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29577477

RESUMO

White blood cells (WBCs) are a major component of immunity in response to pathogen invasion. Neutrophils are the most abundant WBCs in humans, playing a central role in acute inflammation induced by pathogens. Adhesion to vasculature and tissue infiltration of neutrophils are key processes in acute inflammation. Many inflammatory/autoimmune disorders and cancer therapies have been found to be involved in activation and tissue infiltration of neutrophils. A promising strategy to develop novel targeted drug delivery systems is the targeting and exploitation of activated neutrophils. Herein, a new drug delivery platform based on neutrophils is reviewed. There are two types of drug delivery systems: neutrophils as carriers and neutrophil-membrane-derived nanovesicles. It is discussed how nanoparticles hijack neutrophils in vivo to deliver therapeutics across blood vessel barriers and how neutrophil-membrane-derived nanovesicles target inflamed vasculature. Finally, the potential applications of neutrophil-based drug delivery systems in treating inflammation and cancers are presented.


Assuntos
Neutrófilos , Sistemas de Liberação de Medicamentos , Humanos , Inflamação , Nanopartículas , Neoplasias
7.
Ther Deliv ; 9(1): 29-35, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29216803

RESUMO

Nanotherapeutics, nanoparticles (NPs) loaded with drugs, show the ability of tissue targeting, long circulation and low toxicity compared with free drugs. Endothelium lying the lumen of a blood vessel is a barrier to restrain tissue deposition of nanotherapeutics. Neutrophils, a type of white blood cells, migrate across endothelium during inflammation. There is an emerging concept that in situ targeting of neutrophils allows delivery of nanotherapeutics into deep tissues at disease sites. Here we summarize the recent advances in delivery of nanotherapeutics to inflammatory tissues or tumor microenvironments via neutrophil infiltration. The studies would shift the current paradigm of nanomedicine to biology-driven design of nanotherapeutics. [Formula: see text].


Assuntos
Portadores de Fármacos/química , Nanopartículas/química , Neutrófilos/imunologia , Anticorpos/química , Anticorpos/uso terapêutico , Antígeno CD11b/imunologia , Humanos , Nanomedicina , Neoplasias/tratamento farmacológico , Infiltração de Neutrófilos/fisiologia , Neutrófilos/citologia , Neutrófilos/fisiologia , Polietilenoglicóis/química
8.
J Pharm Biomed Anal ; 145: 255-261, 2017 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-28688270

RESUMO

PEGylation is practically one of most important modifications of drugs including small molecules, peptides and proteins, which has been proven to dramatically improve physicochemical properties and pharmacokinetic behavior of the PEGylated drugs. However, it is a challenge currently to quantitatively analyze PEG and PEGylated drugs by various analytical methods, even mass spectrometry because of multiple parent ion distribution of PEG caused by its polydispersity of molecular weight. Here we developed a robust method with MS/MSALL technique using electrospray ionization (ESI) source coupled high resolution Quadrupole Time-of-Flight (Q-TOF) mass spectrometry for the quantification of PEG2K-Paclitaxel (PEG-PTX) and its two metabolites, PEG and Paclitaxel (PTX). The analysis was performed on a 300SB-C18 column with acetonitrile and 0.1% formic acid as the mobile phase. Samples were simply prepared by protein precipitation in a small quantity of plasma (50µL). Calibration curve was linear within the range of 50.0-4000ng/mL for PEG and PEG-PTX and 1.0-1000ng/mL for PTX. The intra- and inter-day precisions were 3.2-6.9% and 3.1-6.9% for PEG, 4.1-7.8% and 4.0-9.9% for PEG-PTX, and 3.3-4.8% and 3.1-6.9% for PTX, respectively. The recoveries were greater than 90% with low matrix effects. Afterwards, the newly developed method was successfully applied to support a preclinical pharmacokinetic study in six rats after single intravenous injection of PEG-PTX (51.7mg/kg).


Assuntos
Paclitaxel/análise , Animais , Plasma , Polietilenoglicóis , Proteínas , Ratos , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem
9.
Adv Mater ; 29(27)2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28504320

RESUMO

Remodeling of tumor microenvironments enables enhanced delivery of nanoparticles (NPs). This study shows that direct priming of a tumor tissue using photosensitization rapidly activates neutrophil infiltration that mediates delivery of nanotherapeutics into the tumor. A drug delivery platform is comprised of NPs coated with anti-CD11b antibodies (Abs) that target activated neutrophils. Intravital microscopy demonstrates that the movement of anti-CD11b Abs-decorated NPs (NPs-CD11b) into the tumor is mediated by neutrophil infiltration induced by photosensitization (PS) because the systemic depletion of neutrophils completely abolishes the nanoparticle tumor deposition. The neutrophil uptake of NPs does not alter neutrophil activation and transmigration. For cancer therapy in mice, tumor PS and photothermal therapy of anti-CD11b Abs-linked gold nanorods (GNRs-CD11b) are combined to treat the carcinoma tumor. The result indicates that neutrophil tumor infiltration enhances nanoparticle cancer therapy. The findings reveal that promoting tumor infiltration of neutrophils by manipulating tumor microenvironments could be a novel strategy to actively deliver nanotherapeutics in cancer therapies.


Assuntos
Microambiente Tumoral , Animais , Camundongos , Nanopartículas , Infiltração de Neutrófilos , Neutrófilos , Transtornos de Fotossensibilidade
10.
Theranostics ; 7(3): 751-763, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28255364

RESUMO

Nanotechnology has become a powerful tool to potentially translate nanomedicine from bench to bedside. Nanotherapeutics are nanoparticles (NPs) loaded with drugs and possess the property of tissue targeting after surfaces of NPs are bio-functionalized. Designing smaller size of nanotherapeutics is presumed to increase tumor targeting based on the EPR (enhanced permeability and retention) effect. Since the immune systems possess a defence mechanism to fight diseases, there is an emerging concept that NPs selectively target immune cells to mediate the active delivery of drugs into sites of disease. In this review, we will focus on a key question of how nanotherapeutics specifically target immune cells and hijack them as a delivery vehicle to transport nanotherapeutics into disease tissues, thus possibly improving current therapies in inflammation, immune disorders and cancers. We will also discuss the challenges and opportunities for this new strategy of leukocyte-mediated delivery of nanotherapeutics.


Assuntos
Anti-Inflamatórios/administração & dosagem , Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Inflamação/tratamento farmacológico , Leucócitos/metabolismo , Nanopartículas/metabolismo , Neoplasias/tratamento farmacológico , Animais , Portadores de Fármacos , Humanos
11.
Adv Healthc Mater ; 5(9): 1088-93, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26989887

RESUMO

Cancer immunotherapy using tumor-specific monoclonal antibodies presents a novel approach for cancer treatment. A monoclonal antibody TA99 specific for gp75 antigen of melanoma initiates neutrophil recruitment in tumor responsible for cancer therapy. Here, a strategy is reported for hijacking neutrophils in vivo using nanoparticles (NPs) to deliver therapeutics into tumor. In a mouse model of melanoma, it is shown that systemically delivered albumin NPs increase in tumor when TA99 antibody is injected; and the NP tumor accumulation is mediated by neutrophils. After the administration of pyropheophorbide-a loaded albumin NPs and TA99, photodynamic therapy significantly suppresses the tumor growth and increases mouse survival compared with treatment with the NPs or TA99. The study reveals a new avenue to treat cancer by NP hitchhiking of immune systems to enhance delivery of therapeutics into tumor sites.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Imunoterapia , Nanopartículas/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Animais , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/imunologia , Camundongos , Nanopartículas/química , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Infiltração de Neutrófilos/imunologia
12.
J Control Release ; 224: 208-216, 2016 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-26778696

RESUMO

Vascular inflammation is the underlying component of most diseases. To target inflamed vasculature, nanoparticles are commonly engineered by conjugating antibody to the nanoparticle surface, but this bottom-up approach could affect nanoparticle targeting and therapeutic efficacy in complex, physiologically related systems. During vascular inflammation endothelium via the NF-κB pathway instantly upregulates intercellular adhesion molecule 1 (ICAM-1) which binds integrin ß2 on neutrophil membrane. Inspired by this interaction, we created a nanovesicle-based drug delivery system using nitrogen cavitation which rapidly disrupts activated neutrophils to make cell membrane nanovesicles. Studies using intravital microscopy of live mouse cremaster venules showed that these vesicles can selectively bind inflamed vasculature because they possess intact targeting molecules of integrin ß2. Administering of nanovesicles loaded with TPCA-1 (a NF-κB inhibitor) markedly mitigated mouse acute lung inflammation. Our studies reveal a new top-down strategy for directly employing a diseased tissue to produce biofunctional nanovesicle-based drug delivery systems potentially applied to treat various diseases.


Assuntos
Membrana Celular/química , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , Amidas/administração & dosagem , Amidas/uso terapêutico , Animais , Linhagem Celular , Humanos , Inflamação/tratamento farmacológico , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , NF-kappa B/metabolismo , Pneumonia/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Tiofenos/administração & dosagem , Tiofenos/uso terapêutico , Doenças Vasculares/tratamento farmacológico , Vênulas/efeitos dos fármacos , Vênulas/metabolismo
13.
Adv Healthc Mater ; 4(17): 2709-18, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26474414

RESUMO

Most drug delivery systems have been developed for efficient delivery to tumor sites via targeting and on-demand strategies, but the carriers rarely execute synergistic therapeutic actions. In this work, C8, a cationic, pH-triggered anticancer peptide, is developed by incorporating histidine-mediated pH-sensitivity, amphipathic helix, and amino acid pairing self-assembly design. We designed C8 to function as a pH-responsive nanostructure whose cytotoxicity can be switched on and off by its self-assembly: Noncytotoxic ß-sheet fibers at high pH with neutral histidines, and positively charged monomers with membrane lytic activity at low pH. The selective activity of C8, tested for three different cancer cell lines and two noncancerous cell lines, is shown. Based on liposome leakage assays and multiscale computer simulations, its physical mechanisms of pore-forming action and selectivity are proposed, which originate from differences in the lipid composition of the cellular membrane and changes in hydrogen bonding. C8 is then investigated for its potential as a drug carrier. C8 forms a nanocomplex with ellipticine, a nonselective model anticancer drug. It selectively targets cancer cells in a pH-responsive manner, demonstrating enhanced efficacy and selectivity. This study provides a novel powerful strategy for the design and development of multifunctional self-assembling peptides for therapeutic and drug delivery applications.


Assuntos
Antineoplásicos/química , Peptídeos/química , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Elipticinas/química , Histidina/química , Humanos , Concentração de Íons de Hidrogênio , Lipossomos/química , Camundongos , Células NIH 3T3 , Nanoestruturas/química
14.
ACS Nano ; 9(12): 11800-11, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26516654

RESUMO

Endothelial cells form a monolayer in lumen of blood vessels presenting a great barrier for delivery of therapeutic nanoparticles (NPs) into extravascular tissues where most diseases occur, such as inflammation disorders and infection. Here, we report a strategy for delivering therapeutic NPs across this blood vessel barrier by nanoparticle in situ hitchhiking activated neutrophils. Using intravital microscopy of TNF-α-induced inflammation of mouse cremaster venules and a mouse model of acute lung inflammation, we demonstrated that intravenously (iv) infused NPs made from denatured bovine serum albumin (BSA) were specifically internalized by activated neutrophils, and subsequently, the neutrophils containing NPs migrated across blood vessels into inflammatory tissues. When neutrophils were depleted using anti-Gr-1 in a mouse, the transport of albumin NPs across blood vessel walls was robustly abolished. Furthermore, it was found that albumin nanoparticle internalization did not affect neutrophil mobility and functions. Administration of drug-loaded albumin NPs markedly mitigated the lung inflammation induced by LPS (lipopolysaccharide) or infection by Pseudomonas aeruginosa. These results demonstrate the use of an albumin nanoparticle platform for in situ targeting of activated neutrophils for delivery of therapeutics across the blood vessel barriers into diseased sites. This study demonstrates our ability to hijack neutrophils to deliver nanoparticles to targeted diseased sites.


Assuntos
Antibacterianos/farmacocinética , Vasos Sanguíneos/metabolismo , Portadores de Fármacos/farmacocinética , Nanopartículas/química , Neutrófilos/metabolismo , Animais , Antibacterianos/química , Células Cultivadas , Portadores de Fármacos/química , Portadores de Fármacos/uso terapêutico , Infecções/tratamento farmacológico , Inflamação/tratamento farmacológico , Masculino , Camundongos , Neutrófilos/química , Soroalbumina Bovina/química , Soroalbumina Bovina/farmacocinética
15.
Acta Biomater ; 21: 74-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25861950

RESUMO

Small interfering RNA (siRNA) shows great therapeutic potential due to its ability to regulate gene expression in a highly selective manner. However, its application has been limited by ineffective cellular uptake of siRNAs. To achieve successful gene-silencing efficiency, a safe and effective delivery vector is generally required. In this study, we designed a series of amphipathic peptides that comprised a variant of a nuclear localization sequence, 0-6 histidine residues and an optional stearic acid group. Among these candidates, STR-HK exhibited good characteristics as a safe and efficient siRNA delivery vector, facilitating efficient siRNA delivery to mammalian cells without causing cytotoxicity. Moreover, the intratumoral injection of STR-HK/siRNA complexes achieved high anti-tumor activity through the downregulation of the Bcl-2 protein in mice, with an inhibition rate of 62.8%. Our data demonstrate that STR-HK is a highly promising siRNA delivery vector for therapeutic applications.


Assuntos
Peptídeos/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Linhagem Celular Tumoral , Humanos , Técnicas In Vitro , Microscopia de Força Atômica , Espectroscopia de Infravermelho com Transformada de Fourier
16.
Nanomedicine ; 11(2): 435-46, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25193363

RESUMO

Recently, cell-penetrating peptides (CPPs) have received much attention for cellular delivery of therapeutic molecules. However, in the case of CPPs as carriers for siRNA delivery, their utility is often restricted by low cellular uptake and/or entrapment in endosomes. Here, in order to deliver siRNAs with high efficiency, oligoarginine, a prominent member in CPPs, is rationally modified with oligohistidine and stearyl moieties (STR-) by fully taking into account the formation of nanoparticles, uptake and intracellular trafficking. We show that when the ratio of histidine/arginine in a peptide sequence is >1.5, pronounced gene silencing is induced. Following this rule, STR-HnR8 (n=16 and 20) are developed, which show a high knockdown efficiency rarely reported before. Finally, we find that endosomal escape of siRNA induced by stearylated and oligohistidylated oligoarginine is only from "proton-sponge" effect. Taken together, our results suggest a new strategy for the improvement of CPP-based siRNA delivery systems. FROM THE CLINICAL EDITOR: This study present a novel cell penetrating peptide-based siRNA delivery system utilizing modified oligo-arginine demonstrating a successful siRNA delivery approach.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , Técnicas de Transferência de Genes , Nanopartículas/administração & dosagem , RNA Interferente Pequeno/uso terapêutico , Animais , Arginina/administração & dosagem , Arginina/química , Arginina/genética , Células CHO , Peptídeos Penetradores de Células/química , Cricetulus , Endossomos/química , Técnicas de Silenciamento de Genes , Humanos , Microscopia de Força Atômica , Nanopartículas/química , Tamanho da Partícula , RNA Interferente Pequeno/genética , Relação Estrutura-Atividade
17.
Mol Pharm ; 12(1): 56-65, 2015 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-25378277

RESUMO

Gene therapy using RNA interference (RNAi) technology has been explored to treat cancers, by regulating the expression of oncogene. However, even though small interfering RNA (siRNA), which triggers RNAi, may have great therapeutic potential, efforts at using them in vivo have been hampered by the difficulty of effective and safe delivery into cells of interest. In this study, to develop a safe and efficient carrier for in vitro and in vivo siRNA delivery, we designed a peptide library. These peptides are improved variants of a known peptide based siRNA carrier C6. All the modifications improved the transfection efficiency of C6 to some degree. After completing prescreening for activity, several promising candidates were used for further evaluation. Selected peptides C6M3 and C6M6 could form stable complexes with siRNA. These complexes could be greatly uptaken by cells and showed a punctate perinuclear distribution. Moreover, peptide/siRNA complexes achieved high transfection efficiency in vitro without inducing substantial cytotoxicity. We have validated the therapeutic potential of this strategy for cancer treatment by targeting Bcl-2 gene in mouse tumor models, and demonstrated that tumor growth was inhibited. In order to address possible immune side effects of these peptide carriers, biocompatibility study in terms of complement activation and cytokine activation assay were carried out, whereas none of the peptides induced such effects. In conclusion, these results support the potential of these peptides as therapeutic siRNA carrier.


Assuntos
Materiais Biocompatíveis/química , Portadores de Fármacos/química , Desenho de Fármacos , Endossomos/química , RNA Interferente Pequeno/metabolismo , Animais , Células CHO , Linhagem Celular Tumoral , Separação Celular , Sobrevivência Celular , Cricetinae , Cricetulus , Ensaios de Seleção de Medicamentos Antitumorais , Citometria de Fluxo , Técnicas de Transferência de Genes , Humanos , Masculino , Camundongos , Microscopia de Força Atômica , Transplante de Neoplasias , Peptídeos/química , Interferência de RNA
18.
Int J Pharm ; 465(1-2): 1-4, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24530387

RESUMO

Macrophage induced surface degradation of poly(ethylene carbonate) (PEC) was investigated under in vitro conditions. Degradation of PEC with the MW of 41 kDa (PEC41) was slower than that of PEC with the MW of 200 kDa (PEC200). In terms of macrophage mediated drug release from PEC matrix, in cell-free medium, less than 1% of levofloxacin was released from both PEC samples in 10 days, while more than 60 and 20% of the drug, levofloxacin, can be detected in medium with macrophages from PEC200 and PEC41 films, respectively. This work indicated that on-demand drug delivery induced by macrophages can be achieved with PEC polymer.


Assuntos
Portadores de Fármacos , Levofloxacino/química , Macrófagos/metabolismo , Polietilenos/química , Animais , Linhagem Celular , Química Farmacêutica , Preparações de Ação Retardada , Estudos de Viabilidade , Cinética , Camundongos , Peso Molecular , Solubilidade , Propriedades de Superfície
19.
J Mater Chem B ; 2(36): 6010-6019, 2014 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-32261853

RESUMO

At the forefront of medicine, gene therapy provides an effective way to treat a range of diseases by regulating defective genes at the root of the disease. Short interfering RNAs (siRNAs) hold great promise as therapeutic agents in this domain; however, intracellular delivery remains a major obstacle to clinical applications of therapeutic siRNAs. Here we report a peptide designed to mediate siRNA delivery. This peptide, C6M1, is rationally designed to promote the endosomal escape ability of an existing peptide sequence. Formed C6M1-siRNA nanoscale complexes are able to deliver siRNA into cells and induce specific gene knockdown with low toxicity. The increased membrane disruption ability under acidic conditions of the peptide with tryptophan residue substitution may contribute to the enhanced gene silence efficacy. Intratumoral injection of the complexes results in a marked reduction of tumor growth through downregulation of antiapoptotic Bcl-2 protein in mice. In addition, the C6M1-siRNA complex was proven safe at transfection concentration by cytotoxicity assay. These results demonstrate that the C6M1-siRNA complex is a potent system for efficient gene delivery in vitro and in vivo.

20.
Eur J Pharm Biopharm ; 85(3 Pt B): 1245-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23791717

RESUMO

The objective of this study was to investigate the effect of molecular weight (MW) on the drug release from poly(ethylene carbonate) (PEC) based surface-eroding in situ forming depots (ISFD). In phosphate buffered saline (PBS) pH 7.4, 63.7% of bovine serum albumin BSA was released from high MW PEC of 200 kDa (PEC200) in DMSO (15%, w/w) in 2 days, while during the same time period, the release of BSA from PEC41 samples was only 22.5%. At higher concentrations of PEC41 (25%, w/w), the initial burst was further reduced, and even after 6 days, only 16.3% was released. Compared to depots based on PEC200, there was lower rate of solvent release, slower phase inversion, and a denser surface in PEC41 samples. An expansion in size of PEC41 depots suggested that the polymer barrier of PEC41 impeded the diffusion of solvent out of the samples effectively. In conclusion, the initial burst of protein from ISFD of PEC41 was significantly reduced, which would be a promising candidate as polymeric carrier.


Assuntos
Polietilenos/química , Animais , Materiais Biocompatíveis/química , Bovinos , Química Farmacêutica/métodos , Difusão , Sistemas de Liberação de Medicamentos , Estabilidade de Medicamentos , Concentração de Íons de Hidrogênio , Infusões Parenterais , Microscopia Eletrônica de Varredura , Peso Molecular , Tamanho da Partícula , Polímeros/química , Soroalbumina Bovina/química , Solventes/química , Viscosidade , Água/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA