Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Pharm ; 14(6): 1969-1979, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28460165

RESUMO

Mefloquine (Mef), a poorly soluble and highly bitter drug, has been used for malaria prophylaxis and treatment. The dosage form for Mef is mostly available as adult tablets, and thus children under the age of 5 suffer from poor medication adherence. We have developed a stable, rapidly dissolvable, and palatable pediatric formulation for Mef using liposomes composed of 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) and cholesterol with a mean diameter of ∼110 nm. Mef was actively loaded into the liposomes via an ammonium sulfate gradient using the solvent-assisted loading technology (SALT) developed in our lab. Complete loading of Mef inside the liposomal core was achieved at a high drug-to-lipid ratio (D/L) of 0.1-0.2 (w/w), and the final drug content in the formulation was ∼8 mg/mL, well above the solubility of Mef (<0.6 mg/mL in simulated fluids). The strong bitterness of Mef was masked by the liposomal encapsulation as measured by an electronic tongue. Incubating the Mef-liposomes (Mef-Lipo) in the simulated gastric fluid (pH 1.2) and the simulated intestinal fluid containing 3 mM sodium taurocholate (pH 6.8) induced changes in liposome size and the polydispersity, resulting in drug release (∼40% in 2 h). However, no drug release from the Mef-Lipo was measured in the bile salt-free intestinal fluid or simulated saliva (0% in 3 h). These data suggest that drug release from the Mef-Lipo was mediated by a low pH and the presence of a surfactant. Pancreatic lipase did not degrade DSPC in the Mef-Lipo after 8 h of incubation nor induce Mef release from the liposomes, indicating that lipid digestion played a minor role for drug release from the Mef-Lipo. In order to improve long-term room temperature storage, the Mef-Lipo was lyophilized to obtain a solid formulation, which was completely dissolvable in water in 10 s and displayed similar in vitro profiles of release as the liquid form. The lyophilized Mef-Lipo was stable at room temperature for >3 months. In mice, orally delivered liquid and lyophilized Mef-Lipo displayed comparable absorption with bioavailability (BA) of 81-86%, while the absorption of the standard Mef suspension was significantly lower with BA of 70% and 20% decreased maximal plasma concentration and area under the curve. Our data suggest that the Mef-Lipo was a stable, palatable, and bioavailable formulation that might be suitable for pediatric use.


Assuntos
Lipossomos/química , Mefloquina/química , Administração Oral , Animais , Dimetil Sulfóxido/química , Sistemas de Liberação de Medicamentos/métodos , Feminino , Malária/tratamento farmacológico , Mefloquina/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Fosfatidilcolinas/química , Solubilidade
2.
Pharm Res ; 33(5): 1104-14, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26758590

RESUMO

PURPOSE: This study was aimed at developing a new active loading method to stably encapsulate staurosporine (STS), a water insoluble drug, into lipid-based nanoparticles (LNPs) for drug targeting to tumors. METHODS: A limited amount of DMSO was included during the active loading process to prevent precipitation and facilitate the loading of insoluble STS into the aqueous core of a LNP. The drug loading kinetics under various conditions was studied and the STS-LNPs were characterized by size, drug-to-lipid ratio, drug release kinetics and in vitro potency. The antitumor efficacy of the STS-LNPs was compared with free STS in a mouse model. RESULTS: The drug loading efficiency reached 100% within 15 min of incubation at a drug-to-lipid ratio of 0.31 (mol) via an ammonium gradient. STS formed nano-aggregates inside the aqueous core of the LNPs and was stably retained upon storage and in the presence of serum. A 3-fold higher dose of the STS-LNPs could be tolerated by BALB/c mice compared with free STS, leading to nearly complete growth inhibition of a multidrug resistant breast tumor, while free STS only exhibited moderate activity. CONCLUSION: This simple and efficient drug loading method produced a stable LNP formulation for STS that was effective for cancer treatment.


Assuntos
Lipídeos/química , Lipossomos/química , Nanopartículas/química , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Estaurosporina/administração & dosagem , Animais , Linhagem Celular Tumoral , Dimetil Sulfóxido/química , Sistemas de Liberação de Medicamentos , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Lipossomos/ultraestrutura , Camundongos Endogâmicos BALB C , Modelos Moleculares , Nanopartículas/ultraestrutura , Neoplasias/patologia , Tamanho da Partícula , Inibidores de Proteínas Quinases/uso terapêutico , Estaurosporina/uso terapêutico
3.
Bioconjug Chem ; 22(12): 2474-86, 2011 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-22014112

RESUMO

A nanoparticle formulation of docetaxel (DTX) was designed to address the strengths and limitations of current taxane delivery systems: PEGylation, high drug conjugation efficiency (>30 wt %), a slow-release mechanism, and a well-defined and stable nanoparticle identity were identified as critical design parameters. The polymer conjugate was synthesized with carboxymethylcellulose (CMC), an established pharmaceutical excipient characterized by a high density of carboxylate groups permitting increased conjugation of a drug. CMC was chemically modified through acetylation to eliminate its gelling properties and to improve solvent solubility, enabling high yield and reproducible conjugation of DTX and poly(ethylene glycol) (PEG). The optimal conjugate formulation (Cellax) contained 37.1 ± 1.5 wt % DTX and 4.7 ± 0.8 wt % PEG, exhibited a low critical aggregation concentration of 0.6 µg/mL, and formed 118-134 nm spherical nanoparticles stable against dilution. Conjugate compositions with a DTX degree of substitution (DS) outside the 12.3-20.8 mol % range failed to form discrete nanoparticles, emphasizing the importance of hydrophobic and hydrophilic balance in molecular design. Cellax nanoparticles released DTX in serum with near zero order kinetics (100% in 3 weeks), was internalized in murine and human cancer cells, and induced significantly higher toxic effects against a panel of tumor cell lines (2- to 40-fold lower IC50 values) compared to free DTX.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Carboximetilcelulose Sódica/química , Portadores de Fármacos/química , Nanopartículas/química , Neoplasias/tratamento farmacológico , Taxoides/administração & dosagem , Taxoides/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Docetaxel , Humanos , Concentração Inibidora 50 , Camundongos , Polietilenoglicóis/química , Taxoides/química , Taxoides/farmacocinética
4.
Nanomaterials (Basel) ; 11(8)2021 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-34443815

RESUMO

BACKGROUND: Pathogenic intracellular mycobacteria are challenging to treat because of the waxy and complex cell wall characterizing the genus. Niosomes are vesicles with biomimetic cell membrane composition, which allow them to efficiently bind to the eukaryotic cells and deliver their cargo into the cytoplasm. The objective of this study was to develop a new platform based on niosomes loaded with antimicrobial agents to target intracellular mycobacteria. Nanoniosomes were fabricated and loaded with antibiotics and lignin-silver nanoparticles. The efficacy of these nanoniosomes was tested against the intracellular pathogen Mycobacterium abscessus used as a model of infection of human-derived macrophages (THP-1). The cytotoxicity and the immunological response of the agents were tested on THP-1 cells using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and the secretion of pro- and anti-inflammatory cytokines, respectively. RESULTS: M. abscessus was susceptible to the nanoniosomes in infected THP-1 macrophages, suggesting that the nanoniosomes were internalized due to their fusion to the macrophage cellular membrane. Moreover, nanoniosomes showed no upregulation of pro-inflammatory cytokines when exposed to THP-1 macrophages. CONCLUSIONS: Nanoniosomes improved drug efficacy while decreasing toxicity and should be considered for further testing in the treatment of intracellular pathogenic mycobacteria or as a new platform for precise intracellular delivery of drugs.

5.
J Control Release ; 330: 1080-1094, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33189786

RESUMO

Resiquimod (R848) is a toll-like receptor 7 and 8 (TLR7/8) agonist with potent antitumor and immunostimulatory activity. However, systemic delivery of R848 is poorly tolerated because of its poor solubility in water and systemic immune activation. In order to address these limitations, we developed an intravenously-injectable formulation with R848 using thermosensitive liposomes (TSLs) as a delivery vehicle. R848 was remotely loaded into TSLs composed of DPPC: DSPC: DSPE-PEG2K (85:10:5, mol%) with 100 mM FeSO4 as the trapping agent inside. The final R848 to lipid ratio of the optimized R848-loaded TSLs (R848-TSLs) was 0.09 (w/w), 10-fold higher than the previously-reported values. R848-TSLs released 80% of R848 within 5 min at 42 °C. These TSLs were then combined with αPD-1, an immune checkpoint inhibitor, and ultrasound-mediated hyperthermia in a neu deletion (NDL) mouse mammary carcinoma model (Her2+, ER/PR negative). Combined with αPD-1, local injection of R848-TSLs showed superior efficacy with complete NDL tumor regression in both treated and abscopal sites achieved in 8 of 11 tumor bearing mice over 100 days. Immunohistochemistry confirmed enhanced CD8+ T cell infiltration and accumulation by R848-TSLs. Systemic delivery of R848-TSLs, combined with local hyperthermia and αPD-1, inhibited tumor growth and extended median survival from 28 days (non-treatment control) to 94 days. Upon re-challenge with reinjection of tumor cells, none of the previously cured mice developed tumors, as compared with 100% of age-matched control mice. The dose of R848 (10 µg for intra-tumoral injection or 6 mg/kg for intravenous injection delivered up to 4 times) was well-tolerated without weight loss or organ hypertrophy. In summary, we developed R848-TSLs that can be administered locally or systematically, resulting in tumor regression and enhanced survival when combined with αPD-1 in mouse models of breast cancer.


Assuntos
Hipertermia Induzida , Neoplasias , Animais , Imidazóis , Imunoterapia , Lipossomos , Camundongos , Neoplasias/tratamento farmacológico
6.
Pharmaceutics ; 11(9)2019 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-31505795

RESUMO

A large proportion of pharmaceutical compounds exhibit poor water solubility, impacting their delivery. These compounds can be passively encapsulated in the lipid bilayer of liposomes to improve their water solubility, but the loading capacity and stability are poor, leading to burst drug leakage. The solvent-assisted active loading technology (SALT) was developed to promote active loading of poorly soluble drugs in the liposomal core to improve the encapsulation efficiency and formulation stability. By adding a small volume (~5 vol%) of a water miscible solvent to the liposomal loading mixture, we achieved complete, rapid loading of a range of poorly soluble compounds and attained a high drug-to-lipid ratio with stable drug retention. This led to improvements in the circulation half-life, tolerability, and efficacy profiles. In this mini-review, we summarize our results from three studies demonstrating that SALT is a robust and versatile platform to improve active loading of poorly water-soluble compounds. We have validated SALT as a tool for improving drug solubility, liposomal loading efficiency and retention, stability, palatability, and pharmacokinetics (PK), while retaining the ability of the compounds to exert pharmacological effects.

7.
Drug Discov Today ; 23(5): 1159-1166, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29660478

RESUMO

A variety of nanoplatforms have been developed and applied for cancer therapy, imaging, or the combination thereof. These nanoplatforms, combined with therapeutic and imaging functionalities, display great potential to enhance medical care. In particular, lipid-based nanoparticles (LNPs) are among the most-studied platforms that have resulted in many encouraging advances in theranostics. LNPs are biodegradable and biocompatible, and their formulation can be tailored for various applications. Here, we provide an overview of recent developments of four representative LNP platforms for theranostics: stealth liposomes, triggered-release liposomes, porphysomes, and lipid-coated calcium phosphate NPs (LCPs). We discuss their potential, limitations, and potential applications for cancer care and highlight perspectives and future directions for the nanotheranostics field.


Assuntos
Fosfatos de Cálcio/administração & dosagem , Lipídeos/administração & dosagem , Nanopartículas/administração & dosagem , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Animais , Fosfatos de Cálcio/química , Humanos , Lipídeos/química , Lipossomos , Nanopartículas/química , Nanomedicina Teranóstica
8.
Biomaterials ; 166: 13-26, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29529479

RESUMO

The solvent-assisted active loading technology (SALT) was developed for encapsulating a water insoluble weak base into the liposomal core in the presence of 5% DMSO. In this study, we further examined the effect of various water miscible solvents in promoting active loading of other types of drugs into liposomes. To achieve complete drug loading, the amount of solvent required must result in complete drug solubilization and membrane permeability enhancement, but must be below the threshold that induces liposomal aggregation or causes bilayer disruption. We then used the SALT to load gambogic acid (GA, an insoluble model drug that shows promising anticancer effect) into liposomes, and optimized the loading gradient and lipid composition to prepare a stable formulation (Lipo-GA) that displayed >95% drug retention after incubation with serum for 3 days. Lipo-GA contained a high drug-to-lipid ratio of 1/5 (w/w) with a mean particle size of ∼75 nm. It also displayed a prolonged circulation half-life (1.5 h vs. 18.6 h) and enhanced antitumor activity in two syngeneic mice models compared to free GA. Particularly, complete tumor regression was observed in the EMT6 tumor model for 14 d with significant inhibition of multiple oncogenes including HIF-1α, VEGF-A, STAT3, BCL-2, and NF-κB.


Assuntos
Lipossomos , Solventes , Xantonas , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Composição de Medicamentos/métodos , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Lipossomos/química , Camundongos , Camundongos Endogâmicos BALB C , Solventes/química , Xantonas/farmacologia
9.
Biomaterials ; 33(5): 1445-54, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22079003

RESUMO

We have developed a polymer conjugate (Cellax) composed of acetylated carboxymethylcellulose (CMC), docetaxel (DTX), and PEG, designed to enhance the pharmacokinetics (PK) and antitumor efficacy of DTX. Our design placed an emphasis on nanoparticle self-assembly to protect DTX during blood transport, stability of the nanoparticle, and PEGylation to enhance PK. Compared to Taxotere, Cellax exhibited a 38.6 times greater area under the curve (AUC), and significantly lower clearance (2.5%) in PK. Less than 10% of DTX was released from Cellax in the blood circulation, indicating that Cellax were stable during blood transport. Cellax reduced non-specific distribution of DTX to the heart, lung and kidney by 48, 90, and 90%, respectively, at 3 h, compared to Taxotere. The uptake of Cellax at 3 h in the liver and spleen was high (15-45 µg DTX/g) but declined rapidly to <10 µg DTX/g in 24 h, and induced no measurable toxicity at 170 mg DTX/kg. Taxotere, on the other hand, displayed non-specific uptake in all the examined normal tissues and induced significant apoptosis in the lung and kidney at 40 mg DTX/kg. The tumor uptake of Cellax was 5.5-fold more than that by Taxotere and the uptake occurred within 3 h after injection and persisted for 10 days. The conjugate exhibited enhanced efficacy in a panel of primary and metastatic mouse tumor models. These results clearly demonstrated that Cellax improved the pharmacokinetics, biodistribution and efficacy of DTX compared to Taxotere with reduced toxicity.


Assuntos
Antineoplásicos/uso terapêutico , Carboximetilcelulose Sódica/química , Nanopartículas/química , Neoplasias/tratamento farmacológico , Taxoides/farmacocinética , Taxoides/uso terapêutico , Animais , Antineoplásicos/farmacocinética , Modelos Animais de Doenças , Docetaxel , Ensaios de Seleção de Medicamentos Antitumorais , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Neoplasias/patologia , Polietilenoglicóis/química , Polímeros/síntese química , Polímeros/química , Taxoides/sangue , Distribuição Tecidual/efeitos dos fármacos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA