Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diseases ; 11(4)2023 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-38131983

RESUMO

The COVID-19 pandemic has had a profound impact on societies, public health, healthcare systems, and the world economy. With over 771 million people infected worldwide and a staggering death toll exceeding 6,960,783 as of 4 October 2023 (according to the World Health Organization), the urgency for a solution was paramount. Since the outbreak, the demand for immediate treatment for COVID-19 viral infection, as well as for effective vaccination against this virus, was soaring, which led scientists, pharmaceutical/biotech companies, government health agencies, etc., to think about a treatment strategy that could control and minimize this outbreak as soon as possible. Vaccination emerged as the most effective strategy to combat this infectious disease. For vaccination strategies, any conventional vaccine approach using attenuated live or inactivated/engineered virus, as well as other approaches, typically requires years of research and assessment. However, the urgency of the situation promoted a faster and more effective approach to vaccine development against COVID-19. The role of nanotechnology in designing, manufacturing, boosting, and delivering vaccines to the host to counter this virus was unquestionably valued and assessed. Several nanoformulations are discussed here in terms of their composition, physical properties, credibility, and applications in past vaccine development (as well as the possibility of using those used in previous applications for the generation of the COVID-19 vaccine). Controlling and eliminating the spread of the virus and preventing future recurrence requires a safe, tolerable, and effective vaccine strategy. In this review, we discuss the potential of nanoformulations as the basis for an effective vaccine strategy against COVID-19.

2.
Water Environ Res ; 95(5): e10871, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37096740

RESUMO

Poor reactivity of extraordinarily strong per- and polyfluoroalkyl substances (PFAS) with TiO2 makes it necessary to advance photocatalytic materials. In this present work, lead (Pb)-doped TiO2 coated with reduced graphene oxide (TiO2 -Pb/rGO) was prepared using hydrothermal method, and then its photocatalytic activity with various PFAS in water, especially perfluorooctanoic acid (PFOA), was investigated. PFAS decomposition kinetics by TiO2 -Pb/rGO was compared with neat TiO2 , Pb-doped TiO2 (TiO2 -Pb), and rGO-coated TiO2 (TiO2 /rGO). TiO2 -Pb/rGO (0.33 g/L) under ultraviolet (UV) showed superior removal of PFOA (10 mg/L) at 98% after 24 h, following TiO2 -Pb/UV at 80%, TiO2 /rGO/UV at 70%, TiO2 /UV at <10%, and UV at <10%. Doping of TiO2 with Pb and introduction of rGO to TiO2 greatly changed the physicochemical properties of TiO2 and the subsequent charge transfer mechanism. Radical scavenger experiments indicated that holes, superoxide radical anion, and singlet oxygen were responsible for the observed PFOA decomposition. Decomposition of PFOA by TiO2 -Pb/rGO under UV led to formation of short-chain perfluorocarboxylic acids (PFCAs) as reaction intermediates through step-by-step removal of CF2 units. Polyfluoroalkyl substance (6:2 fluorotelomer sulfonate [6:2FTS]) and long-chain PFCAs such as PFOA were significantly removed and defluorinated by TiO2 -Pb/rGO, whereas it was ineffective toward perfluorosulfonic acids and short-chain PFCAs. Removal kinetics decreased in the order of 6:2FTS > PFOA >> PFOS > PFHpA ≈ PFHxS ≈ PFBA ≈ PFBS. Pb doping to TiO2 /rGO showed better performance than Fe doping. Overall, this study implied that proper designing of TiO2 photocatalytic materials enables to expedite the decomposition of persistent organic pollutants in water, in particular highly challenging fluorinated chemicals. PRACTITIONER POINTS: Photocatalytic decomposition of various PFAS using TiO2 -Pb/rGO was studied. TiO2 -Pb/rGO shows better photoactivity towards PFAS than TiO2 -Pb and TiO2/rGO system. Scavenger test indicated that h+, • O2-, and iO2 are responsible for PFOA removal. Using TiO2 -Pb/rGO, PFOA removal was comparable under UVA, UVB, and UVC, and it can be explained by spanning UV absorption to 415 nm. Formation of intermediate PFCAs and F- ions confirmed PFOA removal via chemical decomposition.


Assuntos
Fluorocarbonos , Poluentes Químicos da Água , Chumbo , Fluorocarbonos/química , Água , Poluentes Químicos da Água/química
3.
Water Sci Technol ; 84(12): 3738-3754, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34928840

RESUMO

This study reports the removal of per- and polyfluoroalkyl substances (PFAS) in water using various photocatalytic and photochemical processes. PFAS were chosen, based on chain lengths, functional groups, and structural properties: four perfluorocarboxylic acids (PFCAs), including perfluorooctanoic acid (PFOA), three perfluorosulfonic acids (PFSAs), including perfluorooctanesulfonic acid (PFOS), hexafluoropropylene oxide dimer (GenX), and 6:2 fluorotelomer sulfonate (6:2 FTS), and dependency of the photocatalytic decomposition of PFAS on their properties was investigated. Oxidants and reductants were introduced to study the photochemical decomposition of PFAS, and reactive species and reaction byproducts were identified to elucidate the decomposition mechanism of PFAS. Some notable findings include: long chain PFCAs (95% in 48 h) and 6:2 FTS (100%) were removed via chemical decomposition in TiO2/UVC while GenX (37%), long chain PFSAs (60%), short chain PFSAs (0-10%) and short chain PFCAs (5-18%) were removed via physical adsorption. Sulfate radicals generated with persulfate (PS) played an important role in decomposing PFCAs (60-90%). Sulfite activated by UVC worked for defluorination of PFOA (75%) and PFOS (80%). PFOA was removed faster by UVC/sulfite > UVC/TiO2/sulfite ≈ UVC/TiO2/PS ≥ UVC/PS > UVC/TiO2 while PFOS was removed faster by UVC/sulfite ≫ UVC/TiO2/sulfite ≈ UVC/TiO2/PS ≈ UVC/TiO2 ≫ UVC/PS. Susceptibility of PFAS to the chemical reactions could be explained by their properties and the reactive species produced in each system.


Assuntos
Fluorocarbonos
4.
Mater Sci Eng C Mater Biol Appl ; 119: 111460, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33321591

RESUMO

Glycoalkaloids have been widely demonstrated as potential anticancer agents. However, the chemosensitizing effect of these compounds with traditional chemotherapeutic agents has not been explored yet. In a quest for novel effective therapies to treat bladder cancer (BC), we evaluated the chemosensitizing potential of glycoalkaloidic extract (GE) with cisplatin (cDDP) in RT4 and PDX cells using 2D and 3D cell culture models. Additionally, we also investigated the underlying molecular mechanism behind this effect in RT4 cells. Herein, we observed that PDX cells were highly resistant to cisplatin when compared to RT4 cells. IC50 values showed at least 2.16-folds and 1.4-folds higher in 3D cultures when compared to 2D monolayers in RT4 cells and PDX cells, respectively. GE + cDDP inhibited colony formation (40%) and migration (28.38%) and induced apoptosis (57%) in RT4 cells. Combination therapy induced apoptosis by down-regulating the expression of Bcl-2 (p < 0.001), Bcl-xL (p < 0.001) and survivin (p < 0.01), and activating the caspase cascade in RT4 cells. Moreover, decreased expression of MMP-2 and 9 (p < 0.01) were observed with combination therapy, implying its effect on cell invasion/migration. Furthermore, we used 3D bioprinting to grow RT4 spheroids using sodium alginate-gelatin as a bioink and evaluated the effect of GE + cDDP on this system. Cell viability assay showed the chemosensitizing effect of GE with cDDP on bio-printed spheroids. In summary, we showed the cytotoxicity effect of GE on BC cells and also demonstrated that GE could sensitize BC cells to chemotherapy.


Assuntos
Antineoplásicos , Neoplasias da Bexiga Urinária , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Xenoenxertos , Humanos , Extratos Vegetais/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico
5.
Crit Rev Ther Drug Carrier Syst ; 37(3): 205-227, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32749138

RESUMO

In this review, we describe the advances in oral drug delivery approaches for taxanes for successful therapeutic outcome. Taxanes (paclitaxel and docetaxel) have unwanted pharmacokinetic profiles when they are given in their current dosage forms. Taxanes have low bioavailability, are extensively metabolized by CYP3A, and have a high affinity for P-glycoprotein. Regardless of dosage schedule, the overall docetaxel or paclitaxel dose that a patient can tolerate at a given interval remains similar. Currently, there are no commercially available oral taxane nanoformulations, and there are still several challenges to overcome. Nano-based formulations may offer the best solutions to problems involving the safety and effectiveness of taxane delivery. Thus, further research is necessary before such taxane nanoformulations can be manufactured for clinical use.


Assuntos
Docetaxel/administração & dosagem , Paclitaxel/administração & dosagem , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacocinética , Ensaios Clínicos como Assunto , Docetaxel/química , Docetaxel/farmacocinética , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Composição de Medicamentos , Sistemas de Liberação de Medicamentos , Humanos , Lipídeos/administração & dosagem , Lipídeos/química , Micelas , Nanopartículas/administração & dosagem , Nanopartículas/química , Paclitaxel/química , Paclitaxel/farmacocinética
6.
AAPS PharmSciTech ; 21(6): 202, 2020 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-32696338

RESUMO

The adverse side effects and toxicity caused by the non-targeted delivery of doxorubicin has emphasized the demand of emerging a targeted delivery system. The goal of this study is to enhance the delivery of doxorubicin by formulating an aptamer-labeled liposomal nanoparticle delivery system that will carry and deliver doxorubicin specifically into Her-2+ breast cancer cells. Twelve liposomal batches were prepared using different saturated (HSPC and DPPC) and unsaturated (POPC and DOPC) lipids by thin film hydration. The liposomes were characterized for their particle size, zeta potential, and drug encapsulation efficiency. The particles were also assessed for in vitro toxicity and DOX delivery into the breast cancer cells. The formulations, F1 through F12, had a small particle size of less than 200 nm and a high entrapment efficiency of about 88 ± 5%. The best formulation, F5, had a particle size of 101 ± 14nm, zeta potential of + 5.63 ± 0.46 mV, and entrapment efficiency of ≈ 93%. The cytotoxicity studies show that the DOX-loaded liposomal formulations are more effective in killing cancer cells than the free DOX in both MCF-7 and SKBR-3 cells. The uptake studies show a significant increase in the uptake of the aptamer-labeled liposomes (i.e., F5) by more than 60% into Her-2+ MCF-7 and SKBR-3 breast cancer cells compare to non-aptamer-labeled nanoparticles. F5 also shows ≈ 1.79-fold increase in uptake of DOX in the Her-2+ cells compared to the Her-2- cells. This preliminary study indicates that aptamer-labeled F5 nanoparticles among several batches showed the highest uptake as well as the targeted delivery of doxorubicin into Her-2+ breast cancer cells. Thus, aptamer targeted approach results in substantial reduction in the dose of DOX and improves the therapeutic benefits by promoting the target specificity.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/análogos & derivados , Sistemas de Liberação de Medicamentos , Receptor ErbB-2/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Feminino , Humanos , Lipossomos , Nanopartículas , Polietilenoglicóis/administração & dosagem
7.
AAPS PharmSciTech ; 20(6): 240, 2019 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-31250221

RESUMO

Sunscreens are widely prescribed and used to prevent skin cancer; however, they have been reported to contain various chemicals which mimic hormones and disrupt hormonal functioning in humans. The aim of this study was to develop topical nanogel for skin cancer prevention using an antioxidant compound quercetin (Qu) and inorganic titanium dioxide (TiO2). Two formulations of Qu nanocrystals were optimized with low and high concentration of drug using the Box-Behnken design with the quadratic response surface model and further homogenized with TiO2. Qu nanocrystal (0.08% and 0.12%) formulations showed a particle size of 249.65 ± 2.84 nm and 352.48 ± 3.56 nm with zeta potential of - 14.7 ± 0.41 mV and - 19.6 ± 0.37 mV and drug content of 89.27 ± 1.39% and 90.38 ± 1.81% respectively. Scanning electron microscopy (SEM) images showed rod-shaped nanocrystals with a particle size below 400 nm. Qu (0.08%), Qu (0.12%), Qu (0.12%) + TiO2 (5%), and Qu (0.12%) + TiO2 (15%) nanogels showed over 70% drug release with significantly (p < 0.001) enhanced skin deposition of Qu as compare with Qu suspension within 24 h. The average numbers of tumor, tumor volume, and percentage of animals with tumors at onset in the Qu (0.12%) + TiO2 (15%) nanogel-pretreated group was found to be significantly (p < 0.05) less as compared with the UV only exposed group. Further, Qu (0.12%) + TiO2 (15%) nanogel significantly (p < 0.001) downregulated COX-2, EP3, EP4, PCNA, and cyclin D1 expressions in contrast to Qu and TiO2 only pretreated groups. Therefore, novel combination of Qu (0.12%) + TiO2 (15%) with enhanced skin deposition can be used as a chemopreventive strategy in UVB-induced skin photocarcinogenesis.


Assuntos
Quimioprevenção/métodos , Géis/administração & dosagem , Nanoestruturas/administração & dosagem , Neoplasias Induzidas por Radiação/prevenção & controle , Quercetina/farmacologia , Neoplasias Cutâneas/prevenção & controle , Pele/efeitos dos fármacos , Protetores Solares/farmacologia , Titânio/farmacologia , Raios Ultravioleta , Administração Tópica , Animais , Combinação de Medicamentos , Liberação Controlada de Fármacos , Humanos , Tamanho da Partícula , Polietilenoglicóis/química , Quercetina/administração & dosagem , Quercetina/química , Pele/metabolismo , Protetores Solares/administração & dosagem , Titânio/administração & dosagem
8.
Drug Deliv Transl Res ; 9(4): 816-827, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30924025

RESUMO

Solid lipid nanoparticles (SLN) have been formulated using various batch processes, e.g., solvent diffusion evaporation, emulsification solvent evaporation followed by size reduction using high-pressure homogenization (HPH) or ultrasonication. However, for the manufacturing of formulations, continuous processes are always preferred over batch processes since they are more efficient and offer better quality of the end product. Hence, we developed topical SLN of ibuprofen (IBU) using hot melt extrusion (HME), prepared a gel formulation, and performed its in vitro and in vivo evaluation. Effect of different variables of HME equipment and materials used in SLN was optimized using design of experiment (DoE) approach. Stable 0.48% IBU SLN with particle size 60.2 ± 4.81 nm and entrapment efficiency 90.41 ± 3.46% were developed which further gelled using 1% carbopol 981A. Drug release study, skin deposition study, and in vivo anti-inflammatory activity studies showed 84.37 ± 4.65% drug release, 12.05 ± 0.81% drug deposition, and 40.17 ± 2.41% edema inhibition respectively in case of IBU SLN gel (IBU-SLN-G) which was significantly higher (p < 0.05) than control IBU gel (C-IBU-G) with 50.11 ± 0.57% drug release, 4.11 ± 1.10% deposition, and 20.08 ± 3.23% edema inhibition respectively. In conclusion, HME offers a single step process for manufacturing for SLN which avoids high stress particle size reduction techniques used for SLN preparation.


Assuntos
Anti-Inflamatórios não Esteroides , Portadores de Fármacos , Edema/tratamento farmacológico , Ibuprofeno , Nanopartículas , Administração Cutânea , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/química , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Tecnologia de Extrusão por Fusão a Quente , Humanos , Ibuprofeno/administração & dosagem , Ibuprofeno/química , Lipídeos/administração & dosagem , Lipídeos/química , Nanopartículas/administração & dosagem , Nanopartículas/química , Ratos Sprague-Dawley , Pele/metabolismo , Absorção Cutânea , Tecnologia Farmacêutica
9.
AAPS PharmSciTech ; 19(7): 3287-3297, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30218267

RESUMO

Primary standard therapy for ER-positive breast cancer being tamoxifen, newer delivery approach for enhancement of dissolution and therapeutic efficiency of tamoxifen through oral route could be a possible solution. In the present study, we investigated combination of tamoxifen (TAM) with resveratrol (RES) and observed that the combination is effective on MCF-7 breast cancer cells. To ensure co-delivery of the drugs, we explored the hot melt extrusion technique for simultaneously extruding two drugs together in order to enhance their bioavailability. As both are class II drugs with dissolution limited bioavailability, detailed formulation and process parameter analyses were carried out. Detailed characterization using microscopy, Fourier transform infrared spectroscopy (FTIR), differential scanning calorimetry (DSC), and X-ray powder diffraction (XRD) confirmed that both the drugs were molecularly dispersed in the matrix of Soluplus, CremophorRH40, and Poloxamer188, and no interactions between the ingredients were there during hot melt extrusion (HME) process. Dissolution studies confirmed that HME extrudates were able to release drug more rapidly than simple suspension formulation. Further, pharmacokinetic studies in rats were carried out for tamoxifen. Results demonstrated that extrusion significantly increased the tamoxifen oral bioavailability (p < 0.05) (Tmax = 2.00 ± 0.56 h, Cmax = 3.66 ± 1.49 µg/mL, AUC = 39.80 ± 16.24 µg h/mL, MRT = 20.49 ± 5.71) compared to the conventional suspension of tamoxifen (Tmax = 2.00 ± 0.71 h, Cmax = 2.41 ± 0.84 µg/mL, AUC = 12.82 ± 3.99 µg h/mL, MRT = 18.24 ± 5.95 h). In vitro cytotoxicity studies of TAM, RES, and their combination (TAM-RES) were evaluated with MCF7 cells. The combination showed significantly lower IC50 compared to TAM with increasing ratio of RES which is a result of apoptosis. HME-based simultaneous extrusion of TAM and RES formulation provides a suitable formulation strategy for breast cancer treatment and establishes proof of concept for extruding multiple drugs simultaneously for other applications in future.


Assuntos
Antineoplásicos Hormonais/administração & dosagem , Antineoplásicos Fitogênicos/administração & dosagem , Neoplasias da Mama , Desenvolvimento de Medicamentos/métodos , Resveratrol/administração & dosagem , Tamoxifeno/administração & dosagem , Administração Oral , Animais , Antineoplásicos Hormonais/química , Antineoplásicos Hormonais/metabolismo , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Varredura Diferencial de Calorimetria/métodos , Química Farmacêutica/métodos , Sinergismo Farmacológico , Temperatura Alta , Humanos , Células MCF-7 , Ratos , Ratos Sprague-Dawley , Resveratrol/química , Resveratrol/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Tamoxifeno/química , Tamoxifeno/metabolismo , Difração de Raios X/métodos
10.
Pharm Res ; 35(6): 117, 2018 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-29663141

RESUMO

PURPOSE: Poor corneal permeability, nasolacrimal drainage and requirement of chronic administration are major drawbacks of existing therapies for ocular inflammation. Hence, we designed topical micelles of PEG2000 conjugated with cholecalciferol (PEGCCF). METHODS: Integrin targeted tacrolimus loaded PEGCCF micelles (TTM) were prepared by solvent diffusion evaporation method and characterized for particle size, osmolality, encapsulation efficiency and drug loading. Therapeutic potential of TTM was evaluated in benzalkonium chloride induced ocular inflammation model in BALB/c mice. Corneal flourescein staining and histopathological analysis of corneal sections was performed. RESULTS: TTM had a particle size of 45.3 ± 5.3 nm, encapsulation efficiency (88.7 ± 0.9%w/w) and osmolality of 292-296 mOsmol/Kg. TTM significantly reduced the corneal fluorescence as compared to tacrolimus suspension (TACS). H&E staining showed that TTM could restore corneal epithelial thickness, reduce stromal edema (p < 0.05) and decrease number of inflammatory cells (p < 0.01) compared with TACS. Immunohistochemistry analysis demonstrated lower expression of Ki67 + ve cells (p < 0.05) and IL-6 throughout the cornea against TACS (p < 0.01) and the control (p < 0.001). CONCLUSIONS: TTM is an innovative delivery system for improving ocular inflammation due to a) integrin targeting b) PEGCCF in the form of carrier and c) anti-inflammatory and synergistic effect (due to Pgp inhibition) with TAC.


Assuntos
Portadores de Fármacos/química , Oftalmopatias/tratamento farmacológico , Inflamação/tratamento farmacológico , Tacrolimo/administração & dosagem , Administração Oftálmica , Animais , Compostos de Benzalcônio/toxicidade , Colecalciferol/química , Modelos Animais de Doenças , Composição de Medicamentos/métodos , Olho/efeitos dos fármacos , Olho/patologia , Oftalmopatias/induzido quimicamente , Oftalmopatias/patologia , Feminino , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Integrinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Micelas , Polietilenoglicóis/química , Tacrolimo/farmacocinética
11.
AAPS PharmSciTech ; 19(2): 792-802, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29019073

RESUMO

Triple-negative breast cancer (TNBC) is the leading cancer in women. Chemotherapeutic agents used for TNBC are mainly associated with dose-dependent toxicities and development of resistance. Hence, novel strategies to overcome resistance and to offer dose reduction are warranted. In this study, we designed a novel dual-functioning agent, conjugate of cholecalciferol with PEG2000 (PEGCCF) which can self-assemble into micelles to encapsulate doxorubicin (DOX) and act as a chemosensitizer to improve the therapeutic potential of DOX. DOX-loaded PEGCCF (PEGCCF-DOX) micelles have particle size, polydispersity index (PDI), and zeta potential of 40 ± 8.7 nm, 0.180 ± 0.051, and 2.39 ± 0.157 mV, respectively. Cellular accumulation studies confirmed that PEGCCF was able to concentration-dependently enhance the cellular accumulation of DOX and rhodamine 123 in MDA-MB-231 cells through its P-glycoprotein (P-gp) inhibition activity. PEGCCF-DOX exhibited 1.8-, 1.5-, and 2.9-fold enhancement in cytotoxicity of DOX in MDA-MB-231, MDA-MB-468, and MDA-MB-231DR (DOX-resistant) cell lines, respectively. Western blot analyses showed that PEGCCF-DOX caused significant reduction in tumor markers including mTOR, c-Myc, and antiapoptotic marker Bcl-xl along with upregulation of preapoptotic marker Bax. Further, reduction in mTOR activity by PEGCCF-DOX indicates reduced P-gp activity due to P-gp downregulation as well and, hence, PEGCCF causes enhanced chemosensitization and induces apoptosis. Substantially enhanced apoptotic activity of DOX (10-fold) in MDA-MB-231(DR) cells confirmed apoptotic potential of PEGCCF. Conclusively, PEGCCF nanomicelles are promising delivery systems for improving anticancer activity of DOX in TNBC, thereby reducing its side effects and may act as a potential carrier for other chemotherapeutic agents.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Colecalciferol/química , Doxorrubicina/administração & dosagem , Portadores de Fármacos/química , Polietilenoglicóis/química , Neoplasias de Mama Triplo Negativas/metabolismo , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Liberação Controlada de Fármacos , Feminino , Humanos , Micelas , Nanoestruturas , Tamanho da Partícula , Serina-Treonina Quinases TOR/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
12.
Sci Rep ; 7(1): 15824, 2017 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-29158480

RESUMO

Multidrug resistance (MDR) is a major impediment to cancer treatment. Here, for the first time, we investigated the chemo-sensitizing effect of Noscapine (Nos) at low concentrations in conjunction with docetaxel (DTX) to overcome drug resistance of triple negative breast cancer (TNBC). In vitro experiments showed that Nos significantly inhibited proliferation of TNBC wild type (p < 0.01) and drug resistant (p < 0.05) TNBC cells. Nos followed by DTX treatment notably increased the cell viability (~1.3 fold) markedly (p < 0.05) in 3D models compared to conventional 2D systems. In vivo oral administration of Nos (100 mg/kg) followed by intravenous DTX (5 mg/kg) liposome treatment revealed regression of xenograft tumors in both wild type (p < 0.001) and drug-resistant (p < 0.05) xenografts. In wild type xenografts, combination of Nos plus DTX group showed 5.49 and 3.25 fold reduction in tumor volume compared to Nos and DTX alone groups, respectively. In drug-resistant xenografts, tumor volume was decreased 2.33 and 1.41 fold in xenografts treated with Nos plus DTX significantly (p < 0.05) compared to Nos and DTX alone respectively and downregulated the expression of anti-apoptotic factors and multidrug resistance proteins. Collectively, chemo-sensitizing effect of Nos followed by DTX regime provide a promising chemotherapeutic strategy and its significant role for the treatment of drug-resistant TNBC.


Assuntos
Docetaxel/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Noscapina/administração & dosagem , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Docetaxel/efeitos adversos , Feminino , Humanos , Lipossomos/administração & dosagem , Lipossomos/química , Camundongos , Taxoides/administração & dosagem , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Pharm Res ; 34(11): 2371-2384, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28875330

RESUMO

PURPOSE: Non-small cell lung cancer is the leading cause of cancer related deaths globally. Considering the side effects and diminishing chemosensitivity to chemotherapy, novel treatment approaches are sought. Hence, we aim to develop a liposomal co-delivery system of pDNA expressing shRNA against PFKFB3 (pshPFKFB3) and docetaxel (DTX). METHODS: Cationic DTX liposomes complexed with pshPFKFB3 (PSH-DL) were developed. In vitro cell line studies were performed to evaluate transfection, PFKFB3 mRNA silencing, cytotoxicity, pGP inhibition, and protein markers expression. In vivo efficacy study was performed in A549 xenograft nude mice model. RESULTS: Cytotoxicity studies showed significantly enhanced anticancer activity of PSH-DL against individual treatment alone confirming the chemoenhancing effect of pshPFKFB3 on DTX activity. Fluorescence microscopy and RT-PCR showed effective transfection and RNAi by pshPFKFB3. pGP inhibition assay and western blotting revealed that PFKFB3 downregulation caused diminution of pGP activity leading to changes in cell cycle (Cdk2), survival (survivin), apoptosis (Bcl2 and cleaved caspase 3) and stress (p-JNK and p-p38) markers so that induces apoptosis by PSH-DL in NSCLC cells. PSH-DL also showed ~3.8-fold reduction in tumor volume in A549 xenograft model which was significantly higher than individual treatments alone. CONCLUSION: Targeting PFKFB3 through shRNA based RNAi is a promising approach for potentiating activity of DTX in NSCLC.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Lipossomos/química , Neoplasias Pulmonares/tratamento farmacológico , Fosfofrutoquinase-2/genética , RNA Interferente Pequeno/genética , Taxoides/farmacologia , Animais , Antineoplásicos/química , Apoptose , Caspase 3/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Docetaxel , Combinação de Medicamentos , Inativação Gênica , Técnicas de Transferência de Genes , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Camundongos , Camundongos Nus , Tamanho da Partícula , Fosfofrutoquinase-2/metabolismo , Plasmídeos , Complexo de Inativação Induzido por RNA/metabolismo , Propriedades de Superfície , Taxoides/química , Carga Tumoral/efeitos dos fármacos
14.
Oncol Lett ; 14(2): 2341-2346, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28781672

RESUMO

A mechanistic dissociation exists between tumor starvation and vascular normalization after antiangiogenic therapy. Thus, improved understanding of tumor responses (tumor starvation or vascular normalization) is important for optimizing treatment strategies. 18F-fluoromisonidazole (18F-FMISO) is widely used for imaging tumor hypoxia. To clarify the tumor response to the antiangiogenic drug sorafenib, the present study evaluated the changes in the tumor oxygen state using 18F-FMISO in mice bearing a renal cell carcinoma xenograft (A498). Mice bearing A498 xenografts were assigned to the control and three sorafenib-treatment groups and administered sorafenib (0, 10, 20 or 40 mg/kg/day, per os) once daily for 3 days. Following one day after the final administration, the mice were injected with 18F-FMISO and pimonidazole (a hypoxia marker). 18F-FMISO accumulation in the tumor was determined by autoradiography. Immunohistochemistry of pimonidazole and cluster of differentiation (CD)31 (a vascular marker) was also performed. 18F-FMISO accumulation levels in the tumor significantly increased by 4.3-, 8.4- and 8.6-fold compared with in the control group following 10, 20 and 40 mg/kg sorafenib treatments, respectively [0.07±0.04, 0.32±0.11, 0.62±0.15 and 0.63±0.23 (%ID/m2) × kg for the control, and 10, 20 and 40 mg treatments, respectively; all P<0.0083 vs. the control]. The number of pimonidazole-positive cells also significantly increased by 6.8-, 12.3- and 20.2-fold compared with in the control group following 10, 20 and 40 mg/kg sorafenib treatments, respectively (0.78±0.79, 5.36±2.29, 9.66±1.58 and 15.85±4.59% pimonidazole-positive cells; all P<0.0083 vs. the control). The number of microvessels in tumors markedly decreased to 33.5, 17.6, and 14.0% of the control following 10, 20 and 40 mg/kg sorafenib treatments, respectively (17.1±2.5, 5.7±1.0, 3.0±1.0 and 2.4±0.3 vessels/mm2; P<0.0083 vs. the control). The 18F-FMISO expression level in the tumor increased sorafenib-dose-dependently, which is consistent with the increase in the number of pimonidazole-positive cells and decrease in the number of microvessels. These findings indicated that the present sorafenib treatment protocol induces 'tumor hypoxia/starvation' in the renal cell carcinoma xenograft (A498) due to its antiangiogenic properties.

15.
Exp Cell Res ; 346(1): 65-73, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27177833

RESUMO

Chemosensitization and enhanced delivery to solid tumor are widely explored strategies to augment the anticancer efficacy of existing chemotherapeutics agents. The aim of current research was to investigate the role of low dose Noscapine (Nos) in potentiating docetaxel cytotoxicity and enhancing tumor penetration of nanocarriers. The objectives are; (1) To evaluate the chemo-sensitizing effect of Nos in combination with docetaxel (DTX), and to elucidate the possible mechanism (2) To investigate the effect of low dose Nos on tumor stroma and enhancing nanocarrier uptake in triple negative breast cancer (TNBC) bearing nude mice. Cytotoxicity and flow cytometry analysis of DTX in Nos (4µM) pre-treated MDA-MB-231 cells showed 3.0-fold increase in cell killing and 30% increase in number of late apoptotic cells, respectively. Stress transducer p38 phosphorylation was significantly upregulated with Nos exposure. DTX showed remarkable downregulation in expression of bcl-2, survivin and pAKT in Nos pre-treated MDA-MB-231 cells. Nos pre-sensitization significantly (p<0.02) enhanced the anti-migration effect of DTX. In vivo studies in orthotopic TNBC tumor bearing mice showed marked reduction in tumor collagen-I levels and significantly (p<0.03) higher intra-tumoral uptake of coumarin-6 loaded PEGylated liposomes (7-fold) in Nos treated group. Chemo-sensitization and anti-fibrotic effect of Nos could be a promising approach to increase anticancer efficacy of DTX which can be used for other nanomedicinal products.


Assuntos
Antineoplásicos/farmacologia , Portadores de Fármacos/química , Nanopartículas/química , Noscapina/farmacologia , Taxoides/farmacologia , Neoplasias de Mama Triplo Negativas/patologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaios de Migração Celular , Movimento Celular/efeitos dos fármacos , Cumarínicos/metabolismo , Docetaxel , Ativação Enzimática/efeitos dos fármacos , Imunofluorescência , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipossomos , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Fosforilação/efeitos dos fármacos , Polimerização/efeitos dos fármacos , Coloração e Rotulagem , Tiazóis/metabolismo , Tubulina (Proteína)/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Nanomedicine (Lond) ; 11(11): 1377-92, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27171485

RESUMO

AIM: Therapeutic efficacy of anticancer nanomedicine is compromised by tumor stromal barriers. The present study deals with the development of docetaxel loaded PEGylated liposomes (DTXPL) and to investigate the effect of tumor stroma disrupting agent, telmisartan, on anticancer efficacy of DTXPL. METHODS: DTXPL was prepared using proprietary modified hydration method. Effect of oral telmisartan treatment on tumor uptake of coumarin-6 liposomes and anticancer efficacy of DTXPL was evaluated in orthotopic xenograft lung tumor bearing mice. RESULTS: DTXPL (105.7 ± 3.8 nm) showed very high physical stability, negligible hemolysis, 428% enhancement in bioavailability with significantly higher intratumoral uptake. Marked reduction in collagen-I, MMP2/9 and lung tumor weight were observed in DTXPL+telmisartan group. CONCLUSION: Combination of DTXPL with telmisartan could significantly enhance clinical outcome in lung cancer.


Assuntos
Antineoplásicos/farmacologia , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Polietilenoglicóis/química , Taxoides/farmacologia , Células A549 , Animais , Antineoplásicos/administração & dosagem , Benzimidazóis/administração & dosagem , Benzoatos/administração & dosagem , Cumarínicos/química , Docetaxel , Feminino , Xenoenxertos , Humanos , Lipossomos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Tamanho da Partícula , Ratos , Ratos Sprague-Dawley , Propriedades de Superfície , Taxoides/administração & dosagem , Taxoides/farmacocinética , Telmisartan , Tiazóis/química , Carga Tumoral/efeitos dos fármacos
18.
Mol Pharm ; 13(6): 2049-58, 2016 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-27070720

RESUMO

Therapeutic efficacy of nanocarriers can be amplified by active targeting and overcoming the extracellular matrix associated barriers of tumors. The aim of the present study was to investigate the effect of oral antifibrotic agent (telmisartan) on tumor uptake and anticancer efficacy of EphA2 receptor targeted liposomes. Docetaxel loaded PEGylated liposomes (DPL) functionalized with nickel chelated phospholipid were prepared using a modified hydration method. DPL were incubated with various concentrations of histidine tagged EphA2 receptor specific peptide (YSA) to optimize particle size, zeta potential, and percentage YSA binding. Cellular uptake studies using various endocytosis blockers revealed that a caveolae dependent pathway was the major route for internalization of YSA anchored liposomes of docetaxel (YDPL) in A549 lung cancer cell line. Hydrodynamic diameter and zeta potential of optimized YDPL were 157.3 ± 11.8 nm and -3.64 mV, respectively. Orthotopic lung tumor xenograft (A549) bearing athymic nude mice treated with oral telmisartan (5 mg/kg) for 2 days showed significantly (p < 0.05) higher uptake of YDPL in tumor tissues compared to healthy tissue. Average lung tumor weight of the YDPL + telmisartan treated group was 4.8- and 3.8-fold lower than that of the DPL and YDPL treated groups (p < 0.05). Substantially lower expression (p < 0.05) of EphA2 receptor protein, proliferating cell nuclear antigen (PCNA), MMP-9, and collagen 1A level with increased E-cadherin and TIMP-1 levels in immunohistochemistry and Western blot analysis of lung tumor samples of the combination group confirmed antifibrotic effect with enhanced anticancer activity. Active targeting and ECM remodeling synergistically contributed to anticancer efficacy of YDPL in orthotopic lung cancer.


Assuntos
Antineoplásicos/farmacologia , Lipossomos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Peptídeos/farmacologia , Taxoides/farmacologia , Células A549 , Animais , Antineoplásicos/química , Caderinas/metabolismo , Linhagem Celular Tumoral , Colágeno/metabolismo , Docetaxel , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Nus , Peptídeos/química , Polietilenoglicóis/química , Antígeno Nuclear de Célula em Proliferação/metabolismo , Receptor EphA2/metabolismo , Taxoides/química , Inibidor Tecidual de Metaloproteinase-1/metabolismo
19.
Colloids Surf B Biointerfaces ; 143: 156-167, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-27003466

RESUMO

PURPOSE: In this study, we developed cationic ultra-flexible nanocarriers (UltraFLEX-Nano) to surmount the skin barrier structure and to potentiate the topical delivery of a highly lipophilic antioxidative diindolylmethane derivative (DIM-D) for the inhibition of UV-induced DNA damage and skin carcinogenesis. METHODS: UltraFLEX-Nano was prepared with 1,2-dipalmitoyl-sn-glycero-3-phosphocholine, 1,2-dioleoyl-3-trimethylammonium-propane, cholesterol and tween-80 by ethanolic injection method; was characterized by Differential Scanning Calorimetric (DSC), Fourier Transform Infrared (FT-IR) and Atomic Force Microscopic (phase-imaging) analyses and permeation studies were performed in dermatomed human skin. The efficacy of DIM-D-UltraFLEX-Nano for skin cancer chemoprevention was evaluated in UVB-induced skin cancer model in vivo. RESULTS: DIM-D-UltraFLEX-Nano formed a stable mono-dispersion (110.50±0.71nm) with >90% encapsulation of DIM-D that was supported by HPLC, DSC, FT-IR and AFM phase imaging. The blank formulation was non-toxic to human embryonic kidney cells. UltraFLEX-Nano was vastly deformable and highly permeable across the stratum corneum; there was significant (p<0.01) skin deposition of DIM-D for UltraFLEX-Nano that was superior to PEG solution (13.83-fold). DIM-D-UltraFLEX-Nano pretreatment delayed the onset of UVB-induced tumorigenesis (2 weeks) and reduced (p<0.05) the number of tumors observed in SKH-1 mice (3.33-fold), which was comparable to pretreatment with sunscreen (SPF30). Also, DIM-D-UltraFLEX-Nano caused decrease (p<0.05) in UV-induced DNA damage (8-hydroxydeoxyguanosine), skin inflammation (PCNA), epidermal hyperplasia (c-myc, CyclinD1), immunosuppression (IL10), cell survival (AKT), metastasis (Vimentin, MMP-9, TIMP1) but increase in apoptosis (p53 and p21). CONCLUSION: UltraFLEX-Nano was efficient in enhancing the topical delivery of DIM-D. DIM-D-UltraFLEX-Nano was efficacious in delaying skin tumor incidence and multiplicity in SKH mice comparable to sunscreen (SPF30).


Assuntos
Antioxidantes/farmacologia , Portadores de Fármacos/síntese química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Indóis/farmacologia , Nanopartículas/química , Neoplasias Cutâneas/prevenção & controle , 1,2-Dipalmitoilfosfatidilcolina/análogos & derivados , 1,2-Dipalmitoilfosfatidilcolina/química , 8-Hidroxi-2'-Desoxiguanosina , Animais , Antioxidantes/química , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/patologia , Carcinogênese/efeitos da radiação , Quimioprevenção/métodos , Ciclina D1/genética , Ciclina D1/metabolismo , Dano ao DNA , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Portadores de Fármacos/farmacologia , Composição de Medicamentos , Ácidos Graxos Monoinsaturados/química , Feminino , Células HEK293 , Humanos , Indóis/química , Interleucina-10/genética , Interleucina-10/metabolismo , Camundongos , Permeabilidade , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Compostos de Amônio Quaternário/química , Pele , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta
20.
J Pharm Sci ; 104(12): 4417-4426, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26372815

RESUMO

Very low oral bioavailability due to extensive pre-systemic metabolism and P-gp efflux has constrained the oral metronomic chemotherapy of docetaxel (DTX). There is tremendous need of compounds facilitating oral delivery of DTX. The research was aimed to investigate the effect of piperlongumine (PPL) on human liver microsomal metabolism, Caco-2 permeability, and cytotoxicity of DTX in triple-negative breast cancer cell lines. Reduction in testosterone and DTX metabolism (twofold increase in half-life) by PPL was comparable to the standard CYP3A4 inhibitor, cyclosporine A. P-gp efflux ratio of DTX across caco-2 monolayer was reduced from 2.37 to 1.52 on co-incubation with PPL. The IC50 value of DTX was reduced three to five times and combination index values in all the cell lines were below 0.6. PPL at non-cytotoxic concentration showed significant enhancement of the antimigration effect of DTX. Expression of tumor markers such as survivin, bcl2, C-myc, and cyclin D1 were downregulated to a great extent with enhanced p53 expression when treated with combination instead of individual drug. Co-treatment with PPL led to 1.68-fold enhancement in DTX bioavailability in SD rats. PPL could be a potential candidate in overcoming the obstacles associated with oral DTX delivery with synergistic anticancer activity.


Assuntos
Antineoplásicos/farmacologia , Dioxolanos/farmacologia , Taxoides/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Administração Oral , Animais , Disponibilidade Biológica , Células CACO-2 , Linhagem Celular Tumoral , Ciclosporina/farmacologia , Inibidores do Citocromo P-450 CYP3A/farmacologia , Docetaxel , Sinergismo Farmacológico , Feminino , Meia-Vida , Humanos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Ratos , Ratos Sprague-Dawley , Neoplasias de Mama Triplo Negativas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...