Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Control Release ; 370: 421-437, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38701884

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease with a high mortality rate due to limited treatment options. Current therapies cannot effectively reverse the damage caused by IPF. Research suggests that promoting programmed cell death (apoptosis) in myofibroblasts, the key cells driving fibrosis, could be a promising strategy. However, inducing apoptosis in healthy cells like epithelial and endothelial cells can cause unwanted side effects. This project addresses this challenge by developing a targeted approach to induce apoptosis specifically in myofibroblasts. We designed liposomes (LPS) decorated with peptides that recognize VCAM-1, a protein highly expressed on myofibroblasts in fibrotic lungs. These VCAM1-targeted LPS encapsulate Venetoclax (VNT), a small molecule drug that inhibits BCL-2, an anti-apoptotic protein. By delivering VNT directly to myofibroblasts, we hypothesize that VCAM1-VNT-LPS can selectively induce apoptosis in these cells, leading to reduced fibrosis and improved lung function. We successfully characterized VCAM1-VNT-LPS for size, surface charge, and drug loading efficiency. Additionally, we evaluated their stability over three months at different temperatures. In vitro and in vivo studies using a bleomycin-induced mouse model of lung fibrosis demonstrated the therapeutic potential of VCAM1-VNT-LPS. These studies showed a reduction in fibrosis-associated proteins (collagen, α-SMA, VCAM1) and BCL-2, while simultaneously increasing apoptosis in myofibroblasts. These findings suggest that VCAM1-targeted delivery of BCL-2 inhibitors using liposomes presents a promising and potentially selective therapeutic approach for IPF.


Assuntos
Apoptose , Compostos Bicíclicos Heterocíclicos com Pontes , Lipossomos , Camundongos Endogâmicos C57BL , Nanopartículas , Proteínas Proto-Oncogênicas c-bcl-2 , Sulfonamidas , Molécula 1 de Adesão de Célula Vascular , Animais , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Nanopartículas/administração & dosagem , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Molécula 1 de Adesão de Célula Vascular/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Apoptose/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Humanos , Pulmão/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/patologia , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Masculino , Camundongos , Bleomicina/administração & dosagem
2.
J Control Release ; 370: 110-123, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38648957

RESUMO

Continuous and aberrant activation of myofibroblasts is the hallmark of pathological fibrosis (e.g., abnormal wound healing). The deposition of excessive extracellular matrix (ECM) components alters or increases the stiffness of tissue and primarily accounts for multiple organ dysfunctions. Among various proteins, Cadherin-11 (CDH11) has been reported to be overexpressed on myofibroblasts in fibrotic tissues. Anti-apoptotic proteins such as (B cell lymphoma-2) (BCL-2) are also upregulated on myofibroblasts. Therefore, we hypothesize that CDH11 could be a targeted domain for cell-specific drug delivery and targeted inhibition of BCL-2 to ameliorate the development of fibrosis in the skin. To prove our hypothesis, we have developed liposomes (LPS) conjugated with CDH11 neutralizing antibody (antiCDH11) to target cell surface CDH11 and loaded these LPS with a BCL-2 inhibitor, Navitoclax (NAVI), to induce apoptosis of CDH11 expressing fibroblasts. The developed LPS were evaluated for physicochemical characterization, stability, in vitro therapeutic efficacy using dermal fibroblasts, and in vivo therapeutic efficacy in bleomycin-induced skin fibrosis model in mice. The findings from in vitro and in vivo studies confirmed that selectivity of LPS was improved towards CDH11 expressing myofibroblasts, thereby improving therapeutic efficacy with no indication of adverse effects. Hence, this novel research work represents a versatile LPS strategy that exhibits promising potential for treating skin fibrosis.


Assuntos
Apoptose , Caderinas , Fibrose , Lipossomos , Pele , Animais , Apoptose/efeitos dos fármacos , Fibrose/tratamento farmacológico , Caderinas/metabolismo , Pele/patologia , Pele/efeitos dos fármacos , Pele/metabolismo , Humanos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Compostos de Anilina/administração & dosagem , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Compostos de Anilina/química , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Bleomicina/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Camundongos Endogâmicos C57BL , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/uso terapêutico , Camundongos , Masculino
3.
Langmuir ; 40(5): 2754-2763, 2024 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-38275136

RESUMO

Peptide amphiphiles (PAs) are known for their remarkable ability to undergo molecular self-assembly, a process that is highly responsive to the local microenvironment. Herein, we design a pyrene tethered peptide amphiphile Py-VFFAKK, 1 that exhibits pathway-driven self-assembly from metastable nanoparticles to kinetically controlled nanofibers and thermodynamically stable twisted bundles upon modulations in pH, temperature, and chemical cues. The presence of the pyrene moiety ensures donation of the electron to an electron acceptor, namely, 7,7,8,8-tetracyanoquinodimethane (TCNQ), to form a supramolecular charge transfer complex in aqueous solution that was studied in detail with microscopic and spectroscopic techniques. Excitation of the donor species in its excimer state facilitates electron donation to the acceptor moiety, paving away a long-lived charge-separated state that persists for over a nanosecond, as ascertained through transient absorption spectroscopy. Finally, the self-assembled charge transfer complex is explored toward antimicrobial properties with Escherichia coli while maintaining biocompatibility toward L929 mice fibroblast cells.


Assuntos
Sinais (Psicologia) , Nanofibras , Animais , Camundongos , Peptídeos/farmacologia , Peptídeos/química , Análise Espectral , Nanofibras/toxicidade , Nanofibras/química , Pirenos
4.
ACS Appl Mater Interfaces ; 16(1): 305-317, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38157479

RESUMO

Gastric cancer treatment is challenging due to the lack of early-stage diagnostic technology and targeted delivery systems. Currently, the available treatments for gastric cancer are surgery, chemotherapy, immunotherapy, and radiation. These strategies are either invasive or require systemic delivery, exerting toxicities within healthy tissues. By creation of a targeted delivery system to the stomach, gastric cancer can be treated in the early stages. Such an approach reduces the negative effects on the rest of the body by minimizing systemic absorbance and random localization. With this in mind, we developed a mucoadhesive vehicle composed of ß-Glucan And Docosahexaenoic Acid (GADA) for controlled drug/gene delivery. In the current study, we investigated the therapeutic effect of codelivery Bcl2 inhibitors navitoclax (NAVI) and siRNA (Bcl2) via oral using GADA. The therapeutic efficacy of the GADA-mediated oral NAVI/siRNA was investigated in a gastric cancer mouse model. Higher Bcl2 inhibition efficacy was observed in Western blotting and TUNEL assay in mice treated with GADA/NAVI/siRNA compared to free NAVI, siRNA, and NAVI/siRNA. Histology (H&E) and immunohistochemistry (Ki67, TUNEL, and BCl2) analyses confirmed a significant reduction of the tumor region. Interaction between GADA and mucus resulted in retention for over 6 h and thereby sustained local payload release. The developed oral carrier GADA is an emerging vehicle that has promising potential in oral delivery of both small and large molecules, and their mucoadhesive property results in improved therapeutic efficacy with minimal side effects compared to conventional treatment. This study opens a new window for the effective delivery of oral medicine for the treatment of gastric cancer and other gastrointestinal diseases.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias Gástricas , Camundongos , Animais , Neoplasias Gástricas/tratamento farmacológico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química , Sistemas de Liberação de Medicamentos/métodos , RNA Interferente Pequeno , Nanopartículas/química
5.
J Control Release ; 361: 314-333, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37562554

RESUMO

Solid tumors are abnormal mass of tissue, which affects the organs based on its malignancy and leads to the dysfunction of the affected organs. The major problem associated with treatment of solid tumors is delivering anticancer therapeutics to the deepest layers/core of the solid tumor. Deposition of excessive extracellular matrix (ECM) hinders the therapeutics to travel towards the core of the tumor. Therefore, conventional anticancer therapeutics can only reduce the tumor size and that also for a limited duration, and tumor recurrence occurs once the therapy is discontinued. Additionally, by the time the cancer is diagnosed, the cancer cells already started affecting the major organs of the body such as lung, liver, spleen, kidney, and brain, due to their ability to metastasize and lung is the primary site for them to be infiltrated. To facilitate the anticancer therapeutics to penetrate the deeper layers of tumor, and to provide concurrent treatment of both the solid tumor and metastasis, we have designed and developed a Bimodal Light Assisted Skin Tumor and Metastasis Treatment (BLAST), which is a combination of photothermal and chemotherapeutic moieties. The BLAST is composed of 2D boron nitride (BN) nanosheet with adsorbed molecules of BCL-2 inhibitor, Navitoclax (NAVI) on its surface, that can breakdown excessive ECM network and thereby facilitate dissociation of the solid tumor. The developed BLAST was evaluated for its ability to penetrate solid tumors using 3D spheroids for the uptake, cytotoxicity, growth inhibition, reactive oxygen species (ROS) detection, penetration, and downregulation of proteins upon laser irradiation. The in vivo therapeutic studies on a skin cancer mice model revealed that the BLAST with and without laser were able to penetrate the solid tumor, reduce tumor volume in mice, dissociate the protein network, and prevent lung metastasis as confirmed by immunohistochemistry and western blot analysis. Post analysis of serum and blood components revealed the safety and efficacy of BLAST in mice. Hence, the developed BLAST holds strong promise in solid tumor treatment and metastasis prevention simultaneously.


Assuntos
Antineoplásicos , Neoplasias Pulmonares , Melanoma , Animais , Camundongos , Fototerapia , Antineoplásicos/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Luz , Melanoma/tratamento farmacológico , Linhagem Celular Tumoral
6.
ACS Appl Bio Mater ; 6(2): 365-383, 2023 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-36753355

RESUMO

Over the last decades, photomedicine has made a significant impact and progress in treating superficial cancer. With tremendous efforts many of the technologies have entered clinical trials. Photothermal agents (PTAs) have been considered as emerging candidates for accelerating the outcome from photomedicine based cancer treatment. Besides various inorganic and organic candidates, 2D materials such as graphene, boron nitride, and molybdenum disulfide have shown significant potential for photothermal therapy (PTT). The properties such as high surface area to volume, biocompatibility, stability in physiological media, ease of synthesis and functionalization, and high photothermal conversion efficiency have made 2D nanomaterials wonderful candidates for PTT to treat cancer. The targeting or localized activation could be achieved when PTT is combined with chemotherapies, immunotherapies, or photodynamic therapy (PDT) to provide better outcomes with fewer side effects. Though significant development has been made in the field of phototherapeutic drugs, several challenges have restricted the use of PTT in clinical use and hence they have not yet been tested in large clinical trials. In this review, we attempted to discuss the progress, properties, applications, and challenges of 2D materials in the field of PTT and their application in photomedicine.


Assuntos
Grafite , Nanoestruturas , Neoplasias , Fotoquimioterapia , Humanos , Fototerapia , Nanoestruturas/uso terapêutico , Neoplasias/tratamento farmacológico , Grafite/uso terapêutico
7.
Pharmaceutics ; 13(5)2021 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-34067176

RESUMO

DNA vaccines still represent an emergent area of research, giving rise to continuous progress towards several biomedicine demands. The formulation of delivery systems to specifically target mannose receptors, which are overexpressed on antigen presenting cells (APCs), is considered a suitable strategy to improve the DNA vaccine immunogenicity. The present study developed binary and ternary carriers, based on polyethylenimine (PEI), octa-arginine peptide (R8), and mannose ligands, to specifically deliver a minicircle DNA (mcDNA) vaccine to APCs. Systems were prepared at various nitrogen to phosphate group (N/P) ratios and characterized in terms of their morphology, size, surface charge, and complexation capacity. In vitro studies were conducted to assess the biocompatibility, cell internalization ability, and gene expression of formulated carriers. The high charge density and condensing capacity of both PEI and R8 enhance the interaction with the mcDNA, leading to the formation of smaller particles. The addition of PEI polymer to the R8-mannose/mcDNA binary system reduces the size and increases the zeta potential and system stability. Confocal microscopy studies confirmed intracellular localization of targeting systems, resulting in sustained mcDNA uptake. Furthermore, the efficiency of in vitro transfection can be influenced by the presence of R8-mannose, with great implications for gene expression. R8-mannose/PEI/mcDNA ternary systems can be considered valuable tools to instigate further research, aiming for advances in the DNA vaccine field.

8.
Nanomedicine ; 31: 102320, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33075540

RESUMO

The current work aims to explore the biological characteristics of vincristine synergistic co-loading into pegylated liposomal doxorubicin in non-indicated modalities of non-small cell lung cancer (NSCLC) and triple negative breast cancer (TNBC). The combinatorial liposome prepared by active co-loading of the drugs against modified ammonium ion gradient exhibited 95% encapsulation of both drugs. The cellular uptake studies using confocal microscopy and flow cytometry showed significantly increased uptake of dual drug formulation as against liposomal doxorubicin. The co-loaded liposome formulation had significantly increased cell cycle arrest in G2/M phase with subsequent apoptosis and reduced cell viability in both tumor cell lines than doxorubicin liposome. This carrier exhibited similar acute toxicity, pharmacokinetic and tissue distribution profiles with significant increase in tumor regression as compared to liposomal doxorubicin. These results indicate that co-encapsulation of vincristine into clinically used pegylated liposomal doxorubicin significantly improved in-vitro and in-vivo therapeutic efficacy against NSCLC and TNBC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Doxorrubicina/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Vincristina/uso terapêutico , Doxorrubicina/uso terapêutico , Portadores de Fármacos/química , Sinergismo Farmacológico , Feminino , Humanos , Polietilenoglicóis/uso terapêutico
9.
Pharm Res ; 37(2): 18, 2020 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-31897768

RESUMO

PURPOSE: In this study, we developed a polymeric micellar system for glutathione-mediated intracellular delivery of a photosensitizer, chlorin e6 (Ce6) by synthesizing an amphiphilic polymer, methoxy-poly(ethylene glycol)-poly(D,L-lactide)-disulfide-Ce6 (mPEG-PLA-S-S-Ce6), which self-assembled in aqueous environment to form micelles. METHODS: The polymer-drug conjugate was characterized by NMR. The singlet oxygen (2O1) generation and in vitro release of Ce6 micelles were evaluated. Further, glutathione-mediated intracellular drug delivery was assessed in human alveolar adenocarcinoma cells (A549), mouse mammary carcinoma cells (4 T1) and 3D A549 spheroids. RESULTS: The micellar system protected Ce6 from aggregation leading to improved 2O1 generation compared to free Ce6. Due to the availability of glutathione, the disulfide bonds in the micelles were cleaved resulting in rapid release of Ce6 evident from the in vitro study. The Ce6 micelles displayed quicker drug release in presence of glutathione monoester (GSH-OEt) pre-treated A549 and 4 T1 cells compared to without pre-treated cells. In vitro phototoxicity of micelles displayed enhanced toxicity in 10 mM GSH-OEt pre-treated A549 and 4 T1 cells compared to untreated cells. As anticipated, Ce6 micelles showed lower drug release in presence of 0.1 mM of buthionine sulfoximine (BSO) pretreated A549 and 4 T1 cells exhibiting lower phototoxicity. Further, A549 3D spheroids treated with Ce6 micelles showed significant inhibition in growth, enhanced phototoxicity, and cellular apoptosis in comparison to free Ce6. CONCLUSION: The above results showed that the developed strategy could be effective in improving the PDT efficacy of Ce6, and the developed polymeric micellar system could be utilized as a PDT regimen for cancer.


Assuntos
Glutationa/química , Poliésteres/química , Polietilenoglicóis/química , Porfirinas/química , Células A549 , Animais , Apoptose/efeitos dos fármacos , Butionina Sulfoximina/química , Linhagem Celular Tumoral , Clorofilídeos , Portadores de Fármacos/química , Liberação Controlada de Fármacos/efeitos dos fármacos , Humanos , Camundongos , Micelas , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/química , Polímeros/química , Porfirinas/farmacologia
10.
Photodiagnosis Photodyn Ther ; 29: 101633, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31870896

RESUMO

BACKGROUND: To develop a photosensitizer, chlorin e6 (Ce6)-based amphiphilic polymer, DP-Ce6, where DOPE and PEG are conjugated to Ce6, which would self-assemble to form polymeric micelles (DP-Ce6-M) in aqueous environment. METHODS: DP-Ce6-M were characterized for particle size, zeta potential, and singlet oxygen (1O2) generation. Cellular internalization, phototoxicity were investigated against monolayer and 3D spheroids of human lung adenocarcinoma cells (A549). RESULTS AND CONCLUSIONS: DP-Ce6-M formed stable micelles with particles size of 58.2 ±â€¯1.6 nm. Solubility of Ce6 was improved. Photoactivity of DP-Ce6-M was sustained in regard to 1O2 generation compared to free Ce6. The DP-Ce6-M showed enhanced internalization and growth inhibition in monolayer and spheroidal cells. Overall, DP-Ce6-M demonstrated the potential for further exploration as PDT agent for cancer treatment.


Assuntos
Adenocarcinoma/tratamento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Porfirinas/farmacologia , Neoplasias Uterinas/tratamento farmacológico , Linhagem Celular Tumoral , Clorofilídeos , Feminino , Humanos , Lipídeos/química , Lipídeos/farmacologia , Micelas , Nanopartículas , Tamanho da Partícula , Fosfatidiletanolaminas/química , Fosfatidiletanolaminas/farmacologia , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/química , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Polímeros/química , Polímeros/farmacologia , Porfirinas/química
11.
ACS Appl Bio Mater ; 3(5): 3157-3169, 2020 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-35025359

RESUMO

Even though existing chemotherapeutic agents, including paclitaxel (PTX), possess superior anticancer activity, their application is limited because of extreme hydrophobicity. Here, a dendrimer-based nanoparticles system has been prepared for improved delivery of loaded paclitaxel to the tumor. The cell-penetrating peptides, octa-arginine (R) and vitamin-E succinate (VES), had been conjugated to the PEGylated generation 4 polyamidoamine dendrimer (D) to form RVES-PD. The synthesized polymers had been characterized by 1NMR, IR, and gel permeation chromatography. The PTX-loaded RVES-PD (RVES-PD-PTX) was physicochemically characterized for particle size, zeta potential analysis, drug loading, release, and encapsulation studies. Cellular uptake, cytotoxicity, and apoptosis determination studies were carried out in human lung cancer cells (A549) in monolayers and spheroids. In vivo antitumor efficacy of RVES-PD-PTX was determined using B16F10 tumor-bearing mice. The results indicated that the RVES-PD nanoparticles were taken up by cancer cells effectively and demonstrated improved cellular translocation compared to nontargeted VES-PD. The cytotoxicity study revealed that RVES-PD-PTX exhibited the highest cytotoxicity of PTX compared to nontargeted VES-PD-PTX and free PTX in the PTX concentration range of 0-50 µg/mL. RVES-PD-PTX-induced apoptosis as analyzed by the Annexin V assay and suppressed the growth of spheroids to the highest extent compared to VES-PD-PTX and free PTX. Furthermore, the biodistribution study indicated that the RVES-PD had higher accumulation in tumor compared to VES-PD. The in vivo experiment using B16F10 tumor-bearing mice demonstrated that the RVES-PD treatment resulted in the highest rate of tumor volume reduction and apoptosis primarily by upregulating caspase 3/7 to the highest extent compared to VES-PD and free PTX. In conclusion, the developed RVES-PD could load PTX and showed efficient cell penetration and improved PTX-mediated cytotoxicity in all tested in vitro and in vivo assay systems. The results strongly suggest further exploration of this developed PTX-nanoformulation in cancer treatment.

12.
Nanomedicine (Lond) ; 14(24): 3159-3176, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31855118

RESUMO

Aim: Transferrin anchored, poly(ethylene glycol) (PEG) and α-tocopheryl succinate (α-TOS) conjugated generation 4 dendrimer has been prepared in order to develop a tumor targeted delivery system of a hydrophobic chemotherapeutic agent, paclitaxel (PTX). Materials & methods: The dendrimers were characterized physicochemically for size, ζ and encapsulation ability. The cellular uptake, cytotoxicity potential and apoptosis of prepared nanoconstruct were evaluated in human cervical epithelial cells monolayer and 3D spheroids. Results & conclusion: G4-TOS-PEG-Tf demonstrated increased cellular uptake, cytotoxicity and apoptotic potential of PTX compared with free PTX and G4-TOS-PEG-PTX. G4-TOS-PEG-Tf-PTX inhibited growth of human cervical epithelial cells spheroids significantly. The newly developed dendrimers hold promise as an efficient delivery system for PTX or other hydrophobic chemotherapeutic agents for targeted delivery to tumors.


Assuntos
Dendrímeros/química , Paclitaxel/química , Poliaminas/química , Transferrina/química , alfa-Tocoferol/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Citometria de Fluxo , Células HeLa , Humanos , Microscopia Confocal , Nanopartículas/química , Polietilenoglicóis , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos
13.
Mol Pharm ; 16(4): 1541-1554, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30817166

RESUMO

This study involves development of a dendrimer-based nanoconstruct by conjugating α-tocopheryl succinate (α-TOS) and polyethylene glycol (PEG) on a poly(amidoamine) dendrimer (G4 PAMAM) to improve intracellular delivery of a poorly water-soluble chemotherapeutic drug, paclitaxel (PTX). The conjugates were characterized by NMR, and PTX-loaded nanocarriers (G4-TOS-PEG-PTX) were evaluated for hydrodynamic diameter, polydispersity index (PDI), zeta potential, percentage encapsulation efficiency (%EE), and percentage drug loading (%DL). A hemolysis study was performed, which indicated that the synthesized dendrimer conjugates were not toxic to red blood cells; hence, they were biocompatible. A cellular uptake study in (B16F10 and MDA MB231) monolayer cells and 3D spheroids showed that α-TOS conjugation improved the time dependent uptake of nanosized dendrimer conjugates. The cell viability assay revealed that G4-TOS-PEG-PTX enhanced the cytotoxicity of PTX as compared to free PTX and PTX-loaded G4-PEG (G4-PEG-PTX) at tested concentrations. Correspondingly, the α-TOS-anchored dendrimer induced more apoptosis as compared to free PTX and G4-PEG-PTX. Moreover, the fluorescently labeled G4-TOS-PEG penetrated deeper into MDA MB231 3D spheroids as visualized by confocal microscopy. G4-TOS-PEG-PTX showed significant growth inhibition in 3D spheroids as compared to free PTX and G4-PEG-PTX. Further, the in vivo efficacy study using B16F10 xenografted C57Bl6/J mice indicated that the G4-TOS-PEG localized in tumor sections. G4-TOS-PEG-PTX reduced the tumor growth significantly compared to free PTX and G4-PEG-PTX. G4-TOS-PEG-PTX had more apoptotic potential in tumor sections as analyzed by TUNEL assay. Hence, the newly developed dendrimer conjugate, G4-TOS-PEG, has the potential to target loaded drug to the tumor, and G4-TOS-PEG-PTX has the potential to be utilized successfully in cancer treatment.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Dendrímeros/química , Sistemas de Liberação de Medicamentos , Melanoma Experimental/tratamento farmacológico , Nylons/química , Paclitaxel/farmacologia , Poliaminas/química , alfa-Tocoferol/química , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Antioxidantes/química , Apoptose , Neoplasias da Mama/patologia , Proliferação de Células , Portadores de Fármacos/química , Feminino , Humanos , Técnicas In Vitro , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/química , Paclitaxel/química , Polietilenoglicóis/química , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Nanomedicine (Lond) ; 14(7): 819-834, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30874479

RESUMO

AIM: In this study, we developed a chlorin e6-conjugated methoxy-poly(ethylene glycol)-poly(d,l-lactide) (mPEG-PLA-Ce6) amphiphilic polymer, which self-assembled to form stable nanoparticles. MATERIALS & METHODS: The nanoparticles were characterized for particle size, ζ-potential and singlet oxygen (1O2) generation. Cellular internalization and phototoxicity were investigated against monolayer and 3D spheroids of human lung adenocarcinoma cells (A549). RESULTS & CONCLUSION: mPEG-PLA-Ce6 exhibited a size of 149.72 ± 3.51 nm and ζ-potential of -24.82 ± 2.94 mV. The 1O2 generation by mPEG-PLA-Ce6 in water was considerably higher than free chlorin e6. The nanoparticles showed enhanced cellular internalization and phototoxicity in monolayer and 3D spheroids. The developed mPEG-PLA-Ce6 has potential application as a nanocarrier of chlorin e6 for photodynamic therapy of solid tumors.


Assuntos
Antineoplásicos/química , Portadores de Fármacos/química , Nanopartículas/química , Poliésteres/química , Polietilenoglicóis/química , Porfirinas/química , Células A549 , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Transporte Biológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Clorofilídeos , Liberação Controlada de Fármacos , Humanos , Micelas , Tamanho da Partícula , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Porfirinas/farmacologia
15.
Int J Pharm ; 557: 329-341, 2019 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-30599231

RESUMO

In the current study, we employed poly(amidoamine) (PAMAM) dendrimers of generation 4 (G4) to deliver paclitaxel (PTX), a poorly soluble anti-cancer agent precisely to cancer cells via its conjugation on dendrimer surface. Further, G4 PAMAM has been PEGylated (PEG) and tagged with Biotin, an essential micronutrient for cellular functions, receptors of which are overexpressed in certain cancers. The synthesized multifunctional conjugates were characterized by 1H NMR and zeta potential analysis techniques. In addition, the conjugates were evaluated in vitro in cell monolayers and 3D spheroids of biotin receptor over-expressed A549 cell line (human non-small cell lung cancer). G4 PTX PEG-Biotin conjugate penetrated at significantly higher extent in monolayers as well as spheroids as studied by flow cytometry and confocal microscopy by visualizing the cells at varied depth. The G4 PTX PEG-Biotin conjugate demonstrated higher cytotoxicity compared to free PTX and G4 PTX PEG conjugate as assessed by MTT assay in monolayers and Presto Blue assay in detached spheroidal cells. G4 PTX PEG-Biotin demonstrated significant inhibition of growth of tumor spheroids. Therefore, the newly synthesized biotin anchored PTX-conjugated dendrimer system is promising and could be further explored for efficiently delivering PTX to biotin receptor overexpressed cancers.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Biotina/administração & dosagem , Dendrímeros/administração & dosagem , Portadores de Fármacos/administração & dosagem , Paclitaxel/administração & dosagem , Polietilenoglicóis/administração & dosagem , Células A549 , Antineoplásicos Fitogênicos/química , Biotina/química , Sobrevivência Celular/efeitos dos fármacos , Dendrímeros/química , Portadores de Fármacos/química , Humanos , Neoplasias/tratamento farmacológico , Paclitaxel/química , Polietilenoglicóis/química , Esferoides Celulares/efeitos dos fármacos , Células Tumorais Cultivadas
16.
Curr Drug Deliv ; 15(9): 1271-1283, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29732970

RESUMO

BACKGROUND: Solid lipid nanoparticles (SLNs) represent an affordable, easily scalable, stable and biocompatible drug delivery system with a high drug to lipid ratio which also improves solubility of poorly soluble drugs. OBJECTIVE: SLNs were developed by using glyceryl monostearate as the single lipid in the presence of surfactant Poloxamer 188 and evaluated the efficiency of SLNs to load the therapeutic cargo, curcumin (CUR). METHOD: The nano-formulation was optimized by Quality by Design approach to understand the effect of various process parameters on various quality attributes, including drug loadability, particle size and polydispersity. The nanoparticles were characterized using Differential scanning calorimetry (DSC), Fourier Transform Infra-red Spectroscopy (FT-IR) and X-Ray Diffraction (XRD) analysis. These novel SLNs were evaluated for in-vitro anticancer activity using breast adenocarcinoma cells (MDA-MB-231). RESULTS: The optimized formulation had a particle size of 226.802±3.92 nm with low polydispersity index of 0.244±0.018. The % encapsulation of CUR into SLNs was found to be 67.88±2.08 %. DSC, FT-IR and XRD confirmed that the CUR was encapsulated stably into the lipid matrix, thereby improving the solubility of the drug. CUR-SLN showed sustained drug release in comparison to the free CUR solution. CUR-SLNs exhibited higher cellular uptake in human breast adenocarcinoma cells compared to free CUR at both 1 and 4 h time points. CUR-SLNs demonstrated decreased cell viability (43.97±1.53%) compared to free CUR (59.33±0.95%) at a concentration of 50 µg/mL after 24 h treatment. Furthermore, the treatment of MDA-MB-231 cells with CUR-SLNs for 24 h induced significantly higher apoptosis (37.28±5.3%) in cells compared to the free CUR (21.06±0.97%). CONCLUSION: The results provide a strong rationale for further exploration of the newly developed CUR-SLN to be utilized as a potent chemotherapeutic agent in cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Curcumina/farmacologia , Desenho de Fármacos , Glicerídeos/química , Lipídeos/química , Nanopartículas/química , Antineoplásicos/síntese química , Antineoplásicos/química , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Curcumina/síntese química , Curcumina/química , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Tamanho da Partícula , Propriedades de Superfície , Células Tumorais Cultivadas
17.
Chem Phys Lipids ; 208: 10-18, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28842128

RESUMO

The aim of the present research was to develop a novel, biocompatible, amenable to industrial scale up and affordable solid lipid nanoparticles (SLN) preparation of curcumin and evaluate the therapeutic efficacy in vitro using cancer cells. We have incorporated cholesterol as the lipid to prepare SLN along with the Poloxamer-188 as stabilizer. High shear homogenization was used to prepare the SLN and formulation was optimized using Quality by Design The optimized Chol CUR SLN exhibited a narrow size distribution with a particle size of 166.4±3.5nm. Percentage encapsulation (%EE) was found to be 76.9±1.9%. The SLN were further characterized by DSC, FTIR, XRD and drug release. In vitro cell studies in MDA-MB-231 (Human Breast cancer) cell line revealed that the Chol CUR SLN showed superior cytotoxicity and uptake in comparison to the free curcumin. Furthermore, Chol CUR SLN induced a significantly higher apoptosis compared to free CUR treatment. These results indicated that the curcumin encapsulated in Chol SLN was able to significantly improve the cytotoxic potential and induction of apoptosis in MDA-MB-231 cells. The promising result from our study could lead a further exploration of this nanoparticle formulation to be utilized clinically for cancer treatment.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Curcumina/química , Curcumina/farmacologia , Portadores de Fármacos/química , Lipídeos/química , Nanopartículas , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Transporte Biológico , Linhagem Celular Tumoral , Curcumina/metabolismo , Liberação Controlada de Fármacos , Humanos , Tamanho da Partícula
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA