Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Small ; : e2307462, 2024 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-38342698

RESUMEN

The response to treatment is substantially varied between individual patients with ovarian cancer. However, chemotherapy treatment plans rarely pay sufficient attention to the mentioned factors. Instead, standardized treatment protocols are usually employed for most ovarian cancer patients. Variations in an individual's sensitivity to drugs significantly limit the effectiveness of treatment in some patients and lead to severe toxicities in others. In the present investigation, a nanotechnology-based approach for personalized treatment of ovarian carcinoma (the most lethal type of gynecological cancer) constructed on the individual genetic profile of the patient's tumor is developed and validated. The expression of predefined genes and proteins is analyzed for each patient sample. Finally, a mixture of the complex nanocarrier-based targeted delivery system containing drug(s)/siRNA(s)/targeted peptide is selected from the pre-synthesized bank and tested in vivo on murine cancer model using cancer cells isolated from tumors of each patient. Based on the results of the present study, an innovative approach and protocol for personalized treatment of ovarian cancer are suggested and evaluated. The results of the present study clearly show the advantages and perspectives of the proposed individual treatment approach.

2.
Mol Cancer Ther ; 21(9): 1381-1392, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-35732569

RESUMEN

Only a small percentage (<1%) of patients with late-stage lung squamous cell carcinoma (LUSC) are eligible for targeted therapy. Because PI3K/AKT/mTOR signaling, particularly Phosphatidylinositol 3-kinase CA (PIK3CA), is dysregulated in two-thirds of LUSC, and DNA damage response pathways are enriched in LUSC, we tested whether CC-115, a dual mTORC1/2 and DNA-PK inhibitor, sensitizes LUSC to chemotherapy. We demonstrate that CC-115 synergizes with carboplatin in six of 14 NSCLC cell lines, primarily PIK3CA-mutant LUSC. Synergy was more common in cell lines that had decreased basal levels of activated AKT and DNA-PK, evidenced by reduced P-S473-AKT, P-Th308-AKT, and P-S2056-DNA-PKcs. CC-115 sensitized LUSC to carboplatin by inhibiting chemotherapy-induced AKT activation and maintaining apoptosis, particularly in PIK3CA-mutant cells lacking wild-type (WT) TP53. In addition, pathway analysis revealed that enrichments in the IFNα and IFNγ pathways were significantly associated with synergy. In multiple LUSC patient-derived xenograft and cell line tumor models, CC-115 plus platinum-based doublet chemotherapy significantly inhibited tumor growth and increased overall survival as compared with either treatment alone at clinically relevant dosing schedules. IHC and immunoblot analysis of CC-115-treated tumors demonstrated decreased P-Th308-AKT, P-S473-AKT, P-S235/236-S6, and P-S2056-DNA-PKcs, showing direct pharmacodynamic evidence of inhibited PI3K/AKT/mTOR signaling cascades. Because PI3K pathway and DNA-PK inhibitors have shown toxicity in clinical trials, we assessed toxicity by examining weight and numerous organs in PRKDC-WT mice, which demonstrated that the combination treatment does not exacerbate the clinically accepted side effects of standard-of-care chemotherapy. This preclinical study provides strong support for the further investigation of CC-115 plus chemotherapy in LUSC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Animales , Carboplatino/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/genética , ADN/uso terapéutico , Humanos , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazinas , Serina-Treonina Quinasas TOR/metabolismo , Triazoles
3.
Theranostics ; 9(26): 8362-8376, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31754402

RESUMEN

Non-Small Cell Lung Carcinoma (NSCLC), is the most common type of lung cancer (more than 80% of all cases). Small molecule Tyrosine Kinase (TK) Inhibitors acting on the Epidermal Growth Factor Receptors (EGFRs) are standard therapies for patients with NSCLC harboring EGFR-TK inhibitor-sensitizing mutations. However, fewer than 10 % of patients with NSCLC benefit from this therapy. Moreover, even the latest generation of EGFR inhibitors can cause severe systemic toxicities and are ineffective in preventing non-canonical EGFR signaling. In order to minimize and even overcome these limitations, we are proposing a novel multi-tier biotechnology treatment approach that includes: (1) suppression of all four types of EGFR-TKs by a pool of small interfering RNAs (siRNAs); (2) induction of cell death by an anticancer drug, (3) enhancing the efficiency of the treatment by the local inhalation delivery of therapeutic agents directly to the lungs (passive targeting), (4) active receptor-mediated targeting of the therapy specifically to cancer cells that in turn should minimize adverse side effects of treatment and (5) increasing the stability, solubility, and cellular penetration of siRNA and drug by using tumor targeted Nanostructured Lipid Carriers (NLC). Methods: NLCs targeted to NSCLC cells by a synthetic Luteinizing Hormone-Releasing Hormone (LHRH) decapeptide was used for the simultaneous delivery of paclitaxel (TAX) and a pool of siRNAs targeted to the four major forms of EGFR-TKs. LHRH-NLC-siRNAs-TAX nanoparticles were synthesized, characterized and tested in vitro using human lung cancer cells with different sensitivities to gefitinib (inhibitor of EGFR) and in vivo on an orthotopic NSCLC mouse model. Results: Proposed nanoparticle-based complex containing an anticancer drug, inhibitors of different types of EGFR-TKs and peptide targeted to the tumor-specific receptors (LHRH-NLC-siRNAs-TAX) demonstrated a favorable organ distribution and superior anticancer effect when compared with treatment by a single drug, inhibitor of one EGFR-TK and non-targeted therapy. Conclusions: The use of a multifunctional NLC-based delivery system substantially enhanced the efficiency of therapy for NSCLC and possibly will limit adverse side effects of the treatments. The results obtained have the potential to significantly impact the field of drug delivery and to improve the efficiency of therapy of lung and other types of cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/terapia , Nanotecnología/métodos , Células A549 , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Gefitinib/uso terapéutico , Terapia Genética/métodos , Humanos , Ratones
4.
J Control Release ; 303: 109-116, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-30981814

RESUMEN

Chemotherapy-induced peripheral neuropathy (CIPN) is a major adverse effect of paclitaxel. Several liposome-based products have been approved and demonstrated superior efficacy and safety profiles for other drugs. The first objective of this work was to evaluate the effect of liposome formulation of paclitaxel (L-PTX) on neurotoxicity in-vitro and in-vivo in comparison to the standard Taxol® formulation. The second aim was to investigate the effect of formulation on paclitaxel biodistribution following intravenous administration in an animal model. Free paclitaxel was toxic to cell of neuronal origin (IC50 = 18.4 µg/mL) at a lower concentration than to lung cancer cells (IC50 = 59.1 µg/mL), and L-PTX demonstrated a comparable toxicity in both cell lines (IC50 = 31.8 and 33.7 µg/mL). Administration of L-PTX at 2 mg/kg per dose for a total of 4 doses on day 0, 2, 4, and 6 to rats did not result in increased sensitivity in response to mechanical or thermal stimulation of hind paws, in comparison to Taxol® administration at the same dose level that resulted in neuropathy. Paclitaxel biodisposition was evaluated for two formulations in plasma, liver, lung, brain, spinal cord, skin and muscle of rats after single intravenous dose at 6 mg/kg. The exposure to paclitaxel in brain, spinal cord, muscle, and skin was lower in the L-PTX group compared to Taxol® group. PEGylated liposomes containing paclitaxel were successfully developed and demonstrated reduced neurotoxicity in-vitro in neuronal cells and prevented development of peripheral neuropathy in-vivo. This proof of concept study showed that formulation in nanoparticles is a promising approach for reducing (or preventing) neurotoxicity caused by cancer drugs.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Nanopartículas/administración & dosificación , Paclitaxel/administración & dosificación , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacocinética , Encéfalo/metabolismo , Línea Celular Tumoral , Composición de Medicamentos , Humanos , Liposomas , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Músculos/metabolismo , Nanopartículas/química , Paclitaxel/química , Paclitaxel/farmacocinética , Enfermedades del Sistema Nervioso Periférico/prevención & control , Ratas Sprague-Dawley , Piel/metabolismo , Médula Espinal/metabolismo , Distribución Tisular
5.
Drug Deliv Transl Res ; 8(5): 1483-1507, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29978332

RESUMEN

The major current conventional types of metastatic breast cancer (MBC) treatments include surgery, radiation, hormonal therapy, chemotherapy, or immunotherapy. Introducing biological drugs, targeted treatment and gene therapy can potentially reduce the mortality and improve the quality of life in patients with MBC. However, combination of several types of treatment is usually recommended. Triple negative breast cancer (TNBC) accounts for 10-20% of all cases of breast carcinoma and is characterized by the low expression of progesterone receptor (PR), estrogen receptor (ER), and human epidermal growth factor receptor 2 (HER2). Consequently, convenient treatments used for MBC that target these receptors are not effective for TNBC which therefore requires special treatment approaches. This review discusses the occurrence of MBC, the prognosis and predictive biomarkers of MBC, and focuses on the novel advanced tactics for treatment of MBC and TNBC. Nanotechnology-based combinatorial approach for the suppression of EGFR by siRNA and gifitinib is described.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Terapia Combinada/métodos , Neoplasias de la Mama Triple Negativas/terapia , Animales , Ensayos Clínicos como Asunto , Receptores ErbB/antagonistas & inhibidores , Femenino , Gefitinib/uso terapéutico , Terapia Genética , Humanos , Metástasis de la Neoplasia , Pronóstico , ARN Interferente Pequeño/administración & dosificación , Neoplasias de la Mama Triple Negativas/metabolismo
6.
Nanomedicine ; 13(6): 1983-1992, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28434932

RESUMEN

Inhalation delivery of prostaglandin E (PGE2) in combination with selected siRNA(s) was proposed for the efficient treatment of idiopathic pulmonary fibrosis (IPF). Nanostructured lipid carriers (NLC) were used as a delivery system for PGE2 with and without siRNAs targeted to MMP3, CCL12, and HIF1Alpha mRNAs. The model of IPF was developed in SKH1 mice by intratracheal administration of bleomycin at a dose of 1.5U/kg. Results showed that NLC-PGE2 in combination with three siRNAs delivered locally to the lungs by inhalation markedly reduced mouse body mass, substantially limited hydroxyproline content in the lungs and disturbances of the mRNAs and protein expression, restricted lung tissue damage and prevented animal mortality. Our data provide evidence that IPF can be effectively treated by inhalation of the NLC-PGE2 in combination with siRNAs delivered locally into the lungs. This effect could not be achieved by using NLC containing just PGE2 or siRNA(s) alone.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Fibrosis Pulmonar Idiopática/terapia , Nanopartículas/administración & dosificación , Prostaglandinas/uso terapéutico , ARN Interferente Pequeño/administración & dosificación , Administración por Inhalación , Animales , Antibióticos Antineoplásicos/farmacología , Bleomicina/farmacología , Terapia Combinada , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Lípidos/química , Ratones , Ratones Pelados , ARN Interferente Pequeño/genética
7.
J Control Release ; 228: 150-158, 2016 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-26965957

RESUMEN

Delivery of macromolecules such as siRNA into cells that reside in the basal epidermis of the skin is a major challenge due to the transport barriers that need to be overcome. siRNAs have potential therapeutic applications in various dermatological diseases such as psoriasis, atopic dermatitis, and cancer. Unfortunately, a low permeability of siRNA through the stratum corneum and epidermis has significantly limited its use for topical application. The objective of this study was to develop a topical siRNA delivery system that can permeate through the stratum corneum and viable epidermis and efficiently deposit therapeutic levels of siRNA to the basal epidermis/upper dermis where melanoma cells reside. To achieve this objective, a series of liposome compositions that contained various concentrations of edge activator in their structures were prepared and then complexed with siRNA at different ratios to generate a small library of liposome-siRNA complexes (lipoplexes) with different physicochemical properties. In this study we used melanoma as a disease model. Through use of quantitative imaging analysis, we identified the necessary design parameters for effective permeation of lipoplexes through the skin layers and deposition at the upper dermis. The ability of the formulated lipoplexes to internalize into melanoma cells, knockdown the expression of the BRAF protein and induce cell death in melanoma cells was studied by fluorescent microscopy, in-cell immunofluorescence assay and WST-1 cell proliferation assay. By providing direct quantitative and qualitative microscopy evidence, the results of this study demonstrate for the first time that the passive delivery of an edge-activated liposomal formulation can effectively carry siRNA through the stratum corneum and deposit it at the lower epidermis/upper dermis.


Asunto(s)
Epidermis/metabolismo , Técnicas de Transferencia de Gen , Melanoma/terapia , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/uso terapéutico , Tratamiento con ARN de Interferencia , Línea Celular Tumoral , Epidermis/patología , Humanos , Lípidos/química , Liposomas/química , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Proteínas Proto-Oncogénicas B-raf/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacocinética , Absorción Cutánea
8.
Biomaterials ; 81: 46-57, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26720500

RESUMEN

Stem cell-based gene therapies, wherein stem cells are genetically engineered to express therapeutic molecules, have shown tremendous potential for cancer applications owing to their innate ability to home to tumors. However, traditional stem cell-based gene therapies are hampered by our current inability to control when the therapeutic genes are actually turned on, thereby resulting in detrimental side effects. Here, we report the novel application of magnetic core-shell nanoparticles for the dual purpose of delivering and activating a heat-inducible gene vector that encodes TNF-related apoptosis-inducing ligand (TRAIL) in adipose-derived mesenchymal stem cells (AD-MSCs). By combining the tumor tropism of the AD-MSCs with the spatiotemporal MCNP-based delivery and activation of TRAIL expression, this platform provides an attractive means with which to enhance our control over the activation of stem cell-based gene therapies. In particular, we found that these engineered AD-MSCs retained their innate ability to proliferate, differentiate, and, most importantly, home to tumors, making them ideal cellular carriers. Moreover, exposure of the engineered AD-MSCS to mild magnetic hyperthermia resulted in the selective expression of TRAIL from the engineered AD-MSCs and, as a result, induced significant ovarian cancer cell death in vitro and in vivo.


Asunto(s)
Terapia Genética , Hipertermia Inducida , Magnetismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Neoplasias Ováricas/terapia , Animales , Apoptosis , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Calor , Humanos , Ratones Desnudos , Nanopartículas/química , Nanopartículas/ultraestructura , Neoplasias Ováricas/patología , Plásmidos/metabolismo , Polietileneimina/química , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
9.
Langmuir ; 30(43): 12941-9, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25300552

RESUMEN

The aim of the present work is to synthesize, characterize, and test self-assembled anisotropic or Janus particles designed to load anticancer drugs for lung cancer treatment by inhalation. The particles were synthesized using binary mixtures of biodegradable and biocompatible materials. The particles did not demonstrate cyto- and genotoxic effects. Janus particles were internalized by cancer cells and accumulated both in the cytoplasm and nuclei. After inhalation delivery, nanoparticles accumulated preferentially in the lungs of mice and retained there for at least 24 h. Two drugs or other biologically active components with substantially different aqueous solubility can be simultaneously loaded in two-phases (polymer-lipid) of these nanoparticles. In the present proof-of-concept investigation, the particles were loaded with two anticancer drugs: doxorubicin and curcumin as model anticancer drugs with relatively high and low aqueous solubility, respectively. However, there are no obstacles for loading any hydrophobic or hydrophilic chemical agents. Nanoparticles with dual load were used for their local inhalation delivery directly to the lungs of mice with orthotopic model of human lung cancer. In vivo experiments showed that the selected nanoparticles with two anticancer drugs with different mechanisms of action prevented progression of lung tumors. It should be stressed that anticancer effects of the combined treatment with two anticancer drugs loaded in the same nanoparticle significantly exceeded the effect of either drug loaded in similar nanoparticles alone.


Asunto(s)
Antineoplásicos/química , Curcumina/química , Portadores de Fármacos/química , Interacciones Hidrofóbicas e Hidrofílicas , Pulmón/metabolismo , Nanopartículas/química , Administración por Inhalación , Animales , Anisotropía , Antineoplásicos/administración & dosificación , Transporte Biológico , Línea Celular Tumoral , Curcumina/administración & dosificación , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/toxicidad , Humanos , Ratones , Pruebas de Mutagenicidad , Tamaño de la Partícula
10.
J Control Release ; 194: 103-12, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25192941

RESUMEN

The clinical application of gene silencing is hindered by poor stability and low delivery efficiency of naked oligonucleotides. Here, we present the in vitro and in vivo behaviors of a rationally designed, ternary, self-assembled nanoparticle complex, consisting of an anionic copolymer, cationic DOTAP liposome, and antisense oligonucleotide (AON). The multifunctional copolymers are based on backbone poly(propylacrylic acid) (PPAA), a pH-sensitive hydrophobic polymer, with grafted poly(alkylene oxides) (PAOs) varying in extent of grafting and PAO chemistry. The nanoparticle complexes with PPAA-g-PAO copolymers enhance antisense gene silencing effects in A2780 human ovarian cancer cells. A greater amount of AON is delivered to ovarian tumor xenografts using the ternary copolymer-stabilized delivery system, compared to a binary DOTAP/AON complex, following intraperitoneal injection in mice. Further, intratumoral injection of the nanoparticle complexes containing 1 mol% grafted PAO reduced tumoral bcl-2 expression by up to 60%. The data for complexes across the set of PAO polymers support a strong role for the hydrophilic-lipophilic balance of the graft copolymer in achieving serum stability and cellular uptake. Based upon these results, we anticipate that this novel nanoparticle delivery system can be extended to the delivery of plasmid DNA, siRNA, or aptamers for preclinical and clinical development.


Asunto(s)
Resinas Acrílicas/química , Liposomas/química , Oligonucleótidos Antisentido/administración & dosificación , Polietilenglicoles/química , 1,2-Dipalmitoilfosfatidilcolina/química , Acrilamidas , Animales , Cationes/química , Línea Celular Tumoral , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Femenino , Silenciador del Gen/efectos de los fármacos , Genes bcl-2/genética , Terapia Genética/métodos , Hemólisis/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Nanopartículas , Neoplasias/terapia , Oligonucleótidos Antisentido/farmacocinética , Oligonucleótidos Antisentido/farmacología , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Pharm Res ; 31(12): 3487-502, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24919932

RESUMEN

PURPOSE: Design and synthesis of a tumor responsive nanoparticle-based system for imaging and treatment of various cancers. METHODS: Manganese oxide nanoparticles (Mn3O4 NPs) were synthesized and modified with LHRH targeting peptide or anti-melanoma antibodies (cancer targeting moieties) and a MMP2 cleavable peptide (a possible chemotactic factor). Nanostructured lipid carriers (NLCs) were used to entrap the BRAF inhibitor, vemurafenib, and enhance cytotoxicity of the drug. Size distribution, stability, drug entrapment, cytotoxicity and genotoxicity of synthesized nanoparticles were studied in vitro. Enhancement of MRI signal by nanoparticles and their body distribution were examined in vivo on mouse models of melanoma, ovarian and lung cancers. RESULTS: Uniform, stable cancer-targeted nanoparticles (PEGylated water-soluble Mn3O4 NPs and NLCs) were synthesized. No signs of cyto-,genotoxicity and DNA damage were detected for nanoparticles that do not contain an anticancer drug. Entrapment of vemurafenib into nanoparticles significantly enhanced drug toxicity in cancer cells with targeted V600E mutation. The developed nanoparticles containing LHRH and MMP2 peptides showed preferential accumulation in primary and metastatic tumors increasing the MRI signal in mice with melanoma, lung and ovarian cancers. CONCLUSIONS: The proposed nanoparticle-based systems provide the foundation for building an integrated MRI diagnostic and therapeutic approach for various types of cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Medios de Contraste/administración & dosificación , Medios de Contraste/química , Imagen por Resonancia Magnética/métodos , Nanopartículas/química , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Materiales Biocompatibles , Supervivencia Celular/efectos de los fármacos , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Indoles/administración & dosificación , Indoles/uso terapéutico , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Compuestos de Manganeso/química , Melanoma/diagnóstico , Melanoma/tratamiento farmacológico , Ratones , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/tratamiento farmacológico , Óxidos/química , Sulfonamidas/administración & dosificación , Sulfonamidas/uso terapéutico , Distribución Tisular , Vemurafenib
12.
Cancer Biol Med ; 11(1): 44-55, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24738038

RESUMEN

OBJECTIVE: Various nanoparticles have been designed and tested in order to select optimal carriers for the inhalation delivery of anticancer drugs to the lungs. METHODS: THE FOLLOWING NANOCARRIERS WERE STUDIED: micelles, liposomes, mesoporous silica nanoparticles (MSNs), poly propyleneimine (PPI) dendrimer-siRNA complexes nanoparticles, quantum dots (QDs), and poly (ethylene glycol) polymers. All particles were characterized using the following methods: dynamic light scattering, zeta potential, atomic force microscopy, in vitro cyto- and genotoxicity. In vivo organ distribution of all nanoparticles, retention in the lungs, and anticancer effects of liposomes loaded with doxorubicin were examined in nude mice after the pulmonary or intravenous delivery. RESULTS: Significant differences in lung uptake were found after the inhalation delivery of lipid-based and non-lipid-based nanoparticles. The accumulation of liposomes and micelles in lungs remained relatively high even 24 h after inhalation when compared with MSNs, QDs, and PPI dendrimers. There were notable differences between nanoparticle accumulation in the lungs and other organs 1 and 3 h after inhalation or intravenous administrations, but 24 h after intravenous injection all nanoparticles were mainly accumulated in the liver, kidneys, and spleen. Inhalation delivery of doxorubicin by liposomes significantly enhanced its anticancer effect and prevented severe adverse side effects of the treatment in mice bearing the orthotopic model of lung cancer. CONCLUSION: The results of the study demonstrate that lipid-based nanocarriers had considerably higher accumulation and longer retention time in the lungs when compared with non-lipid-based carriers after the inhalation delivery. These particles are most suitable for effective inhalation treatment of lung cancer.

13.
Clin Cancer Res ; 19(22): 6193-204, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24036854

RESUMEN

PURPOSE: The proposed project is aimed at enhancing the efficiency of epithelial ovarian cancer treatment and reducing adverse side effects of chemotherapy using nanotechnology. Overexpression of the CD44 membrane receptor results in tumor initiation, growth, cancer stem cells' specific behavior, development of drug resistance, and metastases. We hypothesize that a developed cancer-targeted delivery system that combines CD44 siRNA with paclitaxel would successfully deliver its payload inside cancer cells, effectively induce cell death, and prevent metastases. EXPERIMENTAL DESIGN: We synthesized, characterized, and tested a nanoscale-based drug delivery system (DDS) containing a modified polypropylenimine (PPI) dendrimer as a carrier; anticancer drug paclitaxel as a cell death inducer; a synthetic analog of luteinizing hormone-releasing hormone (LHRH) peptide as a tumor-targeting moiety; and siRNA targeted to CD44 mRNA. The proposed DDS was tested in vitro and in vivo using metastatic ovarian cancer cells isolated from patients with malignant ascites. RESULTS: We found that in contrast with cells isolated from primary tumors, CD44 was highly overexpressed in metastatic cancer cells. Treatment with the proposed tumor-targeted nanoscale-based nucleic acid and DDS led to the suppression of CD44 mRNA and protein, efficient induction of cell death, effective tumor shrinkage, and prevention of adverse side effects on healthy organs. CONCLUSION: We show a high therapeutic potential for combinatorial treatment of ovarian carcinoma with a novel DDS that effectively transports siRNA targeting to CD44 mRNA simultaneously with cytotoxic agents. Clin Cancer Res; 19(22); 6193-204. ©2013 AACR.


Asunto(s)
Líquido Ascítico/citología , Dendrímeros/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/farmacología , ARN Interferente Pequeño/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Transporte Biológico , Femenino , Expresión Génica , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/biosíntesis , Humanos , Receptores de Hialuranos/biosíntesis , Receptores de Hialuranos/genética , Ratones , Ratones Desnudos , Nanomedicina , Nanopartículas/química , Invasividad Neoplásica , Trasplante de Neoplasias , Paclitaxel/administración & dosificación , Proyectos Piloto , Polipropilenos , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Receptores LHRH/biosíntesis , Trasplante Heterólogo , Células Tumorales Cultivadas
14.
J Control Release ; 171(3): 349-57, 2013 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-23648833

RESUMEN

We developed, synthesized, and tested a multifunctional nanostructured lipid nanocarrier-based system (NLCS) for efficient delivery of an anticancer drug and siRNA directly into the lungs by inhalation. The system contains: (1) nanostructured lipid carriers (NLC); (2) anticancer drug (doxorubicin or paclitaxel); (3) siRNA targeted to MRP1 mRNA as a suppressor of pump drug resistance; (4) siRNA targeted to BCL2 mRNA as a suppressor of nonpump cellular resistance and (5) a modified synthetic analog of luteinizing hormone-releasing hormone (LHRH) as a targeting moiety specific to the receptors that are overexpressed in the plasma membrane of lung cancer cells. The NLCS was tested in vitro using human lung cancer cells and in vivo utilizing mouse orthotopic model of human lung cancer. After inhalation, the proposed NLCS effectively delivered its payload into lung cancer cells leaving healthy lung tissues intact and also significantly decreasing the exposure of healthy organs when compared with intravenous injection. The NLCS showed enhanced antitumor activity when compared with intravenous treatment. The data obtained demonstrated high efficiency of proposed NLCS for tumor-targeted local delivery by inhalation of anticancer drugs and mixture of siRNAs specifically to lung cancer cells and, as a result, efficient suppression of tumor growth and prevention of adverse side effects on healthy organs.


Asunto(s)
Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Neoplasias Pulmonares/tratamiento farmacológico , Nanoestructuras/química , Paclitaxel/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Doxorrubicina/uso terapéutico , Hormona Liberadora de Gonadotropina/química , Humanos , Lípidos/química , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Paclitaxel/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico
15.
Curr Drug Discov Technol ; 10(1): 8-15, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22564170

RESUMEN

The prevention of cyto- and genotoxicity of nanocarriers is an important task in nanomedicine. In the present investigation, we, at the first time using similar experimental conditions, compared genotoxicity of nanocarriers with different composition, architecture, size, molecular weight and charge. Poly(ethylene glycol) polymers, neutral and cationic liposomes, micelles, poly(amindo amine) and poly(propyleneimine) dendrimers, quantum dots, mesoporous silica, and supermagnetic iron oxide (SPIO) nanoparticles were studied. All nanoparticles were used in non-cytotoxic concentrations. However, even in these concentrations, positively charged cationic liposomes, dendrimers, and SPIO nanoparticles induced genotoxicity leading to the significant formation of micronuclei in cells. Negatively charged and neutral nanocarriers were not genotoxic. A strong positive correlation was found between the number of formed micronuclei and the positive charge of nanocarriers. We proposed modifications of both types of dendrimers and SPIO nanoparticles that substantially decreased their genotoxicity and allowed for an efficient intracellular delivery of nucleic acids.


Asunto(s)
Portadores de Fármacos/toxicidad , Micronúcleos con Defecto Cromosómico/inducido químicamente , Nanopartículas/toxicidad , Animales , Células CHO , Supervivencia Celular/efectos de los fármacos , Cricetinae , Cricetulus , Dendrímeros/toxicidad , Portadores de Fármacos/química , Pruebas de Micronúcleos , Nanopartículas/química , Tamaño de la Partícula , Polietilenglicoles/toxicidad , Polipropilenos/toxicidad , ARN Interferente Pequeño/administración & dosificación , Dióxido de Silicio/toxicidad
16.
Eur J Pharm Biopharm ; 84(2): 335-44, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23228437

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and often fatal form of interstitial lung disease. We hypothesized that the local pulmonary delivery of prostaglandin E2 (PGE2) by liposomes can be used for the effective treatment of IPF. To test this hypothesis, we used a murine model of bleomycin-induced IPF to evaluate liposomal delivery of PGE2 topically to the lungs. Animal survival, body weight, hydroxyproline content in the lungs, lung histology, mRNA, and protein expression were studied. After inhalation delivery, liposomes accumulated predominately in the lungs. In contrast, intravenous administration led to the accumulation of liposomes mainly in kidney, liver, and spleen. Liposomal PGE2 prevented the disturbances in the expression of many genes associated with the development of IPF, substantially restricted inflammation and fibrotic injury in the lung tissues, prevented decrease in body weight, limited hydroxyproline accumulation in the lungs, and virtually eliminated mortality of animals after intratracheal instillation of bleomycin. In summary, our data provide evidence that pulmonary fibrosis can be effectively treated by the inhalation administration of liposomal form of PGE2 into the lungs. The results of the present investigations make the liposomal form of PGE2 an attractive drug for the effective inhalation treatment of idiopathic pulmonary fibrosis.


Asunto(s)
Dinoprostona/administración & dosificación , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Administración Intravenosa , Animales , Bleomicina/toxicidad , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Hidroxiprolina/metabolismo , Fibrosis Pulmonar Idiopática/inducido químicamente , Inmunohistoquímica , Riñón/efectos de los fármacos , Liposomas/administración & dosificación , Liposomas/metabolismo , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , ARN Mensajero/metabolismo , Bazo/efectos de los fármacos , Factores de Tiempo , Distribución Tisular
17.
Nanomedicine (Lond) ; 7(2): 185-97, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22339132

RESUMEN

AIMS: To develop an approach for the effective treatment of ovarian tumor, prevention of metastases and limitation of side effects. MATERIALS & METHODS: In order to combine chemotherapy with genotherapy, we constructed a nanoscale-based tumor-targeted system containing two anticancer drugs, two antisense oligonucleotides (suppressors of cellular resistance) and a ligand specific to receptors overexpressed in cancer cells. The system was tested in a mouse metastatic xenograft model using tumor isolates from patients with ovarian carcinoma. RESULTS: Treatment with the proposed nanosystem decreased primary tumor, prevented intraperitoneal metastases and limited adverse side effects. CONCLUSIONS: The advantages of the proposed targeted complex treatment of primary aggressive ovarian tumor and prevention of metastases make the proposed approach promising for clinical applications.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma/secundario , Carcinoma/terapia , Terapia Genética/métodos , Terapia Molecular Dirigida/métodos , Nanocápsulas/uso terapéutico , Neoplasias Ováricas/terapia , Carcinoma/patología , Terapia Combinada , Femenino , Humanos , Nanocápsulas/química , Neoplasias Ováricas/patología , Resultado del Tratamiento , Células Tumorales Cultivadas
18.
Eur J Pharm Sci ; 45(3): 320-9, 2012 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-22155544

RESUMEN

Paclitaxel (PTX) has gained widespread clinical use yet its administration is associated with significant toxicity. In the present study, the toxicity and anti-tumor efficacy of tyrosine-derived nanospheres (NSP) for the delivery of PTX was compared to a clinical formulation of PTX in PBS-diluted Cremophor® EL (PTX-CrEL-D). Maximum tolerated dose was determined using a concentration series of PTX in NSP and CrEL-D, with toxicity assessed by measuring changes in body weight. Healthy mice administered PTX-NSP continued to gain weight normally while treatment with PTX-CrEL-D resulted in significant weight loss that failed to recover following treatment. Even at the dose of 50mg/kg, PTX-NSP showed better tolerance than 25mg/kg of PTX-CrEL-D. Xenograft studies of breast cancer revealed that the anti-tumor efficacy of PTX-NSP was equal to that of PTX-CrEL-D in tumors originating from both MDA-MB-435 and ZR-75-1 cancer lines. Larger volume of distribution and longer half-life were measured for PTX-NSP administration compared to those reported in the literature for a CrEL formulation. This trend suggests the potential for improved therapeutic index of PTX when administered via NSP. The findings reported here confirm that the NSP formulation is an efficient method for PTX administration with significant increase in maximum tolerated dose, offering possible clinical implications in the treatment of breast tumors.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Portadores de Fármacos/química , Glicerol/análogos & derivados , Nanosferas/química , Neoplasias/tratamiento farmacológico , Paclitaxel/administración & dosificación , Vehículos Farmacéuticos/química , Animales , Antineoplásicos Fitogénicos/química , Línea Celular Tumoral , Femenino , Glicerol/química , Humanos , Dosis Máxima Tolerada , Ratones , Ratones Desnudos , Paclitaxel/química , Tirosina/química , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Drug Target ; 19(10): 900-14, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21981718

RESUMEN

A tumor targeted mesoporous silica nanoparticles (MSN)-based drug delivery system (DDS) was developed for inhalation treatment of lung cancer. The system was capable of effectively delivering inside cancer cells anticancer drugs (doxorubicin and cisplatin) combined with two types of siRNA targeted to MRP1 and BCL2 mRNA for suppression of pump and nonpump cellular resistance in non-small cell lung carcinoma, respectively. Targeting of MSN to cancer cells was achieved by the conjugation of LHRH peptide on the surface of MSN via poly(ethylene glycol) spacer. The delivered anticancer drugs and siRNA preserved their specific activity leading to the cell death induction and inhibition of targeted mRNA. Suppression of cellular resistance by siRNA effectively delivered inside cancer cells and substantially enhanced the cytotoxicity of anticancer drugs. Local delivery of MSN by inhalation led to the preferential accumulation of nanoparticles in the mouse lungs, prevented the escape of MSN into the systemic circulation, and limited their accumulation in other organs. The experimental data confirm that the developed DDS satisfies the major prerequisites for effective treatment of non-small cell lung carcinoma. Therefore, the proposed cancer-targeted MSN-based system for complex delivery of drugs and siRNA has high potential in the effective treatment of lung cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Pulmonares/terapia , Nanotecnología , ARN Interferente Pequeño/administración & dosificación , Adenocarcinoma/patología , Adenocarcinoma/terapia , Administración por Inhalación , Animales , Antineoplásicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Línea Celular Tumoral , Cisplatino/administración & dosificación , Cisplatino/farmacología , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos , Hormona Liberadora de Gonadotropina/química , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Nanopartículas , Porosidad , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/metabolismo , Dióxido de Silicio/química
20.
Proc Natl Acad Sci U S A ; 107(23): 10737-42, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20498076

RESUMEN

Development of cancer cell resistance, low accumulation of therapeutic drug in the lungs, and severe adverse treatment side effects represent main obstacles to efficient chemotherapy of lung cancer. To overcome these difficulties, we propose inhalation local delivery of anticancer drugs in combination with suppressors of pump and nonpump cellular resistance. To test this approach, nanoscale-based delivery systems containing doxorubicin as a cell death inducer, antisense oligonucleotides targeted to MRP1 mRNA as a suppressor of pump resistance and to BCL2 mRNA as a suppressor of nonpump resistance, were developed and examined on an orthotopic murine model of human lung carcinoma. The experimental results show high antitumor activity and low adverse side effects of proposed complex inhalatory treatment that cannot be achieved by individual components applied separately. The present work potentially contributes to the treatment of lung cancer by describing a unique combinatorial local inhalation delivery of drugs and suppressors of pump and nonpump cellular resistance.


Asunto(s)
Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Oligonucleótidos/farmacología , Administración por Inhalación , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Ratones , Ratones Desnudos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Oligonucleótidos/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...