Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Intervalo de año de publicación
1.
Nanotechnology ; 33(20)2022 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-35100566

RESUMEN

Short time treatment with reduced dosages of selol-loaded PLGA nanocapsules (NcSel) combined with magnetic hyperthermia (MHT) is evaluated in aged Erhlich tumor-bearing mice. Clinical, hematological, biochemical, genotoxic and histopathological parameters are assessed during 7 d treatment with NcSel and MHT, separately or combined. The time evolution of the tumor volume is successfully modeled using the logistic mathematical model. The combined therapy comprising NcSel and MHT is able to hinder primary tumor growth and a case of complete tumor remission is recorded. Moreover, no metastasis was diagnosed and the adverse effects are negligible. NcSel plus MHT may represent an effective and safe alternative to cancer control in aged patients. Future clinical trials are encouraged.


Asunto(s)
Neoplasias de la Mama/terapia , Hipertermia Inducida , Nanopartículas de Magnetita/uso terapéutico , Nanocápsulas/uso terapéutico , Compuestos de Selenio/uso terapéutico , Animales , Neoplasias de la Mama/patología , Carcinoma de Ehrlich/patología , Carcinoma de Ehrlich/terapia , Ciclo Celular/efectos de los fármacos , Terapia Combinada , Fragmentación del ADN/efectos de los fármacos , Femenino , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/ultraestructura , Ratones , Nanocápsulas/química , Nanocápsulas/ultraestructura , Compuestos de Selenio/química , Factores de Tiempo , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos
2.
Biomed Mater ; 16(3)2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33540396

RESUMEN

Systemic therapy is generally required for breast cancer. However, treatment toxicity and side effects are a concern, especially for triple-negative breast cancer (TNBC), a subtype that usually develops resistance to chemotherapy. To overcome this issue, new nanoformulations capable of targeting cancer cells have been developed and alternative biomarkers have been explored as target molecules for TNBC management. In this study, we performed anin vivoassay in a murine orthotopic TNBC model to evaluate the targeting ability of anti-carcinoembryonic antigen (anti-CEA) loaded nanoparticles (labelled MFCEA), which had been previously synthetized by our research group. 4T1 cells were injected in the mammary gland of balb-c mice, and tumors were evaluated for CEA expression by immunohistochemistry. Tumor-bearing mice received targeted (MFCEA) and non-targeted (MF) nanoparticles intraperitoneally. Tumors were removed 1, 4, 15 and 24 h after treatment, and Prussian blue iron staining was performed. Our results showed, as far as we know for the first time, that 4T1 induced tumors are CEA positive, and this opens up new prospects for treating TNBC. Furthermore, MFCEA nanoparticles were able to target malignant tissue and were retained in the tumor for longer than MF nanoparticles. The retention property of MFCEA, together with the absence of toxicity observed in the MTT assay, make these nanoparticles a promising device for management of CEA positive tumors and perhaps for TNBC. Nevertheless, further studies must be carried out to improve their performance and ensure safety for clinical studies.


Asunto(s)
Nanopartículas , Neoplasias de la Mama Triple Negativas , Animales , Antígeno Carcinoembrionario/uso terapéutico , Línea Celular Tumoral , Humanos , Hierro , Ratones , Nanopartículas/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
3.
Nanomedicine (Lond) ; 15(10): 1019-1036, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32264766

RESUMEN

Aim: Nano-5-aminolevulic acid (NanoALA)-mediated photodynamic therapy (PDT), an oil-in-water polymeric nanoemulsion of ALA, was evaluated in a murine model of breast cancer. Materials & methods: Analysis of ALA-derived protoporphyrin IX production and acute toxicity test, biocompatibility and treatment efficacy, and long-term effect of NanoALA-PDT on tumor progression were performed. Results: The nanoformulation favored the prodrug uptake by tumor cells in a shorter time (1.5 h). As a result, the adverse effects were negligible and the response rates for primary mammary tumor control were significantly improved. Tumor progression was slower after NanoALA-PDT treatment, providing longer survival. Conclusion: NanoALA is a good proactive drug candidate for PDT against cancer potentially applied as adjuvant/neoadjuvant intervention strategy for breast cancer.


Asunto(s)
Ácido Aminolevulínico/uso terapéutico , Neoplasias de la Mama , Fotoquimioterapia , Animales , Neoplasias de la Mama/tratamiento farmacológico , Muerte Celular , Línea Celular Tumoral , Portadores de Fármacos , Humanos , Ratones , Nanomedicina , Fármacos Fotosensibilizantes/uso terapéutico
4.
J Biomed Nanotechnol ; 16(2): 179-192, 2020 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-32252879

RESUMEN

Nanobiotechnology strategies for cancer treatments are currently being tested with increasing interest, except in elderly groups. It is well established that breast cancer incidence increases with age and that traditional therapies usually generate severe adverse effects, especially for elderly groups. To investigate if the benefits of nanotechnology could be extended to treating cancer in this group, citrate-coated maghemite nanoparticles (NpCit) were used for magnetohyperthermia (MHT) in combination with the administration of PLGA-Selol nanocapsule (NcSel), a formulation with antioxidant and antitumor activity. The combined therapies significantly inhibited breast Ehrlich tumor growth and prevented metastases to the lymph nodes, liver and lungs until 45 days after tumor induction, a better result than the group undergoing conventional drug treatment. The levels of TNF-α, associated with poor prognosis in Ehrlich tumor, were also normalized. Therefore, the results evidenced the potential use of these therapies for future clinical trials in elderly breast cancer patients.


Asunto(s)
Adenocarcinoma , Envejecimiento , Animales , Línea Celular Tumoral , Glicoles , Humanos , Ratones , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Compuestos de Selenio
5.
Int J Nanomedicine ; 14: 3375-3388, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31123402

RESUMEN

BACKGROUND: Magnetic nanoparticles (MNPs) have been successfully tested for several purposes in medical applications. However, knowledge concerning the effects of nanostructures on elderly organisms is remarkably scarce. PURPOSE: To fill part of this gap, this work aimed to investigate biocompatibility and bio-distribution aspects of magnetic nanoparticles coated with citrate (NpCit) in both elderly and young healthy mice. METHODS: NpCit (2.4 mg iron) was administered intraperitoneally, and its toxicity was evaluated for 28 days through clinical, biochemical, hematological, and histopathological examinations. In addition, its biodistribution was evaluated by spectrometric (inductively coupled plasma optical emission spectrometry) and histological methods. RESULTS: NpCit presented age-dependent effects, inducing very slight and temporary biochemical and hematological changes in young animals. These changes were even weaker than the effects of the aging process, especially those related to the hematological data, tumor necrosis factor alpha, and nitric oxide levels. On the other hand, NpCit showed a distinct set of results in the elderly group, sometimes reinforcing (decrease of lymphocytes and increase of monocytes) and sometimes opposing (erythrocyte parameters and cytokine levels) the aging changes. Leukocyte changes were still observed on the 28th day after treatment in the elderly group. Slight evidence of a decrease in liver and immune functions was detected in elderly mice treated or not treated with NpCit. It was noted that tissue damage or clinical changes related to aging or to the NpCit treatment were not observed. As detected for aging, the pattern of iron biodistribution was significantly different after NpCit administration: extra iron was detected until the 28th day, but in different organs of elderly (liver and kidneys) and young (spleen, liver, and lungs) mice. CONCLUSION: Taken together, the data show NpCit to be a stable and reasonably biocompatible sample, especially for young mice, and thus appropriate for biomedical applications. The data showed important differences after NpCit treatment related to the animals' age, and this emphasizes the need for further studies in older animals to appropriately extend the benefits of nanotechnology to the elderly population.


Asunto(s)
Envejecimiento/fisiología , Ácido Cítrico/farmacología , Materiales Biocompatibles Revestidos/farmacología , Nanopartículas de Magnetita/química , Animales , Femenino , Hierro/química , Pulmón/efectos de los fármacos , Nanopartículas de Magnetita/ultraestructura , Ratones , Óxido Nítrico/sangre , Especificidad de Órganos/efectos de los fármacos , Distribución Tisular/efectos de los fármacos , Factor de Necrosis Tumoral alfa/sangre
6.
Recent Pat Biotechnol ; 12(4): 269-279, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30062978

RESUMEN

BACKGROUND: Carcinoembryonic Antigen (CEA) is a recommended prognostic marker in Colorectal Cancer (CRC) for tumor diagnosis and monitoring response to therapy. High CEA levels are specifically associated with CRC progression and increased levels of the marker are expected to fall following surgical treatment. Due to its role in CRC, CEA has also been explored as a target for cancer therapy and diagnosis approaches. OBJECTIVE: The goal of this work is to highlight the role of CEA in CRC progression and liver metastasis as well as its potential as a biomarker for clinical and biotechnological approaches. METHOD: A literature search of electronic medical and patent databases Pubmed, Scopus, and Science Direct, Google patents, Esp@cenet and United States Patent and Trademark Office (USPTO), was performed. Information was collected in recent publications, including 81 articles besides 13 patents related to different CEA targeting biotechnological approaches for CRC therapy and diagnosis. RESULTS: CEA enhances CRC metastatic potential through many ways. CEA protects metastatic cells from death, changes the microenvironment of sinusoids, promoting the expression of adhesion molecule and malignant cell survival, besides being considered a proangiogenic molecule. Furthermore CEA has also been evaluated as a target in drug delivery systems, photodynamic therapy, radioimmunotherapy, cancer imaging and nanotechnological devices, leading to many patents concerning to development of anti-CEA antibodies or their fragments with potential to target colorectal cancer and liver metastasis cells. CONCLUSION: CEA is already clinically used to monitor CRC patients, and it is a very promising targeting biomarker for multiple biotechnological applications. As far as we know this is the first report on CEA that addresses patents database.


Asunto(s)
Biomarcadores de Tumor/sangre , Antígeno Carcinoembrionario/sangre , Neoplasias Colorrectales/diagnóstico , Neoplasias Hepáticas/diagnóstico , Animales , Biotecnología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Neoplasias Hepáticas/secundario , Patentes como Asunto
7.
Photodiagnosis Photodyn Ther ; 20: 62-70, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28838760

RESUMEN

BACKGROUND: The development of nanocarriers is an important approach to increase the bioavailability of hydrophilic drugs in target cells. In this work, we evaluated the anti-tumorigenic mechanisms and efficacy of NanoALA, a novel nanoformulation of aminolevulic acid (ALA) based on poly(lactide-co-glycolide) (PLGA) nanocapsules designed for anticancer photodynamic therapy (PDT). METHODS: For this purpose, physicochemical characterization, prodrug incorporation kinetics, biocompatibility and photocytotoxicity tests, analysis of the cell death type and mitochondrial function, measurement of the intracellular reactive oxygen species production and DNA fragmentation were performed in murine mammary carcinoma (4T1) cells. RESULTS: NanoALA formulation, stable over a period of 90days following synthesis, presented hydrodynamic diameter of 220±8.7nm, zeta potential of -30.6mV and low value of polydispersity index (0.28). The biological assays indicated that the nanostructured product promotes greater ALA uptake by 4T1 cells and consequently more cytotoxicity in the PDT process. For the first time in the scientific literature, there is a therapeutic efficacy report of approximately 80%, after only 1h of incubation with 100µgmL-1 prodrug (0.6mM ALA equivalent). The mitochondria are probably the initial target of treatment, culminating in energy metabolism disorders and cell death by apoptosis. CONCLUSIONS: NanoALA emerges as a promising strategy for anticancer PDT. Besides being effective against a highly aggressive tumor cell line, the treatment may be economically advantageous because it allows a reduction in the dose and frequency of application compared to free ALA.


Asunto(s)
Ácido Aminolevulínico/administración & dosificación , Nanocápsulas/química , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Ácido Láctico , Potencial de la Membrana Mitocondrial , Ratones , Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Especies Reactivas de Oxígeno/metabolismo
8.
J Biomed Nanotechnol ; 12(4): 689-99, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27301195

RESUMEN

The objective of this study was to investigate the activity of photodynamic therapy mediated by aluminum-chlorophthalocyanine contained in a polymeric nanostructured carrier composed by methyl vinyl ether-co-maleic anhydride (PVM/MA) against local subcutaneous breast cancer tumors and its effects against distant metastasis in a mouse tumor model. In our results, we observed a decrease in breast cancer tumor growth, prevention of distant lung metastases, and a significant increased survival in mice treated with photodynamic therapy. In addition to these results, we observed that tumor-bearing mice without treatment developed a significant extension of liver hematopoiesis that was significantly reduced in mice treated with photodynamic therapy. We hypothesized and showed that this reduction in (1) metastasis and (2) liver hematopoiesis may be related to the systemic activity of immature hematopoietic cells, specifically the myeloid-derived suppressor cells, which were suppressed in mice treated with photodynamic therapy. These cells produce a tolerogenic tumor environment that protects tumor tissues from immunological surveillance. Therefore, we suggest that photodynamic therapy could be employed in combination with other conventional therapies; such as surgery and radiotherapy, to improve the overall survival of patients diagnosed with breast cancer, as observed in our experimental resuIts.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Indoles/administración & dosificación , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Nanocápsulas/química , Compuestos Organometálicos/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Femenino , Indoles/química , Neoplasias Pulmonares/patología , Maleatos/química , Ratones , Ratones Endogámicos BALB C , Nanocápsulas/administración & dosificación , Nanocápsulas/ultraestructura , Compuestos Organometálicos/química , Fármacos Fotosensibilizantes/administración & dosificación , Fármacos Fotosensibilizantes/química , Polietilenos/química , Resultado del Tratamiento
9.
Nanotechnology ; 26(50): 505101, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26580675

RESUMEN

Selol is a semi-synthetic compound containing selenite that is effective against cancerous cells and safer for clinical applications in comparison with other inorganic forms of selenite. Recently, we have developed a formulation of poly(methyl vinyl ether-co-maleic anhydride)-shelled selol nanocapsules (SPN), which reduced the proliferative activity of lung adenocarcinoma cells and presented little deleterious effects on normal cells in in vitro studies. In this study, we report on the antitumor activity and systemic effects induced by this formulation in chemically induced lung adenocarcinoma-bearing mice. The in vivo antitumor activity of the SPN was verified by macroscopic quantification, immunohistochemistry and morphological analyses. Toxicity analyses were performed by evaluations of the kidney, liver, and spleen; analyses of hemogram and plasma levels of alanine aminotransferase, aspartate transaminase, urea, and creatinine; and DNA fragmentation and cell cycle activity of the bone marrow cells. Furthermore, we investigated the potential of the SPN formulation to cause hemolysis, activate the complement system, provoke an inflammatory response and change the conformation of the plasma proteins. Our results showed that the SPN reduced the area of the surface tumor nodules but not the total number of tumor nodules. The biochemical and hematological findings were suggestive of the low systemic toxicity of the SPN formulation. The surface properties of the selol nanocapsules point to characteristics that are consistent with the treatment of the tumors in vivo: low hemolytic activity, weak inflammatory reaction with no activation of the complement system, and mild or absent conformational changes of the plasma proteins. In conclusion, this report suggests that the SPN formulation investigated herein exhibits anti-tumoral effects against lung adenocarcinoma in vivo and is associated with low systemic toxicity and high biocompatibility.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Maleatos/administración & dosificación , Nanocápsulas/administración & dosificación , Polietilenos/administración & dosificación , Compuestos de Selenio/administración & dosificación , Adenocarcinoma/ultraestructura , Adenocarcinoma del Pulmón , Animales , Antineoplásicos/química , Antineoplásicos/toxicidad , Peso Corporal/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas del Sistema Complemento/metabolismo , Fragmentación del ADN/efectos de los fármacos , Femenino , Inflamación/inducido químicamente , Neoplasias Pulmonares/ultraestructura , Maleatos/química , Maleatos/toxicidad , Ratones , Nanocápsulas/química , Nanocápsulas/toxicidad , Tamaño de los Órganos/efectos de los fármacos , Polietilenos/química , Polietilenos/toxicidad , Compuestos de Selenio/química , Compuestos de Selenio/toxicidad
10.
J Nutr Biochem ; 25(11): 1161-1176, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25127291

RESUMEN

The development of therapeutic strategies to attenuate chemotherapy toxicity represents an area of great interest in cancer research, and among them is nutritional therapy based on antioxidants. As research on this topic is still controversial and scarce, we aim to investigate the effects of antioxidant supplementation with vitamin C, vitamin E or pequi oil, a carotenoid-rich oil extracted from pequi (Caryocar brasiliense), on doxorubicin (DX)-induced oxidative damage to normal cells in Ehrlich solid tumor-bearing mice. Tumor weight and volume, histopathology, morphometry and immunohistochemistry were used to assess the treatments' efficacy in containing tumor aggressiveness and regression, while possible toxicity of treatments was assessed by animals' weight, morphological analysis of the heart, liver and kidneys, hemogram, and serum levels of total bilirubin, direct bilirubin, indirect bilirubin, aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma glutamyl transferase (GGT), alkaline phosphatase, creatinine and urea. Although all the chemotherapeutic treatments increased internal necrosis area and reduced the positive Ki-67 cells compared to non-treated tumors, the treatments with pequi oil provided before tumor inoculation (PTDX) or in continuous and concurrent administration with doxorubicin (PTPDX) were more effective in containing tumor growth, besides increasing lymphocyte-dependent immunity and reducing the adverse side effects associated with DX-induced oxidative damage to normal cells, mainly the PTDX treatment. Vitamins C and E given before tumor inoculation and chemotherapy were not successful against doxorubicin-induced cardiotoxicity, besides increasing doxorubicin-induced nephrotoxicity, indicating that, at least for doxorubicin, pequi oil instead of vitamins C and E would be the best option to reduce its adverse effects.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Carcinoma de Ehrlich/tratamiento farmacológico , Carotenoides/análisis , Doxorrubicina/toxicidad , Aceites de Plantas/química , Animales , Antibióticos Antineoplásicos/uso terapéutico , Ácido Ascórbico/análisis , Carcinoma de Ehrlich/metabolismo , Carcinoma de Ehrlich/patología , Doxorrubicina/uso terapéutico , Femenino , Ratones , Estrés Oxidativo/efectos de los fármacos , Aceites de Plantas/farmacología , Vitamina E/análisis
11.
Tumour Biol ; 35(4): 3391-403, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24292954

RESUMEN

Dextran-functionalized maghemite fluid (DexMF) has been tested to treat Ehrlich-solid-tumor-bearing mice, evidencing its potential use in mediating magnetohyperthermia in breast cancer treatment. However, although magnetic nanoparticles tend to accumulate in tumor tissues, part of the nanomaterial can reach the blood stream, and then the organism. The aim of this study was to investigate the acute systemic effects of the intratumoral injection of DexMF mediating magnetohyperthermia in the treatment of an advanced clinical Ehrlich-solid-tumor, assessed through histopathological analyses of liver, kidneys, heart and spleen, comet assay, micronucleus test, hemogram, and serum levels of bilirubin, aspartate aminotransferase, alanine aminotransferase, gamma glutamyl transferase, alkaline phosphatase, creatinine, and urea. The tumor's histopathology and morphometry were used to assess its aggressiveness and regression. DexMF mediating hyperthermia was effective in containing tumor aggressiveness and in inducing tumor regression, besides showing no toxic effects. Its physical characteristics also suggest that it is safe to use in other biomedical applications.


Asunto(s)
Carcinoma de Ehrlich/terapia , Dextranos/administración & dosificación , Hipertermia Inducida/métodos , Magnetoterapia/métodos , Animales , Carcinoma de Ehrlich/genética , Carcinoma de Ehrlich/patología , Ensayo Cometa , Femenino , Hipertermia Inducida/efectos adversos , Magnetoterapia/efectos adversos , Imagen por Resonancia Magnética , Ratones , Micronúcleos con Defecto Cromosómico , gamma-Glutamiltransferasa/sangre
12.
J Nanobiotechnology ; 11: 41, 2013 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-24341795

RESUMEN

BACKGROUND: The increasing incidence of cancer and the search for more effective therapies with minimal collateral effects have prompted studies to find alternative new treatments. Among these, photodynamic therapy (PDT) has been proposed as a very promising new modality in cancer treatment with the lowest rates of side effects, revealing itself to be particularly successful when the photosensitizer is associated with nanoscaled carriers. This study aimed to design and develop a new formulation based on albumin nanospheres containing zinc-phthalocyanine tetrasulfonate (ZnPcS4-AN) for use in the PDT protocol and to investigate its antitumor activity in Swiss albino mice using the Ehrlich solid tumor as an experimental model for breast cancer. METHODS: Ehrlich tumor's volume, histopathology and morphometry were used to assess the efficacy of intratumoral injection of ZnPcS4-AN in containing tumor aggressiveness and promoting its regression, while the toxicity of possible treatments was assessed by animal weight, morphological analysis of the liver and kidneys, hemogram, and serum levels of total bilirubin, direct bilirubin, indirect bilirubin, aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma glutamyl transferase (GGT), alkaline phosphatase, creatinine and urea. In order to evaluate the efficacy of PDT, groups of animals treated with intratumoral injection of doxorubicin (Dox) were also investigated. RESULTS: Intratumoral injection of ZnPcS4-AN was found to be efficient in mediating PDT to refrain tumor aggressiveness and to induce its regression. Although tumor volume reduction was not significant, PDT induced a remarkable increase in the necrosis area seen in the tumor's central region, as in other experimental groups, including tumor and Dox treated groups, but also in the tumor's peripheral region. Further, PDT showed minimal adverse effects. Indeed, the use of ZnPcS4-AN in mediating PDT revealed anti-neoplastic activity similar to that obtained while using intratumoral Dox therapy. CONCLUSIONS: PDT mediated by the new formulation ZnPcS4-AN enhanced the inhibition of tumor growth while producing practically no adverse effects and thus emerges as a very promising nanotechnology-based strategy for solid cancer treatment.


Asunto(s)
Albúminas/química , Carcinoma de Ehrlich/tratamiento farmacológico , Indoles/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Nanosferas/química , Compuestos Organometálicos/farmacología , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Alanina Transaminasa/sangre , Fosfatasa Alcalina/sangre , Animales , Aspartato Aminotransferasas/sangre , Bilirrubina/sangre , Peso Corporal/efectos de los fármacos , Carcinoma de Ehrlich/patología , Creatinina/sangre , Doxorrubicina/farmacología , Femenino , Indoles/química , Inyecciones Intralesiones , Luz , Neoplasias Mamarias Experimentales/patología , Ratones , Compuestos Organometálicos/química , Fármacos Fotosensibilizantes/química , Carga Tumoral/efectos de los fármacos , Urea/sangre , gamma-Glutamiltransferasa/sangre
13.
J Biomed Nanotechnol ; 9(7): 1261-71, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23909142

RESUMEN

This work aimed to test a dextran-functionalized magnetic fluid (DexMF) sample in mediating magnetohyperthermia to treat an advanced clinical Ehrlich-solid-tumor, to verify the effects of oral antioxidant administration of pequi-oil on this treatment and to investigate the potential of these treatments for future use as an adjuvant in cancer therapy. Animals received the treatments: (a) filtered water (control); (b) tumor implantation and no treatment (tumor group); (c) tumor implantation followed by intratumoral injection of DexMF and alternating current magnetic field exposure (MHT group) for three consecutive days; (d) oral pequi-oil supplementation followed by tumor implantation and the same treatment as group MHT (PMHT group). Analyses took place 1 and 2 weeks after tumor implantation. Both treatments were effective in increasing the tumor necrosis process and controlling tumor growth, besides keeping lymphocyte-dependent immunity. Although the MHT treatment was more efficient after the first week in reducing DNA damage to blood peripheral leucocytes, PMHT therapy appeared to be more effective with the advance of the carcinogenesis process after the second week. Our findings evidence the potential use of DexMF mediating magnetohyperthermia in cancer treatment and also suggest that the preventive pequi oil administration could increase the efficiency of this process.


Asunto(s)
Carcinoma de Ehrlich/terapia , Dextranos/química , Hipertermia Inducida/métodos , Magnetoterapia/métodos , Aceites de Plantas/uso terapéutico , Animales , Antioxidantes/administración & dosificación , Carcinoma de Ehrlich/patología , Línea Celular Tumoral , Terapia Combinada/métodos , Estudios de Factibilidad , Resultado del Tratamiento
14.
Exp Parasitol ; 135(2): 217-22, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23891944

RESUMEN

The major goal of this work was to design a new nanoparticle drug delivery system for desoxycholate amphotericin B (D-AMB), based on controlled particle size, looking for the most successful release of the active agents in order to achieve the best site-specific action of the drug at the therapeutically optimal rate and dose regimen. For this, AMB nanoencapsulated in poly(lactic-co-glycolic acid) (PLGA) and dimercaptosuccinic acid (DMSA) nanoparticles (Nano-D-AMB) has been developed, and its efficacy was evaluated in the treatment of experimental cutaneous leishmaniasis in C57BL/6 mice, to test if our nano-drug delivery system could favor the reduction of the dose frequency required to achieve the same therapeutic level of free D-AMB, and so, an extended dosing interval. Magnetic citrate-coated maghemite nanoparticles were added to this nanosystem (Nano-D-AMB-MG) aiming to increase controlled release of AMB by magnetohyperthermia. Female mice (N=6/group) were infected intradermally in the right footpad with promastigotes of Leishmania amazonensis in the metacyclic phase, receiving the following intraperitoneal treatments: 1% PBS for 10 consecutive days; D-AMB at 2 mg/kg/day for 10 days (totalizing 20 mg/kg/animal); Nano-D-AMB and Nano-D-AMB-MG at 6 mg/kg on the 1st, 4th and 7th days and at 2 mg/kg on the 10th day, also totalizing 20 mg/kg/animal by treatment end. The Nano-D-AMB-MG group was submitted to an AC magnetic field, allowing the induction of magnetohyperthermia. The evaluations were through paw diameter measurements; parasite number and cell viability were investigated by limiting dilution assay. D-AMB-coated PLGA-DMSA nanoparticles showed the same efficacy as free D-AMB to reduce paw diameter; however, the Nano-D-AMB treatment also promoted a significantly greater reduction in parasite number and cell viability compared with free D-AMB. The nano-drug AMB delivery system appeared more effective than free D-AMB therapy to reduce the dose frequency required to achieve the same therapeutic level. It thus favors a longer interval between doses, as expected with development of a new nano drug delivery system, and may be useful in the treatment of many different pathologies, from cancer to neurodegenerative diseases.


Asunto(s)
Anfotericina B/administración & dosificación , Antiprotozoarios/administración & dosificación , Leishmania mexicana , Leishmaniasis Cutánea/tratamiento farmacológico , Nanopartículas , Anfotericina B/farmacocinética , Animales , Antiprotozoarios/farmacocinética , Disponibilidad Biológica , Sistemas de Liberación de Medicamentos , Estabilidad de Medicamentos , Femenino , Ácido Láctico , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Nanopartículas/ultraestructura , Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Succímero
15.
Artículo en Inglés | MEDLINE | ID: mdl-23450821

RESUMEN

BACKGROUND: This paper introduces a new nanoformulation of 5-aminolevulinic acid (nano-ALA) as well as a novel quantitative approach towards evaluating field cancerization for actinic keratosis and/or skin photodamage. In this pilot study, we evaluated field cancerization using nano-ALA and methyl aminolevulinate (MAL), the latter being commercialized as Metvix(®). METHODS AND RESULTS: Photodynamic therapy was used for the treatment of patients with selected skin lesions, whereas the fluorescence of the corresponding photosensitizer was used to evaluate the time evolution of field cancerization in a quantitative way. Field cancerization was quantified using newly developed color image segmentation software. Using photodynamic therapy as the precancer skin treatment and the approach introduced herein for evaluation of fluorescent area, we found that the half-life of field cancerization reduction was 43.3 days and 34.3 days for nano-ALA and MAL, respectively. We also found that nano-ALA targeted about 45% more skin lesion areas than MAL. Further, we found the mean reduction in area of skin field cancerization was about 10% greater for nano-ALA than for MAL. CONCLUSION: Although preliminary, our findings indicate that the efficacy of nano-ALA in treating skin field cancerization is higher than that of MAL.

16.
J Nanobiotechnology ; 11: 4, 2013 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-23414068

RESUMEN

BACKGROUND: Magnetic fluids containing superparamagnetic iron oxide nanoparticles represent an attractive platform as nanocarriers in chemotherapy. Recently, we developed a formulation of maghemite nanoparticles coated with rhodium (II) citrate, which resulted in in vitro cytotoxicity enhanced up to 4.6 times when compared to free rhodium (II) citrate formulation on breast carcinoma cells. In this work, we evaluate the antitumor activity and toxicity induced by these formulations in Balb/c mice bearing orthotopic 4T1 breast carcinoma. METHODS: Mice were evaluated with regard to the treatments' toxicity through analyses of hemogram, serum levels of alanine aminotransferase, iron, and creatinine; DNA fragmentation and cell cycle of bone marrow cells; and liver, kidney and lung histology. In addition, the antitumor activity of rhodium (II) citrate and maghemite nanoparticles coated with rhodium (II) citrate was verified by tumor volume reduction, histology and immunohistochemistry. RESULTS: Regarding the treatments' toxicity, no experimental groups had alterations in levels of serum ALT or creatinine, and this suggestion was corroborated by the histopathologic examination of liver and kidney of mice. Moreover, DNA fragmentation frequency of bone marrow cells was lower than 15% in all experimental groups. On the other hand, the complexes rhodium (II) citrate-functionalized maghemite and free rhodium (II) citrate led to a marked growth inhibition of tumor and decrease in CD31 and Ki-67 staining. CONCLUSIONS: In summary, we demonstrated that both rhodium (II) citrate and maghemite nanoparticles coated with rhodium (II) citrate formulations exhibited antitumor effects against 4T1 metastatic breast cancer cell line following intratumoral administration. This antitumor effect was followed by inhibition of both cell proliferation and microvascularization and by tumor tissue injury characterized as necrosis and fibrosis. Remarkably, this is the first published report demonstrating the therapeutic efficacy of maghemite nanoparticles coated with rhodium (II) citrate. This treatment prolonged the survival period of treated mice without inducing apparent systemic toxicity, which strengthens its use for future breast cancer therapeutic applications.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Férricos/química , Nanopartículas de Magnetita/química , Rodio/farmacología , Alanina Transaminasa/sangre , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ácido Cítrico/química , Ácido Cítrico/farmacología , Creatinina/sangre , Fragmentación del ADN/efectos de los fármacos , Femenino , Compuestos Férricos/análisis , Humanos , Inmunohistoquímica , Hierro/sangre , Antígeno Ki-67/análisis , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos BALB C , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Rodio/química , Rayos Ultravioleta
17.
Food Chem Toxicol ; 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23146696

RESUMEN

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

18.
Int J Nanomedicine ; 7: 5271-82, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23055733

RESUMEN

Nanosized maghemite particles were synthesized, precoated (with dimercaptosuccinic acid) and surface-functionalized with anticarcinoembryonic antigen (anti-CEA) and successfully used to target cell lines expressing the CEA, characteristic of colorectal cancer (CRC) cells. The as-developed nanosized material device, consisting of surface decorated maghemite nanoparticles suspended as a biocompatible magnetic fluid (MF) sample, labeled MF-anti-CEA, was characterized and tested against two cell lines: a high-CEA expressing cell line (LS174T) and a low-CEA expressing cell line (HCT116). Whereas X-ray diffraction was used to assess the average core size of the as-synthesized maghemite particles (average 8.3 nm in diameter), dynamic light scattering and electrophoretic mobility measurements were used to obtain the average hydrodynamic diameter (550 nm) and the zeta-potential (-38 mV) of the as-prepared and maghemite-based nanosized device, respectively. Additionally, surface-enhanced Raman spectroscopy (SERS) was used to track the surface decoration of the nanosized maghemite particles from the very first precoating up to the attachment of the anti-CEA moiety. The Raman peak at 1655 cm(-1), absent in the free anti-CEA spectrum, is the signature of the anti-CEA binding onto the precoated magnetic nanoparticles. Whereas MTT assay was used to confirm the low cell toxicity of the MF-anti-CEA device, ELISA and Prussian blue iron staining tests performed with both cell lines (LS174T and HCT116) confirm that the as-prepared MF-anti- CEA is highly specific for CEA-expressing cells. Finally, transmission electron microscopy analyses show that the association with anti-CEA seems to increase the number of LS174T cells with internalized maghemite nanoparticles, whereas no such increase seems to occur in the HCT116 cell line. In conclusion, the MF-anti-CEA sample is a biocompatible device that can specifically target CEA, suggesting its potential use as a theragnostic tool for CEA-expressing tumors, micrometastasis, and cancer-circulating cells.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígeno Carcinoembrionario/inmunología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapéutico , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Compuestos Férricos/química , Humanos , Nanocápsulas/uso terapéutico
19.
Int J Nanomedicine ; 7: 5287-99, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23055734

RESUMEN

Antitumor activities have been described in selol, a hydrophobic mixture of molecules containing selenium in their structure, and also in maghemite magnetic nanoparticles (MNPs). Both selol and MNPs were co-encapsulated within poly(lactic-co-glycolic acid) (PLGA) nanocapsules for therapeutic purposes. The PLGA-nanocapsules loaded with MNPs and selol were labeled MSE-NC and characterized by transmission and scanning electron microscopy, electrophoretic mobility, photon correlation spectroscopy, presenting a monodisperse profile, and positive charge. The antitumor effect of MSE-NC was evaluated using normal (MCF-10A) and neoplastic (4T1 and MCF-7) breast cell lines. Nanocapsules containing only MNPs or selol were used as control. MTT assay showed that the cytotoxicity induced by MSE-NC was dose and time dependent. Normal cells were less affected than tumor cells. Cell death occurred mainly by apoptosis. Further exposure of MSE-NC treated neoplastic breast cells to an alternating magnetic field increased the antitumor effect of MSE-NC. It was concluded that selol-loaded magnetic PLGA-nanocapsules (MSE-NC) represent an effective magnetic material platform to promote magnetohyperthermia and thus a potential system for antitumor therapy.


Asunto(s)
Neoplasias de la Mama/terapia , Hipertermia Inducida/métodos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapéutico , Nanocápsulas/química , Nanocápsulas/uso terapéutico , Compuestos de Selenio/administración & dosificación , Animales , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Femenino , Nanopartículas de Magnetita/ultraestructura , Ratones , Nanocápsulas/ultraestructura , Resultado del Tratamiento
20.
Genet Mol Biol ; 35(1): 153-8, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22481889

RESUMEN

Metformin (dimethyl-biguanide) is an insulin-sensitizing agent that lowers fasting plasma-insulin concentration, wherefore it's wide use for patients with a variety of insulin-resistant and prediabetic states, including impaired glucose tolerance. During pregnancy it is a further resource for reducing first-trimester pregnancy loss in women with the polycystic ovary syndrome. We tested metformin genotoxicity in cells of Chinese hamster ovary, CHO-K1 (chromosome aberrations; comet assays) and in mice (micronucleus assays). Concentrations of 114.4 µg/mL and 572 µg/mL were used in in vitro tests, and 95.4 mg/kg, 190.8 mg/kg and 333.9 mg/kg in assaying. Although the in vitro tests revealed no chromosome aberrations in metaphase cells, DNA damage was detected by comet assaying after 24 h of incubation at both concentrations. The frequency of DNA damage was higher at concentrations of 114.4 µg/mL. Furthermore, although mortality was not observed in in vitro tests, the highest dose of metformin suppressed bone marrow cells. However, no statistically significant differences were noted in micronuclei frequencies between treatments. In vitro results indicate that chronic metformin exposure may be potentially genotoxic. Thus, pregnant woman undergoing treatment with metformin should be properly evaluated beforehand, as regards vulnerability to DNA damage.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA