Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
FASEB J ; 33(8): 9453-9465, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31136203

RESUMEN

Oral consumption of curcumin, a natural polyphenol, is associated with reduced incidence of cancer. Yet, a significant amount of the orally dosed compound is eliminated in the feces, and a major fraction of the absorbed compound is metabolized to inactive glucuronides, resulting in poor bioavailability (<1%). It is not known how oral curcumin exhibits chemopreventive activity. We propose curcumin glucuronide is an inflammation-responsive natural prodrug that is converted back to curcumin on demand at the site of action. Our studies show elevated levels of ß-glucuronidase, an enzyme that hydrolyzes the glycosidic bond of glucuronides to generate the parent compound, in human breast cancer. Oral administration of curcumin in mouse tumor models generated significant tumor levels of the polyphenol. Intravenous administration of the glucuronide resulted in the formation of curcumin in the tumor tissue. Chronic daily oral curcumin dosing led to tumor accumulation of curcumin and inhibition of tumor growth in tumor models with high ß-glucuronidase activity. Overall, the study presented here provides preliminary evidence for a novel mechanism of action for orally administered curcumin.-Liu, G., Khanna, V., Kirtane, A., Grill, A., Panyam, J. Chemopreventive efficacy of oral curcumin: a prodrug hypothesis.


Asunto(s)
Curcumina/análogos & derivados , Curcumina/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Glucurónidos/farmacocinética , Administración Oral , Animales , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Curcumina/administración & dosificación , Curcumina/uso terapéutico , Femenino , Glucuronidasa/metabolismo , Glucurónidos/administración & dosificación , Glucurónidos/uso terapéutico , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Profármacos/administración & dosificación , Profármacos/farmacocinética , Profármacos/uso terapéutico
2.
Mol Pharm ; 17(6): 2109-2124, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32383885

RESUMEN

Activated natural killer (NK) cells can kill malignant tumor cells via granule exocytosis and secretion of IFN-γ, a key regulator of the TH1 response. Thus, mobilization of NK cells can augment cancer immunotherapy, particularly when mediated through antibody-dependent cellular cytotoxicity (ADCC). Stimulation of toll-like receptor (TLR)7/8 activity in dendritic cells promotes pro-inflammatory cytokine secretion and costimulatory molecule upregulation, both of which can potentiate NK cell activation. However, currently available TLR7/8 agonists exhibit unfavorable pharmacokinetics, limiting their in vivo efficacy. To enable efficient delivery to antigen-presenting cells, we encapsulated a novel imidazoquinoline-based TLR7/8 agonist in pH-responsive polymeric NPs. Enhanced costimulatory molecule expression on dendritic cells and a stronger pro-inflammatory cytokine response were observed with a NP-encapsulated agonist, compared to that with the soluble form. Treatment with NP-encapsulated agonists resulted in stronger in vivo cytotoxicity and prolonged activation of NK cells compared to that with a soluble agonist. In addition, TLR7/8 agonist-loaded NPs potentiated stronger NK cell degranulation, which resulted in enhanced in vitro and in vivo ADCC mediated by the epidermal growth factor receptor-targeting antibody cetuximab. TLR7/8 agonist-loaded NP treatment significantly enhanced the antitumor efficacy of cetuximab and an anti-HER2/neu antibody in mouse tumor models. Collectively, our data show that a pH-responsive NP-encapsulating TLR7/8 agonist could be used as a potent immunostimulatory adjuvant for antibody-based cancer immunotherapy by promoting NK cell activation.


Asunto(s)
Inmunoterapia/métodos , Células Asesinas Naturales/metabolismo , Nanopartículas/química , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Células A549 , Animales , Células Dendríticas/metabolismo , Citometría de Flujo , Humanos , Imiquimod/química , Células Asesinas Naturales/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanomedicina/métodos
3.
Mol Pharm ; 16(3): 1200-1210, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30620878

RESUMEN

Cancer vaccines composed of tumor-associated antigens (TAAs) and toll-like receptor (TLR) agonists have shown promising antitumor efficacy in preclinical studies by generating antigen-specific CD8 T cells, but translation of cancer vaccines to the clinic has been limited due to variables responses and development of resistance. The tumor microenvironment deploys various immune escape mechanisms that neutralize CD8 T cell-mediated tumor rejection. Therefore, we hypothesized that modulation of the tumor microenvironment can augment CD8 T cell activation and enhance therapeutic efficacy of cancer vaccines. To accomplish this, we aimed to eliminate immune suppressive cells and block their inhibitory signaling. Combination of the tyrosine kinase inhibitor (TKI) sunitinib with a nanoparticle-based cancer vaccine (nanovaccine) resulted in the reduction of immune-suppressive myeloid-derived suppressive cells (MDSCs) and regulatory T cells (Tregs). Blockade of programmed death-ligand 1 (PD-L1) using anti-PD-L1 antibody was used to reduce CD8 T cell exhaustion. Combination of nanovaccine+sunitinib+PD-L1 antibody treatment reduced PD-L1high M2 macrophages and MDSCs and upregulated activation of CD8 T cells in the tumor. Nanovaccine+sunitinib+PD-L1 antibody treatment also stimulated antigen-specific CD8 T cell response, which led to improved therapeutic efficacy in MB49 and B16F10 murine tumor models. These results suggest that modulation of tumor microenvironment using sunitinib and PD-L1 blockade can significantly enhance the antitumor efficacy of cancer nanovaccine.


Asunto(s)
Anticuerpos/uso terapéutico , Antígeno B7-H1/inmunología , Vacunas contra el Cáncer/uso terapéutico , Glicoproteínas de Membrana/agonistas , Neoplasias/terapia , Sunitinib/uso terapéutico , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Animales , Antígenos de Neoplasias/inmunología , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Femenino , Interleucina-10/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Linfocitos T Reguladores/inmunología , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Vacunación
4.
J Pharm Bioallied Sci ; 14(Suppl 1): S233-S235, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36110830

RESUMEN

Objectives: To assess the utility of lycopene, selenium, and vitamin E in the management of leukoplakia. Materials and Methods: In this cross-sectional observational prospective study, 52 patients of oral leukoplakia were divided into 2 groups. Group I patients were prescribed 6 mg of lycopene, 400 I.U. of vitamin E, and 200 µg of selenium twice a day. Group II patients were prescribed a placebo capsule. The clinical improvement was recorded in both groups. Results: The mean pretreatment size of lesion in group I was 9.24 cm2 and in group II was 8.52 cm2. Post treatment size was 1.52 cm2 in group I and 6.28 cm2 in group II. There was a significant reduction in the size of lesion in group I compared to group II (P < 0.05). Conclusion: The treatment with a combination of lycopene, selenium, and vitamin E is useful and effective in managing the cases of oral leukoplakia.

5.
Sci Rep ; 11(1): 3346, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33558639

RESUMEN

There is a significant interest in designing therapeutic agents that can enhance ADCC and thereby improve clinical responses with approved antibodies. We recently reported the combination of an imidazoquinoline-based TLR7/8 agonist (522) with a monoclonal antibody improved ADCC in vitro and in vivo. In the present study, we tested several new small molecule TLR7/8 agonists that induce significantly higher cytokines compared to both the FDA-approved TLR7 agonist, imiquimod, and 522. We evaluated these agonists in combination with monoclonal antibody therapy, with the main goal of enhancing ADCC. Our studies show these TLR7/8 agonists induce robust pro-inflammatory cytokine secretion and activate NK cells. Specifically, we found the agonists 574 and 558 significantly enhanced NK cell-mediated ADCC in vitro as well as enhanced the anti-cancer efficacy of monoclonal antibodies in two different in vivo mouse models. Additionally, we found the agonists were able to stimulate CD8 T cells, likely indicative of an early adaptive immune response.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Antineoplásicos Inmunológicos/farmacología , Linfocitos T CD8-positivos/inmunología , Imiquimod/farmacología , Células Asesinas Naturales/inmunología , Neoplasias Experimentales , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Células A549 , Animales , Humanos , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Immunotherapy ; 12(4): 255-267, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32208784

RESUMEN

Over the last decade, antibodies have become an important component in the arsenal of cancer therapeutics. High-specificity, low off-target effects, desirable pharmacokinetics and high success rate are a few of the many attributes that make antibodies amenable for development as drugs. To design antibodies for successful clinical applications, however, it is critical to have an understanding of their structure, functions, mechanisms of action and pharmacokinetic/pharmacodynamic properties. This review highlights some of these key aspects, as well as certain limitations encountered, with monoclonal antibody therapy. Further, we discuss rational combination therapies for clinical applications, some of which could help overcome the limitations.


Asunto(s)
Anticuerpos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Anticuerpos/química , Anticuerpos/clasificación , Anticuerpos/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/inmunología , Quimioterapia Combinada , Humanos , Inhibidores de Puntos de Control Inmunológico , Neoplasias/inmunología
7.
Future Drug Discov ; 1(1): FDD8, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31448368

RESUMEN

AIM: We previously developed two antibodies that bind to a cell surface protein, perlecan, overexpressed in triple-negative breast cancer (TNBC). The goal of this study was to investigate these antibodies as targeting ligands for nanoparticle-mediated drug delivery. METHODS: Paclitaxel-loaded poly(D,L-lactide-co-glycolide) nanoparticles were functionalized with antibodies using thiol-maleimide chemistry. Effect of antibody functionalization on therapeutic efficacy of drug-loaded nanoparticles was investigated using in vitro and in vivo models of TNBC. RESULTS: The antibodies were covalently conjugated to nanoparticles without affecting antibody binding affinity or nanoparticle properties. Perlecan-targeted nanoparticles showed improved cell uptake, retention, cytotoxicity in vitro and enhanced tumor growth inhibition in vivo. CONCLUSION: The data presented here indicates that perlecan-targeted nanoparticles can improve tumor drug delivery to TNBC.

8.
Sci Rep ; 9(1): 12492, 2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31462656

RESUMEN

In recent years, there have been significant advances in the treatment of breast cancer resulting in remarkably high survival rates. However, treatment options for metastatic triple negative breast cancer (TNBC) are quite limited due to a lack of identifiable, unique markers. Using a phage display-based whole cell biopanning procedure, we developed two human antibodies that bind to tumor cells with a metastatic TNBC phenotype. Our studies further identified domain 1 of HSPG2 (perlecan) protein as the cognate cell surface antigen bound by the antibody. Immunohistochemistry studies utilizing patient tissue samples revealed significant cell surface expression of HSPG2 in both primary tumors and metastatic lesions. Further, higher HSPG2 expression correlated with poor survival in TNBC. The affinity-matured antibody inhibited the growth of triple negative MDA-MB-231 tumors to a greater extent in nude mice than in NSG mice, pointing to the potential role of natural killer cell-mediated antibody-dependent cell cytotoxicity. This mechanism of action was confirmed through in vitro assays using mouse splenocytes and human peripheral blood mononuclear cells (PBMCs). These results suggest that HSPG2 is a promising target in metastatic TNBC and HSPG2-targeted antibodies could represent a potentially novel class of targeted therapeutics for TNBC.


Asunto(s)
Antineoplásicos Inmunológicos , Proteoglicanos de Heparán Sulfato , Proteínas de Neoplasias , Neoplasias de la Mama Triple Negativas , Adulto , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Femenino , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/inmunología , Humanos , Ratones Desnudos , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Res ; 77(6): 1465-1475, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28108516

RESUMEN

Elevated interstitial fluid pressure and solid stress within tumors contribute to poor intratumoral distribution of nanomedicine. In this study, we hypothesized that the presence of fibrin in tumor extracellular matrix contributes to hindered intratumoral distribution of nanocarriers and that this can be overcome through the use of a fibrinolytic enzyme such as tissue plasminogen activator (tPA). Analysis of fibrin expression in human tumor biopsies showed significant fibrin staining in nearly all tumor types evaluated. However, staining was heterogeneous across and within tumor types. We determined the effect of fibrin on the diffusion, intratumoral distribution, and therapeutic efficacy of nanocarriers. Diffusivity of nanocarriers in fibrin matrices was limited and could be improved significantly by coincubation with tPA. In vivo, coadministration of tPA improved the anticancer efficacy of nanoparticle-encapsulated paclitaxel in subcutaneous syngeneic mouse melanoma and orthotopic xenograft lung cancer models. Furthermore, treatment with tPA led to decompression of blood vessels and improved tumor perfusion. Cotreatment with tPA resulted in greater intratumoral penetration of a model nanocarrier (Doxil), leading to enhanced availability of the drug in the tumor core. Fibrinolytics such as tPA are already approved for other indications. Fibrinolytic cotherapy is therefore a rapidly translatable strategy for improving therapeutic effectiveness of anticancer nanomedicine. Cancer Res; 77(6); 1465-75. ©2017 AACR.


Asunto(s)
Albúminas/administración & dosificación , Fibrina/metabolismo , Fibrinolíticos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Nanomedicina , Paclitaxel/administración & dosificación , Activador de Tejido Plasminógeno/farmacología , Animales , Femenino , Fibrinólisis , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Int Oral Health ; 5(5): 124-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24324316

RESUMEN

Adenomatoid odontogenic tumor is a relatively uncommon distinct odontogenic neoplasm. It is an uncommon tumor of odontogenic origin with varying number of ductlike structures and inductive changes in the stroma. It is a benign and slow growing epithelial tumor and represents 3% of all odontogenic tumors. Its occurrence is more common in anterior region of the maxilla than mandible. Most of the adenomatoid odontogenic tumors occur intra-osseously but few peripheral variant have been reported which are attached to the gingival structures. The intra-osseous Adenomatoid odontogenic tumor may be related to unerrupted tooth (follicular varient) or may not (extrafollicular varient) be related to unerrupted tooth. This paper is to present a rare case of an extrafollicular Adenomatoid odontogenic tumor occurring in the body of the mandible in a male patient which is distinct and secondly it was clinically and radiographically diagnosed as residual cyst. The diagnosis of Adenomatoid odontogenic tumor was confirmed by Histopathological investigation. How to cite this article: Shivali V, Khanna VD, Khanna P, Singh A, Pandey A, Ahuja T. A Rare Case of Extrafollicular Adenomatoid Odontogenic Tumour in the Posterior Region of the Mandible: Misdiagnosed as Residual Cyst. J Int Oral Health 2013; 5(5):124-8.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA