Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nanomaterials (Basel) ; 14(3)2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38334590

RESUMO

Approximately 10% of women suffer from endometriosis during their reproductive years. This disease is a chronic debilitating condition whose etiology for lesion implantation and survival heavily relies on adhesion and angiogenic factors. Currently, there are no clinically approved agents for its detection. In this study, we evaluated cRGD-peptide-conjugated nanoparticles (RGD-Cy5.5-MN) to detect lesions using magnetic resonance imaging (MRI) in a mouse model of endometriosis. We utilized a luciferase-expressing murine suture model of endometriosis. Imaging was performed before and after 24 h following the intravenous injection of RGD-Cy5.5-MN or control nanoparticles (Cy5.5-MN). Next, we performed biodistribution of RGD-Cy5.5-MN and correlative fluorescence microscopy of lesions stained for CD34. Tissue iron content was determined using inductively coupled plasma optical emission spectrometry (ICP-OES). Our results demonstrated that targeting endometriotic lesions with RGD-Cy5.5-MN resulted in a significantly higher delta T2* upon its accumulation compared to Cy5.5-MN. ICP-OES showed significantly higher iron content in the lesions of the animals in the experimental group compared to the lesions of the animals in the control group. Histology showed colocalization of Cy5.5 signal from RGD-Cy5.5-MN with CD34 in the lesions pointing to the targeted nature of the probe. This work offers initial proof-of-concept for targeting angiogenesis in endometriosis which can be useful for potential clinical diagnostic and therapeutic approaches for treating this disease.

2.
Commun Biol ; 6(1): 815, 2023 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-37542121

RESUMO

Small extracellular vesicles (sEV) in TNBC patients' plasma promote T cell dysfunction and tumor progression. Here we show that tumor cell-derived exosomes (TEX) carrying surface PDL-1, PD-1, Fas, FasL, TRAIL, CTLA-4 and TGF-ß1 induce apoptosis of CD8+T and CD4+T cells but spare B and NK cells. Inhibitors blocking TEX-induce receptor/ligand signals and TEX pretreatments with proteinase K or heat fail to prevent T cell apoptosis. Cytochalasin D, Dynosore or Pit Stop 2, partly inhibit TEX uptake but do not prevent T cell apoptosis. TEX entry into T cells induces cytochrome C and Smac release from mitochondria and caspase-3 and PARP cleavage in the cytosol. Expression of survival proteins is reduced in T cells undergoing apoptosis. Independently of external death receptor signaling, TEX entry into T cells induces mitochondrial stress, initiating relentless intrinsic apoptosis, which is responsible for death of activated T cells in the tumor-bearing hosts. The abundance of TEX in cancer plasma represents a danger for adoptively transferred T cells, limiting their therapeutic potential.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Caspases/metabolismo , Apoptose , Proteínas Reguladoras de Apoptose , Linfócitos T/metabolismo
3.
Front Mol Biosci ; 10: 1179343, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37398551

RESUMO

Introduction: Recent studies have shown that miRNA-10b is highly expressed in high-grade glioblastoma multiforme (GBM), and its inhibition leads to deregulation of multiple pathways in tumorigenesis, resulting in repression of tumor growth and increased apoptosis. Thus, we hypothesized that suppressing miR-10b could enhance the cytotoxicity of conventional GBM chemotherapy with temozolomide (TMZ). Methods: Inhibition of miR-10b in glioblastoma cells was achieved using an experimental therapeutic consisting of anti-miR10b antagomirs conjugated to iron oxide nanoparticles (termed MN-anti-miR10b). The nanoparticles serve as delivery vehicles for the antagomirs as well as imaging reporters guiding the delivery in future animal studies. Results: Treatment of U251 and LN229 human glioblastoma cells with MN-anti-miR10b led to inhibition of miR-10b accompanied by repression of growth and increase in apoptosis. We next explored whether MN-anti-miR10b could enhance the cytotoxic effect of TMZ. During these studies, we unexpectedly found that TMZ monotherapy increased miR-10b expression and changed the expression of corresponding miR-10b targets. This discovery led to the design of a sequence-dependent combination treatment, in which miR-10b inhibition and induction of apoptosis by MN-anti-miR10b was followed by a sub-therapeutic dose of TMZ, which caused cell cycle arrest and ultimately cell death. This combination was highly successful in significant enhancement of apoptosis and decrease in cell migration and invasiveness. Discussion: Considering the unexpected effects of TMZ on miR-10b expression and possible implications on its clinical application, we reasoned that comprehensive in vitro studies were warranted before embarking on studies in animals. These intriguing findings serve as a solid foundation for future in vivo studies and offer promise for the successful treatment of GBM.

4.
Mol Pharm ; 20(6): 2822-2835, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37134112

RESUMO

Progesterone (PR) is an endogenous steroid hormone that activates the progesterone receptor (PgR) and is known to play a critical role in cancer progression. Herein, we report the development of cationic lipid-conjugated PR derivatives by covalently conjugating progesterone with cationic lipids of varying hydrocarbon chain lengths (n = 6-18) through a succinate linker. Cytotoxicity studies performed on eight different cancer cell lines reveal that PR10, one of the lead derivatives, exerts notable toxicity (IC50 = 4-12 µM) in cancer cells irrespective of their PgR expression status and remains largely nontoxic to noncancerous cells. Mechanistic studies show that PR10 induces G2/M-phase cell cycle arrest in cancer cells, leading to apoptosis and cell death by inhibiting the PI3K/AKT cell survival pathway and p53 upregulation. Further, in vivo study shows that PR10 treatment significantly reduces melanoma tumor growth and prolongs the overall survival of melanoma tumor-bearing C57BL/6J mice. Interestingly, PR10 readily forms stable self-aggregates of ∼190 nm size in an aqueous environment and exhibits selective uptake into cancerous cell lines. In vitro uptake mechanism studies in various cell lines (cancerous cell lines B16F10, MCF7, PC3, and noncancerous cell line HEK293) using endocytosis inhibition proves that PR10 nanoaggregates enter selectively into the cancer cells predominantly using macropinocytosis and/or caveolae-mediated endocytosis. Overall, this study highlights the development of a self-aggregating cationic derivative of progesterone with anticancer activity, and its cancer cell-selective accumulation in nanoaggregate form holds great potential in the field of targeted drug delivery.


Assuntos
Melanoma , Progesterona , Camundongos , Animais , Humanos , Progesterona/farmacologia , Linhagem Celular Tumoral , Fosfatidilinositol 3-Quinases/metabolismo , Células HEK293 , Camundongos Endogâmicos C57BL , Apoptose , Melanoma/tratamento farmacológico , Lipídeos/farmacologia , Proliferação de Células
5.
Chem Asian J ; 18(2): e202201136, 2023 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-36482874

RESUMO

Paclitaxel (PTX) is a widely used chemotherapeutic agent in the clinic. However, its clinical benefit is limited due to its low water solubility, off-target toxicity, and for being a multidrug-resistant (MDR) substrate. To overcome these limitations in this study, a tumor-targeting peptide (CRGDK peptide, a ligand for NRP-1 receptor) conjugate of α-tocopheryl succinate (α-TOS) was synthesized and modified on PTX-loaded lipid aggregate (TL-PTX) to leverage the benefits of α-TOS, which include a) anti-cancer activity, b) increased PTX loading, and c) inhibition of MDR activity. Use of peptide conjugate of α-TOS (α-TOS-CRGDK) in lipid aggregate increased PTX entrapment efficiency by 20%, helped in NRP-1 specific cellular uptake and significantly enhanced apoptotic and cell killing activity (p <0.01) of PTX compared to control formulation (CL-PTX) by inhibiting MDR-activity in melanoma resulting in ∼70% increment in overall survival of melanoma tumor-bearing mice. In conclusion, CRGDK- α-TOS conjugate in association with PTX-loaded liposome provided a unique NRP-1 targeted, drug-resistant reversing anticancer regimen for treating aggressive melanoma.


Assuntos
Melanoma , Paclitaxel , Camundongos , Animais , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , alfa-Tocoferol/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Peptídeos/farmacologia , Lipídeos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos
6.
Melanoma Res ; 31(5): 472-475, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34284459

RESUMO

Cancer liquid biopsy encompassing circulating tumor cells (CTC), circulating tumor DNA (ctDNA) and/or tumor-derived exosomes (TEX) emerges as a novel approach to early detection, noninvasive monitoring of responses to therapy and predicting patient survival. TEX are a key component of liquid biopsy because they mimic tumor cells in their proteomic and genetic content. Two recent proteomic analyses of TEX released into plasma by melanoma cells confirms the potential of TEX as diagnostic and prognostic markers in melanoma.


Assuntos
Biomarcadores Tumorais/sangue , Exossomos/patologia , Melanoma/patologia , Células Neoplásicas Circulantes/patologia , Proteoma/metabolismo , Progressão da Doença , Exossomos/metabolismo , Humanos , Melanoma/metabolismo , Células Neoplásicas Circulantes/metabolismo , Prognóstico , Proteoma/análise , Taxa de Sobrevida
7.
Nanomedicine (Lond) ; 16(8): 641-656, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33769068

RESUMO

Background: Thymoquinone (TQ) has potential anti-inflammatory, immunomodulatory and anticancer effects but its clinical use is limited by its low solubility, poor bioavailability and rapid clearance. Aim: To enhance systemic bioavailability and tumor-specific toxicity of TQ. Materials & methods: Cationic liposomal formulation of TQ (D1T) was prepared via ethanol injection method and their physicochemical properties, anticancer effects in orthotopic xenograft pancreatic tumor model and pharmacokinetic behavior of D1T relative to TQ were evaluated. Results: D1T showed prominent inhibition of pancreatic tumor progression, significantly greater in vivo absorption, approximately 1.5-fold higher plasma concentration, higher bioavailability, reduced volume of distribution and improved clearance relative to TQ. Conclusion: Encapsulation of TQ in cationic liposomal formulation enhanced its bioavailability and anticancer efficacy against xenograft pancreatic tumor.


Assuntos
Lipossomos , Benzoquinonas , Disponibilidade Biológica , Linhagem Celular Tumoral , Humanos , Solubilidade
8.
Methods Mol Biol ; 2265: 305-321, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33704724

RESUMO

Tumor-derived exosomes (TEX), a subset of small extracellular vesicles (EVs) which originate from the endocytic compartment of tumor cells, are emerging as key players in cancer progression. TEX circulate freely in patients' body fluids and transfer bioactive cargos from tumor to various recipient cells. The molecular cargo of melanoma cell-derived exosomes (MTEX) mimics that of the tumor, and MTEX serve as a liquid biopsy that provides potentially useful information for cancer diagnosis, prognosis, or responses to therapy. Plasma of melanoma patients contains a mix of MTEX and exosomes produced by nonmalignant cells (NMTEX). Isolation of these exosome subtypes from the bulk of plasma exosomes is necessary to evaluate contributions of each as potential biomarkers of melanoma progression and outcome. Here, methods for separation of MTEX from T cell-derived exosomes from a single small volume of plasma and their subsequent molecular and functional characterization are described. Following size exclusion chromatography (SEC) to isolate total plasma exosomes, immune affinity-based capture of MTEX with anti-CSPG4 antibody and then of exosomes produced by T cells with anti-CD3 antibody is used to sequentially isolate the two subsets. This immune capture method enables the recovery of MTEX and CD3+ exosomes in quantities sufficient both for molecular profiling by flow cytometry or western blotting and for functional analyses.


Assuntos
Biomarcadores Tumorais/sangue , Western Blotting , Exossomos/metabolismo , Citometria de Fluxo , Melanoma/sangue , Linfócitos T/metabolismo , Cromatografia em Gel , Exossomos/patologia , Humanos , Biópsia Líquida , Plasma/metabolismo , Linfócitos T/patologia
9.
Oncogene ; 39(48): 7114-7126, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33005016

RESUMO

Glioblastoma multiforme (GBM) is a highly proliferative and locally invasive cancer with poor prognosis and a high recurrence rate. Although anti-VEGF (vascular endothelial growth factor) therapy offers short-term benefit to GBM patients, this approach fails as the tumor develops into a more invasive and drug-resistant phenotype and ultimately recurs. Recently, both glioma stemlike cells (GSCs) and brain tumor-initiating cells (BTICs) have been implicated in GBM recurrence and its resistance to therapy. We observed that patient-derived GBM cells expressing shRNAs of VEGF or neuropilin-1 (NRP-1) attenuate cancer stem cell markers, inhibit the tumor-initiating cell's neurosphere-forming capacity, and migration. Furthermore, both VEGF and NRP-1 knockdown inhibit the growth of patient-derived GBM xenografts in both zebrafish and mouse models. Interestingly, NRP-1-depleted patient-derived GBM xenografts substantially prolonged survival in mice compared to that of VEGF depletion. Our results also demonstrate that NRP-1 ablation of patient-derived GBM cells improves the sensitivity of TMZ and enhances the overall survival of the respective tumor-bearing mice. This improved outcome may provide insight into the inhibition of GBM progression and effective treatment strategies by targeting NRP-1 in addition to chemotherapy and radiotherapy.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Técnicas de Silenciamento de Genes , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Neuropilina-1/deficiência , Neuropilina-1/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transformação Celular Neoplásica , Glioblastoma/patologia , Humanos , Camundongos , Fenótipo , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Nanotoxicology ; 13(9): 1161-1175, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31294672

RESUMO

Cationic lipids are well-known excipients for nanometric liposomal gene delivery systems. However, because of the suspected, collateral toxicity in normal cells, the use of cationic lipids for the treatment of human tumor is largely limited. Recently, we developed a glucocorticoid receptor (GR)-targeted liposomal, anticancer delivery system (DXE nano-lipoplex), which carried cationic lipid of saturated twin aliphatic chains. It exhibited efficient anti-tumor effect in aggressive and drug-resistant tumor models. Toward exploring lipoplex's human clinical use, we incorporated another nano-lipoplex (D1XE) group that carried cationic lipid with one of its aliphatic chain carrying unsaturation and compared in vivo genotoxicological profiling-based safety assessment and the respective anti-tumor efficacy of the lipoplexes. Thus, both the lipoplexes differ only by the chemical identity of one of their constituent cationic lipid. Unsaturated aliphatic chains in lipid generally impart efficient cell surface fusogenic property in lipid formulations. Herein, we report that nanoplex with unsaturated cationic lipid (D1XE) exhibited better physical appearance with less flocculent behavior than nanoplex with saturated lipid (DXE). Upon multiple injections, D1XE nanoplex imparted better tumor regression but most importantly, exhibited much lower overall toxicity (e.g. genotoxicity, weight loss, etc.) than DXE nanoplex. With a higher antitumor effect but a lower genotoxic effect, D1XE is proved to be a better nanoplex than DXE for the potential clinical trial. Thus, this study clearly delineates the importance of incorporating a constituent lipid that carries a single unsaturated aliphatic chain toward developing efficient anti-tumor nano-lipoplexes with reduced genotoxicity.


Assuntos
Lipídeos/química , Nanoestruturas/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Cátions , Aberrações Cromossômicas , Dano ao DNA , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Lipossomos , Masculino , Camundongos , Nanoestruturas/toxicidade , Transfecção
11.
Life Sci ; 203: 305-314, 2018 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-29729261

RESUMO

AIMS: Zoledronic acid (ZA), a FDA approved drug has used widely in the treatment of bone metastasis complications, has been linked to renal toxicity with unclear mechanism. The present study is aimed at investigating the genotoxic and cytotoxic effects of ZA in renal epithelial cells. MAIN METHODS: The genotoxic effect of ZA in Vero and MDCK cells determined by cytokinesis block micronucleus (CBMN) assay. The cytotoxic effect assessed by analysing cell cycle profile, cell death and mitochondrial membrane potential by flow cytometry using propidium iodide, AnnexinV-FITC/PI and JC1 dye staining, respectively, BAX and Bcl-2 expression by Western blotting and caspase activity by spectrofluorimetry. KEY FINDINGS: The cytotoxic effect of ZA based on MTT assay revealed variable sensitivities of Vero and MDCK cells, with IC50 values of 7.41 and 109.58 µM, respectively. The CBMN assay has shown prominent dose-dependent (IC10-50) induction of micronuclei formation in both cells, indicating ZA's clastogenic and aneugenic potential. Further, the ZA treatment led the cells to apoptosis, evident from dose-dependent increase in the percentage of cells in subG1 phase and display of membranous phosphatidylserine translocation. Studies also confirmed apoptosis through mitochondria, evident from the prominent increase in BAX/Bcl-2 ratio, mitochondrial membrane depolarization and caspase-3/7 activity. In addition, ZA reduces cytokinetic activity of renal cells, evident from dose-wise lowered replicative indices. SIGNIFICANCE: The study depict ZA's potential genotoxic effect along with cytotoxic effect in renal epithelial cells, could be key factors for the development of renal complications associated with it, which prompts renal safety measures in lieu with ZA usage.


Assuntos
Apoptose/efeitos dos fármacos , Citocinese/efeitos dos fármacos , Difosfonatos/farmacologia , Imidazóis/farmacologia , Rim/patologia , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Mitocôndrias/patologia , Animais , Conservadores da Densidade Óssea/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Chlorocebus aethiops , Cães , Rim/efeitos dos fármacos , Células Madin Darby de Rim Canino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Células Vero , Ácido Zoledrônico
12.
Biomed Pharmacother ; 99: 956-969, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29710496

RESUMO

Dehydrocostus lactone (DHCL), a sesquiterpene lactone is well-known for its antiulcer, anti-hepatotoxic and anticancer activity. However, the studies concerning the safety/toxicity potential of DHCL toward the cells of normal origin remain unclear. The present study is aimed at investigating the toxicity potential of DHCL in renal distal tubular and interstitial fibroblast cell lines (MDCK and NRK-49F cells, respectively), and also in ovarian epithelial cell line (CHO cells). The MTT assay has predicted potential cytotoxic activity of DHCL against the cell line types with IC50 values of 0.99, 2.1 and 5.15??M, respectively. The prominent dose-dependent (IC30,50 & 70) increase in the percentage of cells at subG1 phase in all the cell lines revealed apoptosis induction, further establishing the cytotoxic effect of DHCL. The DHCL exposure (4?h) revealed the induction of ROS in both renal cell lines, which is responsible for apoptosis induction. The NRK-49F cells displayed dose-wise (IC30-70) increase in chromatin condensation and membranous phosphatidylserine translocation further confirming apoptotic cell death. Also, their increase in BAX/Bcl-2 ratio, mitochondrial membrane permeability and caspase-3/7 activity establishes mitochondrial mediated apoptosis. In case of CHO cells, the higher percentage of cells at G2/M phase and expression of Cyclin B1 at lower concentration of DHCL (?IC30), indicate mitotic arrest. The incidence of chromatid gaps and negligible micronuclei formation in treated cells (IC10-30) suggest that sub-lethal concentrations of DHCL exposure causes mitotic arrest in response to the damages by steady expression of Cyclin B1. Under in vitro condition, the study of DHCL's potential cytotoxic effect on both kidney cells and ovarian epithelial cells indicated the possibility of adverse effects on normal healthy cells as well. Hence, the study recommends in-depth investigations on DHCL usage concerning its safety in therapeutic applications.


Assuntos
Apoptose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Lactonas/farmacologia , Sesquiterpenos/farmacologia , Animais , Células CHO , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Cricetinae , Cricetulus , Cães , Relação Dose-Resposta a Droga , Células Epiteliais/citologia , Feminino , Fibroblastos/citologia , Concentração Inibidora 50 , Túbulos Renais Distais/citologia , Túbulos Renais Distais/efeitos dos fármacos , Lactonas/administração & dosagem , Células Madin Darby de Rim Canino , Ovário/citologia , Ovário/efeitos dos fármacos , Ratos , Sesquiterpenos/administração & dosagem
13.
Cell Biol Toxicol ; 34(2): 125-142, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28914393

RESUMO

Costunolide (CE) is a sesquiterpene lactone well-known for its antihepatotoxic, antiulcer, and anticancer activities. The present study focused on the evaluation of the cytogenetic toxicity and cellular death-inducing potential of CE in CHO cells, an epithelial cell line derived from normal ovary cells of Chinese hamster. The cytotoxic effect denoting MTT assay has shown an IC50 value of 7.56 µM CE, where 50% proliferation inhibition occurs. The oxidative stress caused by CE was confirmed based on GSH depletion induced cell death, conspicuously absent in N-acetylcysteine (GSH precursor) pretreated cells. The evaluation of genotoxic effects of CE using cytokinesis block micronucleus assay and chromosomal aberration test has shown prominent induction of binucleated micronucleated cells and aberrant metaphases bearing chromatid and chromosomal breaks, indicating CE's clastogenic and aneugenic potential. The apoptotic death in CE treated cells was confirmed by an increase in the number of cells in subG1 phase, exhibiting chromatin condensation and membranous phosphatidylserine translocation. The apoptosis induction follows mitochondrial mediation, evident from an increase in the BAX/Bcl-2 ratio, caspase-3/7 activity, and mitochondrial membrane permeability. CE also induces cytostasis in addition to apoptosis, substantiated by the reduced cytokinetic (replicative indices) and mitotic (mitotic indices and histone H3 Ser-10 phosphorylation) activities. Overall, the cellular GSH depletion and potential genotoxic effects by CE led the CHO cells to commit apoptosis and lowered cell division. The observed sensitivity of CHO cells doubts unintended adverse effects of CE on normal healthy cells, suggesting higher essentiality of further studies in order to establish its safety efficacy in therapeutic explorations.


Assuntos
Apoptose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Micronúcleos com Defeito Cromossômico/induzido quimicamente , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Sesquiterpenos/toxicidade , Animais , Células CHO , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Células Epiteliais/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Saussurea/química , Sesquiterpenos/isolamento & purificação
14.
Mol Cell Biochem ; 436(1-2): 119-136, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28585089

RESUMO

Glucocorticoid, such as dexamethasone (Dex) is often used along with chemotherapy to antagonize side effects of chemotherapy. However, sustained use of Dex frequently develops drug resistance in patients. As a strategy to re-induce drug sensitivity, we planned to modify Dex by chemically conjugating it with twin ten carbon aliphatic chain containing cationic lipid. The resultant molecule, DX10, inhibited STAT3 activation through lowering the production of IL-6. To enhance the STAT3 inhibitory effect of DX10, we used WP (a commercially available STAT3 inhibitor) along with DX10. Combination treatment of both significantly inhibited STAT3 activation when compared to either of the individual treatment. The effect of DX10, either in combination or alone, was mediated through glucocorticoid receptor (GR), thereby repurposing the role of GR in the context of p-STAT3 inhibition-mediated cancer treatment. Cellular viability study proved the synergistic effect of WP and DX10. Further, combination treatment led to induction of early stage of apoptosis and cell cycle arrest. In vivo melanoma tumor regression study confirmed the enhanced anti-tumor activity of co-treatment over individual treatment of DX10 or WP. Thus, together our result demonstrates that DX10 may be used in combination therapy with STAT3 inhibitor like WP for combating cancer with constitutively active STAT3.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Melanoma/tratamento farmacológico , Proteínas de Neoplasias/antagonistas & inibidores , Fator de Transcrição STAT3/antagonistas & inibidores , Células A549 , Animais , Células CHO , Ensaios Clínicos Fase I como Assunto , Cricetulus , Dexametasona/análogos & derivados , Dexametasona/farmacologia , Humanos , Células MCF-7 , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/metabolismo , Piridinas/farmacologia , Tirfostinas/farmacologia
15.
Biomed Chromatogr ; 30(12): 2024-2030, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27306429

RESUMO

A lipid-conjugated, estrogenic derivative molecule, ESC8, compared with other estrogenic molecules, encourages cell death in both ER-positive and ER-negative breast cancer cells. A rapid and highly sensitive assay method has been developed and validated for the estimation of a ESC8 in rat plasma using liquid chromatography coupled with mass spectrometry under positive-ion mode with electrospray ionization. The sample process includes using methanol for precipitation of ESC8 and dextromethorphan (internal standard, IS) from plasma. Chromatographic separation was achieved with methanol-water-formic acid (70:30:0.1% v/v/v) pumped at a flow rate of 0.3mL/min and a C18 column (50 × 2.1 mm i.d., 1.7 µm particle size) with a total run time of 5 min. The m/z ions monitored were 568.5 and 272.1 for ESC8 and IS, respectively. The lower limit of quantitation achieved was 1.08 ng/mL and linearity was observed from 5 to 500 ng/mL. The intra- and inter-day precisions were <4%. The proposed method was successfully applied to a preliminary pharmacokinetic study of ESC8 liposomal formulation following an intraperitoneal administration of 3.67 mg/kg in rats. The concentrations of ESC8 in plasma were quantifiable up to 36 h. The peak concentration of ESC8 was found to be 110.72 ng/mL, the area under the concentration-time curve was 1625.23ng/mL h and the half-life was 11.72 h.


Assuntos
Cromatografia Líquida/métodos , Estrogênios/sangue , Lipídeos/química , Espectrometria de Massas/métodos , Animais , Calibragem , Estrogênios/farmacocinética , Limite de Detecção , Ratos , Padrões de Referência , Reprodutibilidade dos Testes
16.
Mol Pharm ; 13(7): 2507-23, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27184196

RESUMO

Many cancers including the late stage ones become drug-resistant and undergo epithelial-to-mesenchymal transition (EMT). These lead to enhanced invasion, migration, and metastasis toward manifesting its aggressiveness and malignancy. One of the key hallmarks of cancer is its overdependence on glycolysis as its preferred energy metabolism pathway. The strict avoidance of alternate energy pathway gluconeogenesis by cancer cells points to a yet-to-be hoisted role of glucocorticoid receptor (GR) especially in tumor microenvironment, where cells are known to become drug-sensitive through induction of gluconeogenesis. However, since GR is involved in metabolism, anti-inflammatory reactions, immunity besides inducing gluconeogenesis, a greater role of GR in tumor microenvironment is envisaged. We have shown previously that GR, although ubiquitously expressed in all cells; afford to be an effective cytoplasmic target for killing cancer cells selectively. Herein, we report the therapeutic use of a newly developed GR-targeted liposomal concoction (DXE) coformulating a lipophilic drug (ESC8) and an anti-Hsp90 anticancer gene against aggressive tumor models. This induced drug-sensitivity and apoptosis while reversing EMT in tumor cells toward effective retardation of aggressive growth in pancreas and skin tumor models. Additionally, the ESC8-free lipid formulation upon cotreatment with hydrophilic drugs, gemcitabine and doxorubicin, could effectively sensitize and kill pancreatic cancer and melanoma cells, respectively. The formulation-triggered EMT-reversal was GR-dependent. Overall, we found a new strategy for drug sensitization that led to the advent of new GR-targeted anticancer therapeutics.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Proteínas de Choque Térmico HSP90/metabolismo , Lipossomos/química , Receptores de Glucocorticoides/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , DNA/química , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Transição Epitelial-Mesenquimal/genética , Proteínas de Choque Térmico HSP90/genética , Humanos , Lipossomos/administração & dosagem , Camundongos , Reação em Cadeia da Polimerase , Receptores de Glucocorticoides/genética , Gencitabina
17.
Mater Sci Eng C Mater Biol Appl ; 63: 393-410, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27040234

RESUMO

Two different silica based (MSU-2 and MCM-41) curcumin loaded mesoporous materials V3 and V6 were synthesized and characterized by several physico-chemical techniques. Release kinetic study revealed the slow and sustained release of curcumin from those materials in blood simulated fluid (pH: 7.4). The materials V3 and V6 were found to be biocompatible in non-cancerous CHO cell line while exhibiting significant cytotoxicity in different cancer cells (human lung carcinoma cells: A549, human breast cancer cells: MCF-7, mouse melanoma cells: B16F10) compared to pristine curcumin indicating the efficacy of the mesoporous silica materials based drug delivery systems (DDSs). The generation of intracellular reactive oxygen species (ROS) and down regulation of anti-apoptotic protein leading to the induction of apoptosis were found to be the plausible mechanisms behind the anti-cancer activity of these DDSs. These results suggest that curcumin-loaded drug delivery system may be successfully employed as an alternative treatment strategy for cancer therapeutics through a nanomedicine approach in near future.


Assuntos
Antineoplásicos/química , Curcumina/química , Portadores de Fármacos/química , Dióxido de Silício/química , Células A549 , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Curcumina/farmacologia , Citometria de Fluxo , Humanos , Células MCF-7 , Camundongos , Nanomedicina , Porosidade , Espécies Reativas de Oxigênio
18.
Mol Pharm ; 13(3): 1081-8, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26832839

RESUMO

The objective of the present study is to develop a liposomal formulation for delivering anticancer drug to breast cancer stem-cell-like cells, ANV-1, and evaluate its pharmacokinetics in an animal model. The anticancer drug ESC8 was used in dexamethasone (Dex)-associated liposome (DX) to form ESC8-entrapped liposome named DXE. ANV-1 cells showed high-level expression of NRP-1. To enhance tumor regression, we additionally adapted to codeliver the NRP-1 shRNA-encoded plasmid using the established DXE liposome. In vivo efficacy of DXE-NRP-1 was carried out in mice bearing ANV-1 cells as xenograft tumors and the extent of tumor growth inhibition was evaluated by tumor-size measurement. A significant difference in tumor volume started to reveal between DXE-NRP-1 group and DXE-Control group. DXE-NRP-1 group showed ∼4 folds and ∼2.5 folds smaller tumor volume than exhibited by untreated and DXE-Control-treated groups, respectively. DXE disposition was evaluated in Sprague-Dawley rats following an intraperitoneal dose (3.67 mg/kg of ESC8 in DXE). The plasma concentrations of ESC8 in the DXE formulation were measured by liquid chromatography mass spectrometry and pharmacokinetic parameters were determined using a noncompartmental analysis. ESC8 had a half-life of 11.01 ± 0.29 h, clearance of 2.10 ± 3.63 L/kg/h, and volume of distribution of 33.42 ± 0.83 L/kg. This suggests that the DXE liposome formulation could be administered once or twice daily for therapeutic efficacy. In overall, we developed a potent liposomal formulation with favorable pharmacokinetic and tumor regressing profile that could sensitize and kill highly aggressive and drug-resistive cancer stem-cell-like cells.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/farmacocinética , Neoplasias da Mama/tratamento farmacológico , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Lipossomos/química , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Composição de Medicamentos , Transição Epitelial-Mesenquimal , Feminino , Humanos , Masculino , Camundongos , Células-Tronco Neoplásicas/metabolismo , Neuropilina-1/antagonistas & inibidores , Neuropilina-1/genética , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...