Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Bioelectron Med ; 10(1): 10, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38594769

RESUMO

BACKGROUND: Glioblastoma (GBM) presents as an aggressive brain cancer, notorious for its recurrence and resistance to conventional treatments. This study aimed to assess the efficacy of the EMulate Therapeutics Voyager®, a non-invasive, non-thermal, non-ionizing, battery-operated, portable experimental medical device, in treating GBM. Using ultra-low radiofrequency energy (ulRFE) to modulate intracellular activity, previous preliminary results in patients have been encouraging. Now, with a focus on murine models, our investigation seeks to elucidate the device's mechanistic impacts, further optimizing its therapeutic potential and understanding its limitations. METHODS: The device employs a silicone over molded coil to deliver oscillating magnetic fields, which are believed to interact with and disrupt cellular targets. These fields are derived from the magnetic fluctuations of solvated molecules. Xenograft and syngeneic murine models were chosen for the study. Mice were injected with U-87 MG or GL261 glioma cells in their flanks and were subsequently treated with one of two ulRFE cognates: A1A, inspired by paclitaxel, or A2, based on murine siRNA targeting CTLA4 + PD1. A separate group of untreated mice was maintained as controls. RESULTS: Mice that underwent treatments with either A1A or A2 exhibited significantly reduced tumor sizes when compared to the untreated cohort. CONCLUSION: The EMulate Therapeutics Voyager® demonstrates promising potential in inhibiting glioma cells in vivo through its unique ulRFE technology and should be further studied in terms of biological effects in vitro and in vivo.

2.
Nature ; 618(7966): 842-848, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37258671

RESUMO

Nonsense mutations are the underlying cause of approximately 11% of all inherited genetic diseases1. Nonsense mutations convert a sense codon that is decoded by tRNA into a premature termination codon (PTC), resulting in an abrupt termination of translation. One strategy to suppress nonsense mutations is to use natural tRNAs with altered anticodons to base-pair to the newly emerged PTC and promote translation2-7. However, tRNA-based gene therapy has not yielded an optimal combination of clinical efficacy and safety and there is presently no treatment for individuals with nonsense mutations. Here we introduce a strategy based on altering native tRNAs into  efficient suppressor tRNAs (sup-tRNAs) by individually fine-tuning their sequence to the physico-chemical properties of the amino acid that they carry. Intravenous and intratracheal lipid nanoparticle (LNP) administration of sup-tRNA in mice restored the production of functional proteins with nonsense mutations. LNP-sup-tRNA formulations caused no discernible readthrough at endogenous native stop codons, as determined by ribosome profiling. At clinically important PTCs in the cystic fibrosis transmembrane conductance regulator gene (CFTR), the sup-tRNAs re-established expression and function in cell systems and patient-derived nasal epithelia and restored airway volume homeostasis. These results provide a framework for the development of tRNA-based therapies with a high molecular safety profile and high efficacy in targeted PTC suppression.


Assuntos
Códon sem Sentido , Regulador de Condutância Transmembrana em Fibrose Cística , RNA de Transferência , Animais , Camundongos , Aminoácidos/genética , Códon sem Sentido/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , RNA de Transferência/administração & dosagem , RNA de Transferência/genética , RNA de Transferência/uso terapêutico , Pareamento de Bases , Anticódon/genética , Biossíntese de Proteínas , Mucosa Nasal/metabolismo , Perfil de Ribossomos
3.
Biomed Pharmacother ; 151: 113156, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35643066

RESUMO

BACKGROUND AND AIMS: The sexual dimorphism in fat-mass distribution and circulating leptin and insulin levels is well known, influencing the progression of obesity-associated metabolic disease. Here, we aimed to investigate the possible role of lipopolysaccharide-binding protein (LBP) in this sexual dimorphism. METHODS: The relationship between plasma LBP and fat mass was evaluated in 145 subjects. The effects of Lbp downregulation, using lipid encapsulated unlocked nucleomonomer agent containing chemically modified-siRNA delivery system, were evaluated in mice. RESULTS: Plasma LBP levels were associated with fat mass and leptin levels in women with obesity, but not in men with obesity. In mice, plasma LBP downregulation led to reduced weight, fat mass and leptin gain after a high-fat and high-sucrose diet (HFHS) in females, in parallel to increased expression of adipogenic and thermogenic genes in visceral adipose tissue. This was not observed in males. Plasma LBP downregulation avoided the increase in serum LPS levels in HFHS-fed male and female mice. Serum LPS levels were positively correlated with body weight and fat mass gain, and negatively with markers of adipose tissue function only in female mice. The sexually dimorphic effects were replicated in mice with established obesity. Of note, LBP downregulation led to recovery of estrogen receptor alpha (Esr1) mRNA levels in females but not in males. CONCLUSION: LBP seems to exert a negative feedback on ERα-mediated estrogen action, impacting on genes involved in thermogenesis. The known decreased estrogen action and negative effects of metabolic endotoxemia may be targeted through LBP downregulation.


Assuntos
Leptina , Lipopolissacarídeos , Proteínas de Fase Aguda , Tecido Adiposo , Animais , Proteínas de Transporte , Dieta Hiperlipídica , Regulação para Baixo , Estrogênios/metabolismo , Feminino , Humanos , Leptina/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo
4.
Sci Rep ; 10(1): 8096, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32415084

RESUMO

Abnormal regulation of ß-catenin initiates an oncogenic program that serves as a main driver of many cancers. Albeit challenging, ß-catenin is an attractive drug target due to its role in maintenance of cancer stem cells and potential to eliminate cancer relapse. We have identified C2, a novel ß-catenin inhibitor, which is a small molecule that binds to a novel allosteric site on the surface of ß-catenin. C2 selectively inhibits ß-catenin, lowers its cellular load and significantly reduces viability of ß-catenin-driven cancer cells. Through direct binding to ß-catenin, C2 renders the target inactive that eventually activates proteasome system for its removal. Here we report a novel pharmacologic approach for selective inhibition of ß-catenin via targeting a cryptic allosteric modulation site. Our findings may provide a new perspective for therapeutic targeting of ß-catenin.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/antagonistas & inibidores , Regulação Alostérica , Animais , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Apoptose , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias/metabolismo , Neoplasias/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/isolamento & purificação , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncotarget ; 8(14): 22370-22384, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-26517684

RESUMO

Transcription factors (TFs) are a major class of protein signaling molecules that play key cellular roles in cancers such as the highly lethal brain cancer-glioblastoma (GBM). However, the development of specific TF inhibitors has proved difficult owing to expansive protein-protein interfaces and the absence of hydrophobic pockets. We uniquely defined the dimerization surface as an expansive parental pharmacophore comprised of several regional daughter pharmacophores. We targeted the OLIG2 TF which is essential for GBM survival and growth, we hypothesized that small molecules able to fit each subpharmacophore would inhibit OLIG2 activation. The most active compound was OLIG2 selective, it entered the brain, and it exhibited potent anti-GBM activity in cell-based assays and in pre-clinical mouse orthotopic models. These data suggest that (1) our multiple pharmacophore approach warrants further investigation, and (2) our most potent compounds merit detailed pharmacodynamic, biophysical, and mechanistic characterization for potential preclinical development as GBM therapeutics.


Assuntos
Antineoplásicos/uso terapêutico , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Neoplasias Encefálicas/tratamento farmacológico , Desenho de Fármacos , Glioblastoma/tratamento farmacológico , Guanidinas/uso terapêutico , Terapia de Alvo Molecular , Proteínas do Tecido Nervoso/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Processos de Crescimento Celular , Sobrevivência Celular/genética , Simulação por Computador , Humanos , Camundongos , Camundongos Nus , Estrutura Molecular , Proteínas do Tecido Nervoso/química , Fator de Transcrição 2 de Oligodendrócitos , Ligação Proteica , Conformação Proteica , Bibliotecas de Moléculas Pequenas , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Oncotarget ; 7(36): 57932-57942, 2016 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-27462771

RESUMO

Zoledronic acid, a potent nitrogen-containing bisphosphonate (NBP), has been extensively used to limit bone turnover in a various diseases including tumors. Recent clinical studies have demonstrated direct anti-cancer effects of zoledronic acid, in addition to its clinical benefits for skeletal-related events. Here we investigated the effects of 4 clinically available NBPs on human tumor cell proliferation. Our data demonstrate a potent anti-proliferative effect of zoledronic acid against glioblastoma (GBM) cell lines, breast cancer cells and GBM patient-derived lines. Zoledronic acid also effectively inhibited GBM tumor growth in xenograft mouse models. Zoledronic acid strongly stimulated autophagy but not apoptotic signals in all tested cells. Only one intermediate product of cholesterols synthesis pathway, geranylgeranyl diphosphate (GGPP) rescued cells from the cytotoxic effects of zoledronic acid. To further investigate the effect of GGPP, we knocked down RABGGTA, which encodes a subunit of the Rabgeranylgeranyltransferase protein. This knockdown induced an effect similar to zoledronic acid in cancer cell lines. These data are promising and suggested a potential for zoledronic acid as an anti-cancer agent, through its ablation of the function of Rab proteins.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Difosfonatos/farmacologia , Nitrogênio/química , Animais , Antineoplásicos/química , Autofagia , Conservadores da Densidade Óssea/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Difosfonatos/química , Ensaios de Seleção de Medicamentos Antitumorais , Glioblastoma/tratamento farmacológico , Humanos , Imidazóis/química , Células MCF-7 , Camundongos , Transplante de Neoplasias , Ácido Zoledrônico
7.
J Transl Med ; 13: 269, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26283544

RESUMO

BACKGROUND: The STAT3 transcription factor is a major intracellular signaling protein and is frequently dysregulated in the most common and lethal brain malignancy in adults, glioblastoma multiforme (GBM). Activation of STAT3 in GBM correlates with malignancy and poor prognosis. The phosphorylating signal transducer JAK2 activates STAT3 in response to cytokines and growth factors. Currently there are no JAK-STAT pathway inhibitors in clinical trials for GBM, so we sought to examine the anti-GBM activity of SAR317461 (Sanofi-Aventis), a newer generation, highly potent JAK2 inhibitor that exhibits low toxicity and good pharmacokinetics. SAR317461 was initially approved for patient testing in the treatment of primary myelofibrosis (PMF), and has shown activity in preclinical models of melanoma and pulmonary cancer, but has not been tested in GBM. METHODS: We hypothesized that a potent small molecule JAK2 inhibitor could overcome the heterogeneous nature of GBM, and suppress a range of patient derived GBM tumorsphere lines and immortalized GBM cell lines. We treated with SAR317461 to determine IC50 values, and using Western blot analysis we asked whether the response was linked to STAT3 expression. Western blot analysis, FACS, and cell viability studies were used to identify the mechanism of SAR317461 induced cell death. RESULTS: We report for the first time that the JAK2 inhibitor SAR317461 clearly inhibited STAT3 phosphorylation and had substantial activity against cells (IC50 1-10 µM) from 6 of 7 different patient GSC derived GBM tumorsphere lines and three immortalized GBM lines. One patient GSC derived line did not constitutively express STAT3 and was more resistant to SAR317461 (IC50 ≈25 µM). In terms of mechanism we found cleaved PARP and clear apoptosis following SAR317461. SAR317461 also induced autophagy and the addition of an autophagy inhibitor markedly enhanced cell killing by SAR317461. CONCLUSIONS: We conclude that SAR317461 potently inhibits STAT3 phosphorylation and that it has significant activity against those GBM cells which express activated STAT3. Further studies are warranted in terms of the potential of SAR317461 as single and combined therapy for selectively treating human patients afflicted with GBMs expressing activation of the JAK2-STAT3 signaling axis.


Assuntos
Neoplasias Encefálicas/metabolismo , Inibidores Enzimáticos/química , Glioblastoma/metabolismo , Janus Quinase 2/antagonistas & inibidores , Mielofibrose Primária/metabolismo , Pirimidinas/química , Fator de Transcrição STAT3/antagonistas & inibidores , Sulfonamidas/química , Adolescente , Adulto , Idoso , Autofagia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Separação Celular , Sobrevivência Celular , Regulação para Baixo , Feminino , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Humanos , Concentração Inibidora 50 , Masculino , Pessoa de Meia-Idade , Fosforilação , Modelos de Riscos Proporcionais , Adulto Jovem
8.
Oncotarget ; 6(2): 1157-70, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25528767

RESUMO

Metabolic reprogramming is a key feature of tumorigenesis that is controlled by oncogenes. Enhanced utilization of glucose and glutamine are the best-established hallmarks of tumor metabolism. The oncogene c-Myc is one of the major players responsible for this metabolic alteration. However, the molecular mechanisms involved in Myc-induced metabolic reprogramming are not well defined. Here we identify p32, a mitochondrial protein known to play a role in the expression of mitochondrial respiratory chain complexes, as a critical player in Myc-induced glutamine addiction. We show that p32 is a direct transcriptional target of Myc and that high level of Myc in malignant brain cancers correlates with high expression of p32. Attenuation of p32 expression reduced growth rate of glioma cells expressing Myc and impaired tumor formation in vivo. Loss of p32 in glutamine addicted glioma cells induced resistance to glutamine deprivation and imparted sensitivity to glucose withdrawal. Finally, we provide evidence that p32 expression contributes to Myc-induced glutamine addiction of cancer cells. Our findings suggest that Myc promotes the expression of p32, which is required to maintain sufficient respiratory capacity to sustain glutamine metabolism in Myc transformed cells.


Assuntos
Neoplasias Encefálicas/genética , Proteínas de Transporte/genética , Glioma/genética , Glutamina/metabolismo , Proteínas Mitocondriais/genética , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proteínas de Transporte/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Glioma/patologia , Humanos , Immunoblotting , Imuno-Histoquímica , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Proteínas Mitocondriais/metabolismo , Modelos Genéticos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Transl Med ; 12: 128, 2014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24884660

RESUMO

BACKGROUND: Glioblastoma (GBM) is an aggressive disease associated with poor survival. It is essential to account for the complexity of GBM biology to improve diagnostic and therapeutic strategies. This complexity is best represented by the increasing amounts of profiling ("omics") data available due to advances in biotechnology. The challenge of integrating these vast genomic and proteomic data can be addressed by a comprehensive systems modeling approach. METHODS: Here, we present an in silico model, where we simulate GBM tumor cells using genomic profiling data. We use this in silico tumor model to predict responses of cancer cells to targeted drugs. Initially, we probed the results from a recent hypothesis-independent, empirical study by Garnett and co-workers that analyzed the sensitivity of hundreds of profiled cancer cell lines to 130 different anticancer agents. We then used the tumor model to predict sensitivity of patient-derived GBM cell lines to different targeted therapeutic agents. RESULTS: Among the drug-mutation associations reported in the Garnett study, our in silico model accurately predicted ~85% of the associations. While testing the model in a prospective manner using simulations of patient-derived GBM cell lines, we compared our simulation predictions with experimental data using the same cells in vitro. This analysis yielded a ~75% agreement of in silico drug sensitivity with in vitro experimental findings. CONCLUSIONS: These results demonstrate a strong predictability of our simulation approach using the in silico tumor model presented here. Our ultimate goal is to use this model to stratify patients for clinical trials. By accurately predicting responses of cancer cells to targeted agents a priori, this in silico tumor model provides an innovative approach to personalizing therapy and promises to improve clinical management of cancer.


Assuntos
Ensaios de Seleção de Medicamentos Antitumorais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Simulação por Computador , Humanos , Estudos Retrospectivos
10.
J Control Release ; 183: 146-53, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24685706

RESUMO

There is a great interest in targeting and selective ablation of populations of circulating cells for research or therapeutic purposes. Red blood cells (RBCs) are readily available and fully biocompatible long-circulating intravascular carriers (natural life is 120days) that are amenable to chemical modifications, drug loading and reinjection. Here we demonstrate that using our previously described lipophilic ligand painting strategy, red blood cells (RBCs) could be in one step converted into targeted entities that selectively seek and bind various cells in vitro and in vivo. In vitro, RBCs modified with lipophilic anti-EpCAM or anti-CD45 antibodies efficiently bound to cancer cells and leukocytes, forming characteristic rosettes. In vivo, intravenously injected RBCs painted with anti-CD45 antibody immediately associated with CD45 positive cells in blood, forming RBC-leukocyte rosettes. Moreover, anti-CD45-modified RBCs, but not the same amount of anti-CD45 antibody or anti-CD45-lipid conjugate (1-2µg/mouse), depleted over 50% of CD45+ leukocytes from circulation, with main clearance organs of leukocytes being liver and spleen with no visible deposition in kidneys and lungs. Anti-CD20 (Rituximab)-painted RBCs efficiently (over 90%) depleted CD19+/CD20+/CD45+ human lymphoma cells in mantle cell lymphoma (MCL) JeKo-1 model, while the same amount of rituximab-lipid (2µg/mouse) was much less efficient in lymphoma cell depletion. Treatment of MCL mice with rituximab-modified RBCs carrying only 2µg of the antibody resulted in a significant prolongation of survival as compared to the same amount of antibody-lipid control. Lipophilic ligand-painted RBCs is a novel tool that can be utilized for targeting blood borne cells for experimental immunology and drug delivery applications.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Antígenos de Neoplasias/imunologia , Antineoplásicos/administração & dosagem , Portadores de Fármacos , Eritrócitos/imunologia , Leucócitos/imunologia , Células Neoplásicas Circulantes/imunologia , Animais , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Reações Antígeno-Anticorpo/imunologia , Antígenos de Neoplasias/sangue , Antígenos de Neoplasias/metabolismo , Antineoplásicos/sangue , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Portadores de Fármacos/química , Eritrócitos/química , Eritrócitos/citologia , Feminino , Humanos , Procedimentos de Redução de Leucócitos , Leucócitos/patologia , Ligantes , Camundongos Endogâmicos BALB C , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patologia , Fosfatidiletanolaminas/química , Polietilenoglicóis/química , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Transl Med ; 12: 13, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24433351

RESUMO

BACKGROUND: Glioblastoma (GBM) is a therapeutic challenge, associated with high mortality. More effective GBM therapeutic options are urgently needed. Hence, we screened a large multi-class drug panel comprising the NIH clinical collection (NCC) that includes 446 FDA-approved drugs, with the goal of identifying new GBM therapeutics for rapid entry into clinical trials for GBM. METHODS: Screens using human GBM cell lines revealed 22 drugs with potent anti-GBM activity, including serotonergic blockers, cholesterol-lowering agents (statins), antineoplastics, anti-infective, anti-inflammatories, and hormonal modulators. We tested the 8 most potent drugs using patient-derived GBM cancer stem cell-like lines. Notably, the statins were active in vitro; they inhibited GBM cell proliferation and induced cellular autophagy. Moreover, the statins enhanced, by 40-70 fold, the pro-apoptotic activity of irinotecan, a topoisomerase 1 inhibitor currently used to treat a variety of cancers including GBM. Our data suggest that the mechanism of action of statins was prevention of multi-drug resistance protein MDR-1 glycosylation. This drug combination was synergistic in inhibiting tumor growth in vivo. Compared to animals treated with high dose irinotecan, the drug combination showed significantly less toxicity. RESULTS: Our data identifies a novel combination from among FDA-approved drugs. In addition, this combination is safer and well tolerated compared to single agent irinotecan. CONCLUSIONS: Our study newly identifies several FDA-approved compounds that may potentially be useful in GBM treatment. Our findings provide the basis for the rational combination of statins and topoisomerase inhibitors in GBM.


Assuntos
Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Aprovação de Drogas , Glioblastoma/tratamento farmacológico , United States Food and Drug Administration , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Autofagia/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Camptotecina/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Irinotecano , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/patologia , Quinolinas/administração & dosagem , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Esferoides Celulares/patologia , Estados Unidos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Int J Nanomedicine ; 8: 3991-4006, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24174874

RESUMO

Staurosporine (STS) is a potent pan-kinase inhibitor with marked activity against several chemotherapy-resistant tumor types in vitro. The translational progress of this compound has been hindered by poor pharmacokinetics and toxicity. We sought to determine whether liposomal encapsulation of STS would enhance antitumor efficacy and reduce toxicity, thereby supporting the feasibility of further preclinical development. We developed a novel reverse pH gradient liposomal loading method for STS, with an optimal buffer type and drug-to-lipid ratio. Our approach produced 70% loading efficiency with good retention, and we provide, for the first time, an assessment of the in vivo antitumor activity of STS. A low intravenous dose (0.8 mg/kg) inhibited U87 tumors in a murine flank model. Biodistribution showed preferential tumor accumulation, and body weight data, a sensitive index of STS toxicity, was unaffected by liposomal STS, but did decline with the free compound. In vitro experiments revealed that liposomal STS blocked Akt phosphorylation, induced poly(ADP-ribose) polymerase cleavage, and produced cell death via apoptosis. This study provides a basis to explore further the feasibility of liposomally encapsulated STS, and potentially related compounds for the management of resistant solid tumors.


Assuntos
Lipossomos/administração & dosagem , Lipossomos/química , Nanocápsulas/administração & dosagem , Nanocápsulas/química , Neoplasias Experimentais/tratamento farmacológico , Proteína Quinase C/antagonistas & inibidores , Estaurosporina/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Difusão , Feminino , Humanos , Lipossomos/ultraestrutura , Masculino , Camundongos , Camundongos Nus , Nanocápsulas/ultraestrutura , Neoplasias Experimentais/patologia , Estaurosporina/química , Resultado do Tratamento
13.
Methods ; 64(2): 102-7, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23974072

RESUMO

With the emerging interest in personalized medicine, there is strong demand for new technologies for clinical sample interrogation. Exfoliated tumor cells in variety of pathological samples (e.g., blood, bone marrow, urine) could provide invaluable information for diagnosis and prognosis of cancers. Here we describe a detailed method for capture and isolation of tumor cells in medium, blood, or large issue buffy coat using EpCAM-targeted buoyant microbubbles (MBs). Perflorohexane gas lipid shell MBs were prepared with emulsification method and conjugated with antibody as described by us before [25]. The binding of EpCAM-targeted MBs to A549 (human lung carcinoma) and 4T1 (mouse breast carcinoma) cells spiked into BSA/PBS or blood was more than 90%, which was comparable with commercial anti-EpCAM immunomagnetic beads (DynaBeads). Anti-EpCAM MBs efficiently (75-82%) isolated BxPC3 pancreatic tumor cells spiked into medium, blood or a buffy coat, within 15-30 min of incubation. We discuss MB parameters and experimental conditions critical to achieve efficient cells binding and isolation. In conclusion, MB-assisted cell isolation is a promising method for rapid enrichment of cells and biomarkers from biological samples.


Assuntos
Separação Celular/métodos , Fluorocarbonos , Microbolhas , Células Neoplásicas Circulantes/patologia , Animais , Antígenos de Neoplasias/imunologia , Moléculas de Adesão Celular/imunologia , Linhagem Celular Tumoral , Células Imobilizadas/citologia , Molécula de Adesão da Célula Epitelial , Feminino , Humanos , Neoplasias Mamárias Animais/patologia , Camundongos
14.
ACS Nano ; 7(5): 4289-98, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23614696

RESUMO

Scavenger receptors (SRs) are molecular pattern recognition receptors that have been shown to mediate opsonin-independent uptake of therapeutic and imaging nanoparticles, underlying the importance of SRs in nanomedicine. Unlike pathogens, engineered nanomaterials offer great flexibility in control of surface properties, allowing addressing specific questions regarding the molecular mechanisms of nanoparticle recognition. Recently, we showed that SR-type AI/II mediates opsonin-independent internalization of dextran superparamagnetic iron oxide (SPIO) nanoparticles via positively charged extracellular collagen-like domain. To understand the mechanism of opsonin-independent SPIO recognition, we tested the binding and uptake of nanoparticles with different surface coatings by SR-AI. SPIO coated with 10 kDa dextran was efficiently recognized and taken up by SR-AI transfected cells and J774 macrophages, while SPIO with 20 kDa dextran coating or cross-linked dextran hydrogel avoided the binding and uptake. Nanoparticle negative charge density and zeta-potential did not correlate with SR-AI binding/uptake efficiency. Additional experiments and computer modeling revealed that recognition of the iron oxide crystalline core by the positively charged collagen-like domain of SR-AI is sterically hindered by surface polymer coating. Importantly, the modeling revealed a strong complementarity between the surface Fe-OH groups of the magnetite crystal and the charged lysines of the collagen-like domain of SR-AI, suggesting a specific recognition of SPIO crystalline surface. These data provide an insight into the molecular recognition of nanocrystals by innate immunity receptors and the mechanisms whereby polymer coatings promote immune evasion.


Assuntos
Compostos Férricos/química , Compostos Férricos/metabolismo , Macrófagos/metabolismo , Nanopartículas/química , Receptores Depuradores Classe A/metabolismo , Animais , Transporte Biológico , Dextranos/química , Células HEK293 , Humanos , Camundongos , Modelos Moleculares , Conformação Molecular , Ligação Proteica , Receptores Depuradores Classe A/química
15.
PLoS One ; 8(3): e58017, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23516425

RESUMO

Circulating tumor cells (CTCs) are exfoliated at various stages of cancer, and could provide invaluable information for the diagnosis and prognosis of cancers. There is an urgent need for the development of cost-efficient and scalable technologies for rare CTC enrichment from blood. Here we report a novel method for isolation of rare tumor cells from excess of blood cells using gas-filled buoyant immuno-microbubbles (MBs). MBs were prepared by emulsification of perfluorocarbon gas in phospholipids and decorated with anti-epithelial cell adhesion molecule (EpCAM) antibody. EpCAM-targeted MBs efficiently (85%) and rapidly (within 15 minutes) bound to various epithelial tumor cells suspended in cell medium. EpCAM-targeted MBs efficiently (88%) isolated frequent tumor cells that were spiked at 100,000 cells/ml into plasma-depleted blood. Anti-EpCAM MBs efficiently (>77%) isolated rare mouse breast 4T1, human prostate PC-3 and pancreatic cancer BxPC-3 cells spiked into 1, 3 and 7 ml (respectively) of plasma-depleted blood. Using EpCAM targeted MBs CTCs from metastatic cancer patients were isolated, suggesting that this technique could be developed into a valuable clinical tool for isolation, enumeration and analysis of rare cells.


Assuntos
Separação Imunomagnética/métodos , Microbolhas , Células Neoplásicas Circulantes , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Células Sanguíneas/imunologia , Células Sanguíneas/metabolismo , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Molécula de Adesão da Célula Epitelial , Humanos , Modelos Teóricos , Células Neoplásicas Circulantes/metabolismo , Ligação Proteica/imunologia
16.
Bioconjug Chem ; 22(8): 1638-44, 2011 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-21786821

RESUMO

Quantum dots (QDs) are attracting intense interest as fluorescence labeling agents for biomedical imaging because biocompatible coatings and relatively nontoxic rare earth metal QDs have emerged as possible options. QD photoemissions are bright, of narrow wavelength range, and very stable. We sought to encapsulate QDs within targeted PEGylated liposomes to reduce their propensity for liver uptake and to amplify the already strong QD emission signal. A novel lipid-QD conjugate initialized a process by which lipids in solution coalesced around the QDs. The liposomal structure was confirmed with size measurements, SEM, and IR spectroscopy. PEGylated QD liposomes injected into a xenograft tumor model largely cleared from the body within 24 h. Residual liver labeling was low. Targeted QD liposomes exhibited robust tumor labeling compared with controls. This study highlights the potential of these near IR emitting QD liposomes for preclinical/clinical applications.


Assuntos
Lipossomos/farmacocinética , Neoplasias Experimentais/tratamento farmacológico , Pontos Quânticos , Animais , Cápsulas , Corantes Fluorescentes , Lipossomos/administração & dosagem , Fígado/metabolismo , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/uso terapêutico , Polietilenoglicóis , Transplante Heterólogo
17.
Mol Cancer Ther ; 10(6): 972-82, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21518727

RESUMO

Although nanoparticle-based drug delivery formulations can improve the effectiveness and safety of certain anticancer drugs, many drugs, due to their chemical composition, are unsuitable for nanoparticle loading. Here, we describe a targeted nanogel drug delivery platform that can (i) encapsulate a wide range of drug chemotypes, including biological, small molecule, and cytotoxic agents; (ii) display targeting ligands and polymeric coatings on the surface; (iii) enhance drug retention within the nanogel core after photo-cross-linking; and (iv) retain therapeutic activity after lyophilization allowing for long-term storage. For therapeutic studies, we used integrin αvß3-targeted lipid-coated nanogels with cross-linked human serum albumin in the core for carrying therapeutic cargoes. These particles exhibited potent activity in tumor cell viability assays with drugs of distinct chemotype, including paclitaxel, docetaxel, bortezomib, 17-AAG, sorafenib, sunitinib, bosutinib, and dasatinib. Treatment of orthotopic breast and pancreas tumors in mice with taxane-loaded nanogels produced a 15-fold improvement in antitumor activity relative to Abraxane by blocking both primary tumor growth and spontaneous metastasis. With a modifiable surface and core, the lipid-coated nanogel represents a platform technology that can be easily adapted for specific drug delivery applications to treat a wide range of malignant diseases.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Neoplasias Pancreáticas/tratamento farmacológico , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Polietilenoimina/administração & dosagem , Polietilenoimina/química , Paclitaxel Ligado a Albumina , Albuminas/administração & dosagem , Animais , Antineoplásicos/química , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Liofilização/métodos , Humanos , Integrina alfaVbeta3/metabolismo , Lipídeos/química , Camundongos , Camundongos Nus , Nanogéis , Paclitaxel/administração & dosagem , Neoplasias Pancreáticas/metabolismo , Polietilenoglicóis/síntese química , Polietilenoimina/síntese química , Polímeros/química , Taxoides/administração & dosagem
18.
Nat Med ; 16(8): 909-14, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20676106

RESUMO

Although it is well established that tumors initiate an angiogenic switch, the molecular basis of this process remains incompletely understood. Here we show that the miRNA miR-132 acts as an angiogenic switch by targeting p120RasGAP in the endothelium and thereby inducing neovascularization. We identified miR-132 as a highly upregulated miRNA in a human embryonic stem cell model of vasculogenesis and found that miR-132 was highly expressed in the endothelium of human tumors and hemangiomas but was undetectable in normal endothelium. Ectopic expression of miR-132 in endothelial cells in vitro increased their proliferation and tube-forming capacity, whereas intraocular injection of an antagomir targeting miR-132, anti-miR-132, reduced postnatal retinal vascular development in mice. Among the top-ranking predicted targets of miR-132 was p120RasGAP, which we found to be expressed in normal but not tumor endothelium. Endothelial expression of miR-132 suppressed p120RasGAP expression and increased Ras activity, whereas a miRNA-resistant version of p120RasGAP reversed the vascular response induced by miR-132. Notably, administration of anti-miR-132 inhibited angiogenesis in wild-type mice but not in mice with an inducible deletion of Rasa1 (encoding p120RasGAP). Finally, vessel-targeted nanoparticle delivery of anti-miR-132 restored p120RasGAP expression in the tumor endothelium, suppressed angiogenesis and decreased tumor burden in an orthotopic xenograft mouse model of human breast carcinoma. We conclude that miR-132 acts as an angiogenic switch by suppressing endothelial p120RasGAP expression, leading to Ras activation and the induction of neovascularization, whereas the application of anti-miR-132 inhibits neovascularization by maintaining vessels in the resting state.


Assuntos
Endotélio Vascular/patologia , MicroRNAs/fisiologia , Neovascularização Patológica/genética , Proteína p120 Ativadora de GTPase/genética , Animais , Anticorpos Monoclonais/farmacologia , Proliferação de Células , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Endotélio Vascular/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , MicroRNAs/metabolismo , Neovascularização Patológica/metabolismo , Interferência de RNA/fisiologia , RNA Interferente Pequeno/farmacologia , Artéria Retiniana/efeitos dos fármacos , Artéria Retiniana/metabolismo , Artéria Retiniana/patologia , Regulação para Cima/genética , Regulação para Cima/fisiologia , Proteína p120 Ativadora de GTPase/metabolismo
19.
Biomaterials ; 30(12): 2369-84, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19157538

RESUMO

Toward probing an hitherto unexplored structure-activity issue namely, the relative in vitro and in vivo efficacies of cationic glycolipids with cyclic and acyclic sugar heads for targeting of genes to liver, we have designed and synthesized two novel series of cationic glycolipids with cyclic (lipids 1-5) and open d-galactose heads (lipids 6-10) containing varying spacer arm lengths in between the sugar and positively charged nitrogen atoms. Among the cyclic glycolipids, lipid 3 with six methylene units spacer in between the quaternary nitrogen atom and among the glycolipids with the open-sugar heads, lipid 6 with only two methylene units spacer were found to be the most efficacious in targeting genes to cultured HepG2 (human hepatocarcinoma cells) and primary hepatocytes. Findings in the fluorescence resonance energy transfer (FRET) studies revealed biomembrane fusibilities as important physico-chemical parameters behind the varying spacer arm dependencies in the two series. Importantly, both the serum compatible glycolipids 3 &6 were found to be equally efficacious in selectively targeting genes to mouse livers under systemic settings. The significantly reduced efficiencies of the glycolipids 3 &6 in transfecting primary hepatocytes as well as mice pretreated with asialofetuin (the ligands of asialoglycoprotein receptors) support the notion that the cellular uptake of the lipoplexes prepared from both the open and the cyclic sugar-head series is mediated via asialoglycoprotein receptor. In summary, our present findings demonstrate for the first time that cationic glycolipids with cyclic sugar-head require longer spacer arms than their acyclic sugar-head counterparts for efficient gene transfection and both the series hold equal promise for selective gene targeting to liver under systemic settings.


Assuntos
Galactose/química , Glicolipídeos/química , Fígado/metabolismo , Transgenes/genética , Animais , Cátions/química , Células Cultivadas , Fenômenos Químicos , Glicolipídeos/síntese química , Humanos , Lipossomos , Masculino , Camundongos , Estrutura Molecular , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA