Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 20(1)2018 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-30587795

RESUMO

Triple negative breast cancer (TNBC) remains a serious health problem with poor prognosis and limited therapeutic options. To discover novel approaches to treat TNBC, we screened cholera toxin (CT) and the members of the bacterial type II heat-labile enterotoxin family (LT-IIa, LT-IIb, and LT-IIc) for cytotoxicity in TNBC cells. Only LT-IIc significantly reduced viability of the TNBC cell lines BT549 and MDA-MB-231 (IC50 = 82.32 nM). LT-IIc had no significant cytotoxic effect on MCF10A (IC50 = 2600 nM), a non-tumorigenic breast epithelial cell line, and minimal effects on MCF7 and T47D, ER⁺ cells, or SKBR-3 cells, HER2⁺ cells. LT-IIc stimulated autophagy through inhibition of the mTOR pathway, while simultaneously inhibiting autophagic progression, as seen by accumulation of LC3B-II and p62. Morphologically, LT-IIc induced the formation of enlarged LAMP2+ autolysosomes, which was blocked by co-treatment with bafilomycin A1. LT-IIc induced apoptosis as demonstrated by the increase in caspase 3/7 activity and Annexin V staining. Co-treatment with necrostatin-1, however, demonstrated that the lethal response of LT-IIc is elicited, in part, by concomitant induction of necroptosis. Knockdown of ATG-5 failed to rescue LT-IIc-induced cytotoxicity, suggesting LT-IIc can exert its cytotoxic effects downstream or independently of autophagophore initiation. Collectively, these experiments demonstrate that LT-IIc acts bifunctionally, inducing autophagy, while simultaneously blocking autolysosomal progression in TNBC cells, inducing a specific cytotoxicity in this breast cancer subtype.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Enterotoxinas/toxicidade , Proteína 5 Relacionada à Autofagia/antagonistas & inibidores , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Temperatura Alta , Humanos , Imidazóis/metabolismo , Indóis/metabolismo , Lisossomos/metabolismo , Necrose , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
2.
Ann Pharmacother ; 45(11): 1399-405, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22028422

RESUMO

OBJECTIVE: To summarize the preclinical and clinical data on vemurafenib, approved by the Food and Drug Administration (FDA) on August 17, 2011, and discuss the drug's clinical advantages and limitations. DATA SOURCES: An English-language literature search of MEDLINE/PubMed (1966-August 2011), using the terms PLX4032, RG7204, RO5185426, vemurafenib, and metastatic melanoma, was conducted. In addition, information and data were obtained from meeting abstracts, clinical trial registries, and news releases from the FDA. STUDY SELECTION AND DATA EXTRACTION: All relevant published articles and abstracts on vemurafenib were included. Clinical trial registries and meeting abstracts were used to obtain information regarding ongoing trials. All peer-reviewed articles containing information regarding the clinical safety and efficacy of vemurafenib were evaluated for inclusion. DATA SYNTHESIS: Before the recent approval (March 2011) of ipilimumab, there was no first-line standard-of-care therapy, with proven overall survival benefit, for the treatment of malignant metastatic melanoma. Unlike ipilimumab, which acts through immune-modulation, vemurafenib is a novel, molecularly targeted therapeutic with preferential efficacy toward a specific mutated oncogenic BRAF-signaling mediator. In recently published results of a Phase 3 clinical trial comparing dacarbazine with vemurafenib, vemurafenib prolonged progression-free and overall survival, with confirmed objective response rate of 48% (95% CI 42 to 55) versus 5% (95% CI 3 to 9) for patients who received dacarbazine (p < 0.001). CONCLUSIONS: Vemurafenib offers a novel, first-line, personalized therapy for patients who have mutated BRAF. Clinical trials of vemurafenib in combination with ipilimumab or other targeted or cytotoxic chemotherapeutic agents may provide more effective regimens with long-term clinical benefit.


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Indóis/efeitos adversos , Indóis/uso terapêutico , Melanoma/tratamento farmacológico , Melanoma/enzimologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Sulfonamidas/efeitos adversos , Sulfonamidas/uso terapêutico , Ensaios Clínicos como Assunto , Intervalo Livre de Doença , Avaliação Pré-Clínica de Medicamentos , Humanos , Melanoma/genética , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Ensaios Clínicos Controlados Aleatórios como Assunto , Vemurafenib
3.
Cancer Biol Ther ; 12(7): 574-85, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21795855

RESUMO

Poor prognosis cancers, such as pancreatic cancer, represent inherent challenges for ceramide-based nanotherapeutics due to metabolic pathways, which neutralize ceramide to less toxic or pro-oncogenic metabolites. We have recently developed a novel 80 nanometer diameter liposomal formulation that incorporates 30 molar percent C6-ceramide, a bioactive lipid that is pro-apoptotic to many cancer cells, but not to normal cells. In this manuscript, we evaluated the efficacy of combining nanoliposomal C6-ceramide (Lip-C6) with either gemcitabine or an inhibitor of glucosylceramide synthase. We first assessed the biological effect of Lip-C6 in PANC-1 cells, a gemcitabine-resistant human pancreatic cancer cell line, and found that low doses alone did not induce cell toxicity. However, cytotoxicity was achieved by combining Lip-C6 with either non-toxic sub-therapeutic concentrations of gemcitabine or with the glucosylceramide synthase inhibitor D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP). Furthermore, these combinations with Lip-C6 cooperatively inhibited PANC-1 tumor growth in vivo. Mechanistically, Lip-C6 inhibited pro-survival Akt and Erk signaling, whereas the nucleoside analog gemcitabine did not. Furthermore, by including PDMP within the nanoliposomes, which halted ceramide neutralization as evidenced by LC-MS3, the cytotoxic effects of Lip-C6 were enhanced. Collectively, we have demonstrated that nanoliposomal ceramide can be an effective anti-pancreatic cancer therapeutic in combination with gemcitabine or an inhibitor of ceramide neutralization.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Ceramidas/farmacologia , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Linhagem Celular Tumoral , Ceramidas/administração & dosagem , Ceramidas/farmacocinética , Desoxicitidina/administração & dosagem , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Portadores de Fármacos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Feminino , Glucosiltransferases/antagonistas & inibidores , Humanos , Lipossomos/uso terapêutico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Nus , Morfolinas/administração & dosagem , Morfolinas/farmacologia , Nanopartículas/administração & dosagem , Nanopartículas/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
4.
Cancer Biol Ther ; 12(5): 427-35, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21725197

RESUMO

Neurotensin receptor-1 (NTSR-1) is a G-protein coupled receptor (GPCR) that has been recently identified as a mediator of cancer progression. NTSR-1 and its endogenous ligand, neurotensin (NTS), are co-expressed in several breast cancer cell lines and breast cancer tumor samples. Based on our previously published study demonstrating that intact structured membrane microdomains (SMDs) are required for NTSR-1 mitogenic signaling, we hypothesized that regulated receptor palmitoylation is responsible for NTSR-1 localization and signaling within SMDs upon NTS stimulation. Site-directed mutagenesis and pharmacological strategies were utilized to assess NTRS-1 post-translational modifications in an over-expression cell model (HEK293T) as well as a native breast cancer cell model (MDA-MB-231). NTSR-1 palmitoylation was confirmed by multiple chemical and fluororadiographic methodologies. NTSR-1 glycosylation was confirmed by pharmacological (tunicamycin) and chemical (PGNaseF and O-type glycosidase) approaches. Physiological correlates including cell viability (MTS assay), apoptosis (caspase 3/7 assay) and ERK phosphorylation were utilized to assess the consequences of NTRS-1 palmitoylation. The interaction between palmitoylated NTRS-1 and Gαq/11 within SMDS was confirmed with immunopreciptation analysis of detergent-free isolated fractions of caveolin-rich microdomains. We identified dual-palmitoylation at Cys381 and Cys383 of endogenously-expressed NTSR-1 in MDA-MB-231 breast adeno-carcinomas as well as exogenously-expressed NTSR-1 in HEK293T cells (which do not normally express NTSR-1). Pharmacological inhibition of NTSR-1 palmitoylation in MDA-MB-231 cells as well as NTSR-1-expressing HEK293T cells diminished NTS-mediated ERK 1/2 phosphorylation. Additionally, NTSR-1 mutated at Cys381 and Cys383 showed diminished ERK1/2 stimulation and reduced ability to protect HEK293T cells against apoptosis induced by serum starvation. Mechanistically, mutated C381,383S-NTSR-1 showed reduced ability to interact with Gαq/11 and diminished localization to structured membrane microdomains (SMDs), where Gαq/11 preferentially resides. We also demonstrated that only glycosylated isoforms of NTRS-1 localize within SMDs by palmitotylation. Collectively, our data establish palmitoylation as a novel pharmacological target to inhibit NTSR-1 mitogenic signaling in breast cancer cells.


Assuntos
Membrana Celular/efeitos dos fármacos , Lipoilação/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases , Neurotensina/farmacologia , Receptores de Neurotensina/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias da Mama , Linhagem Celular , Membrana Celular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Glicosilação/efeitos dos fármacos , Humanos , Mutagênese Sítio-Dirigida , Neurotensina/biossíntese , Palmitatos/farmacologia , Fosforilação , Receptores Mitogênicos/metabolismo , Receptores de Neurotensina/genética , Tunicamicina/farmacologia
5.
Mol Cancer Res ; 7(5): 724-34, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19435815

RESUMO

Neurotensin (NTS) receptor 1 (NTSR1) is a G protein-coupled receptor that has been recently identified as a mediator of tumorigenicity and metastasis. NTSR1, as well as its endogenous ligand, NTS, are coexpressed in several breast cancer cell lines and breast cancer tumor samples but not in normal breast tissue. We have previously published that ceramide mimetics could inhibit breast cancer growth in vitro and in vivo. Thus, understanding the biochemical and biophysical regulation of NTSR1 by ceramide can help further define NTSR1 as a novel target in breast cancer. Our results show that nanoliposomal formulations of ceramide inhibit NTSR1-mediated MDA-MB-231 breast cancer progression (mitogenesis, migration, and matrix metalloproteinase-9 activity). In addition, liposomal ceramide inhibited NTSR1-mediated, but not phorbol 12-myristate 13-acetate-mediated, activation of the mitogen-activated protein kinase pathway. Mechanistically, nanoliposomal short-chain ceramide reduces NTSR1 interaction with Galphaq/11 subunits within structured membrane microdomains, consistent with diminished NTS-induced translocation of NTSR1 into membrane microdomains. Collectively, our findings suggest that exogenous short-chain ceramide has the potential to be used as an adjuvant therapy to inhibit NTS-dependent breast cancer progression.


Assuntos
Ceramidas/farmacologia , Microdomínios da Membrana/metabolismo , Neurotensina/metabolismo , Receptores de Neurotensina/metabolismo , Western Blotting , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ceramidas/química , Ativação Enzimática/efeitos dos fármacos , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Humanos , Imunoprecipitação , Lipossomos/química , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Nanotecnologia , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia
6.
Drug Metab Dispos ; 36(8): 1709-15, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18490436

RESUMO

Ceramide, an endogenous sphingolipid, has demonstrated antieoplastic activity in vitro and in vivo. However, the chemotherapeutic utility of ceramide is limited because of its insolubility. To increase the solubility of ceramide, liposomal delivery systems have been used. The objective of the present study was to characterize the pharmacokinetics and tissue distribution of C6-ceramide and control (non-C6-ceramide) nanoliposomes in rats, using [14C]C6-ceramide and [3H]distearylphosphatidylcholine (DSPC) as tracers of the ceramide and liposome components, respectively. Ceramide liposomes were administered at 50 mg of liposomes/kg by jugular vein to female Sprague-Dawley rats. The apparent volume of distribution (Vd) of [3H]DSPC was approximately 50 ml/kg, suggesting that the liposomes were confined to the systemic circulation. In contrast, the Vd of [14C]C6-ceramide was 20-fold greater than that of liposomes, indicating extensive tissue distribution. This high Vd of [14C]C6-ceramide in relation to that of [3H]DSPC suggests that ceramide and liposomes distribute independently of each other. This disparate disposition was confirmed by tissue distribution studies, in which [14C]C6-ceramide exhibited rapid tissue accumulation compared with to [3H]DSPC. Examination of ceramide liposome blood compartmentalization in vitro also demonstrated divergent partitioning, with liposomes being confined to the plasma fraction and ceramide rapidly equilibrating between red blood cell and plasma fractions. A bilayer exchange mechanism for ceramide transfer is proposed to explain the results of the present study, as well as give insight into the documented antineoplastic efficacy of short-chain ceramide liposomes. Our studies suggest that this nanoscale PEGylated drug delivery system for short-chain ceramide offers rapid tissue distribution without adverse effects for a neoplastic-selective, insoluble agent.


Assuntos
Ceramidas/farmacocinética , Lipossomos , Animais , Ceramidas/sangue , Eritrócitos/metabolismo , Feminino , Técnicas In Vitro , Tamanho da Partícula , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
7.
Nano Lett ; 8(12): 4116-21, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19367878

RESUMO

Paradigm-shifting modalities to more efficiently deliver drugs to cancerous lesions require the following attributes: nanoscale-size, targetability, and stability under physiological conditions. Often, these nanoscale drug delivery vehicles are limited due to agglomeration, poor solubility, or cytotoxicity. Thus, we have designed a methodology to encapsulate hydrophobic antineoplastic chemotherapeutics within a 20-30 nm diameter, pH-responsive, nonagglomerating, nontoxic calcium phosphate nanoparticle matrix. In the present study, we report on calcium phosphate nanocomposite particles (CPNPs) that encapsulate both fluorophores and chemotherapeutics, are colloidally stable in physiological solution for an extended time at 37 degrees C and can efficaciously deliver hydrophobic antineoplastic agents, such as ceramide, in several cell model systems.


Assuntos
Antineoplásicos/uso terapêutico , Fosfatos de Cálcio/química , Portadores de Fármacos , Nanocompostos , Neoplasias/tratamento farmacológico , Antineoplásicos/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...