Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
3.
J Control Release ; 192: 157-66, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25041999

RESUMO

Neuroblastoma (NB), the most common and deadly extracranial solid tumour of childhood, represents a challenging in paediatric oncology. Soluble tumour necrosis factor (TNF)-related apoptosis-inducing ligand (sTRAIL) is a cancer cell-specific molecule exerting remarkable anti-tumour activities against paediatric malignancies both in vitro and in preclinical settings. However, due to its too fast elimination and to the undesired related side effects, the improvement of sTRAIL in vivo bioavailability and the specific delivery to the tumour is mandatory for increasing its therapeutic efficacy. In this manuscript, we developed an innovative pegylated liposomal formulation carrying the sTRAIL at the outer surface (sTRAIL-SL) with the intent to improve its serum half-life and increase its efficacy in vivo, while reducing side effects. Furthermore, the possibility to combine sTRAIL-SL with the proteasome inhibitor Bortezomib (BTZ) was investigated, being BTZ able to sensitize tumour cells toward TRAIL-induced apoptosis. We demonstrated that sTRAIL preserved and improved its anti-tumour activity when coupled to nanocarriers. Moreover, sTRAIL-SL ameliorated its PK profile in blood allowing sTRAIL to exert a more potent anti-tumour activity, which led, upon BTZ priming, to a statistically significant enhanced life spans in two models of sTRAIL-resistant NB-bearing mice. Finally, mechanistic studies indicated that the combination of sTRAIL with BTZ sensitized sTRAIL-resistant NB tumour cells to sTRAIL-induced cell death, both in vitro and in vivo, through the Akt/GSK3/ß-catenin axis-dependent mechanism. In conclusion, our results suggest that sTRAIL-SL might be an efficient vehicle for sTRAIL delivery and that its use in clinic, in combination with BTZ, might represent an adjuvant strategy for the treatment of stage IV, sTRAIL-resistant, NB patients.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Ácidos Borônicos/administração & dosagem , Ácidos Borônicos/uso terapêutico , Neuroblastoma/tratamento farmacológico , Pirazinas/administração & dosagem , Pirazinas/uso terapêutico , Ligante Indutor de Apoptose Relacionado a TNF/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Animais , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptose/efeitos dos fármacos , Bortezomib , Linhagem Celular Tumoral , Feminino , Humanos , Lipossomos , Camundongos , Camundongos Nus , Neuroblastoma/patologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacocinética
4.
Curr Drug Targets ; 10(10): 1021-7, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19663770

RESUMO

Novel anti-vasculature strategies that are emerging for the treatment of cancer and for the inhibition of angiogenesis may be a promising new tool for the adjuvant therapy of malignant tumours. Over the last fifteen years, several reports have been published concerning the relationship between tumour progression and angiogenesis in experimental models of neuroblastoma in vitro and in vivo. Moreover, a high vascular index in neuroblastoma correlates with poor prognosis, suggesting dependence of aggressive tumour growth on active angiogenesis. Here, we present an overview of the most recent advances in anti-vasculature therapy of neuroblastoma, and describe some preclinical results as well as future perspectives.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Neuroblastoma/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Progressão da Doença , Sistemas de Liberação de Medicamentos , Humanos , Neovascularização Patológica/tratamento farmacológico , Neuroblastoma/irrigação sanguínea , Neuroblastoma/patologia , Prognóstico
5.
Ann Ig ; 18(6): 491-505, 2006.
Artigo em Italiano | MEDLINE | ID: mdl-17228607

RESUMO

Aim of the study is to describe the application of surgical site infection (SSI) control procedures in general surgery operating rooms of Piedmont region hospitals. A specific data collection form was designed to record information. 54 questionnaires were compiled. Piedmont legislation related to operating rooms' equipment is obeyed in more than 90% of hospitals. Nevertheless, there are some critical aspects than could be risk factors for SSI or that are not useful in order to prevent them: use of UV radiation (11.3%), use of tacky mats at the entrance of the operating room (5.7%), special cleaning of operating rooms after contaminated or dirty operations (73.6%) and routine environmental sampling (10%) that is strongly recommended by ISPESL guideline in disagreement with international recommendations. Steam autoclave is used for surgical instruments sterilization by 100% of hospitals, but only 50% of them performs an annual validation of both autoclave performance and sterilization procedures. The study gave useful information in order to promote some structural modifications and personnel education for efficacious SSI prevention and control.


Assuntos
Ambiente Controlado , Controle de Infecções/legislação & jurisprudência , Legislação Hospitalar , Salas Cirúrgicas/normas , Guias de Prática Clínica como Assunto , Infecção da Ferida Cirúrgica/prevenção & controle , Infecção Hospitalar/prevenção & controle , Humanos , Agências Internacionais , Itália , Salas Cirúrgicas/legislação & jurisprudência , Fatores de Risco , Inquéritos e Questionários
6.
Cancer Lett ; 197(1-2): 151-5, 2003 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-12880975

RESUMO

Neuroblastoma (NB) is the most common extracranial solid tumor of childhood. In advanced disease stages, prognosis is poor and treatments have limited efficacy, thus novel strategies are warranted. The synthetic retinoid fenretinide (HPR) induces apoptosis in NB and melanoma cell lines. We reported an in vitro potentiation of HPR effects on melanoma cells when the drug is incorporated into GD2-targeted immunoliposomes (anti-GD2-SIL-HPR). Here, we investigated the antitumor activity of anti-GD2-SIL-HPR against NB cells, both in vitro and in vivo. Anti-GD2-immunoliposomes (anti-GD2-SIL) showed specific, competitive binding to, and uptake by, various NB cell lines. Moreover, anti-GD2-SIL-HPR presented increased selectivity and efficacy in inhibiting NB cell proliferation through the induction of apoptosis, compared to free drug and SL-HPR. In an in vivo NB metastatic model, we demonstrated that anti-GD2-SIL-HPR completely inhibited the development of macroscopic and microscopic metastases in comparison to controls. However, similar, but significantly less potent antitumor effect was observed also in mice treated with anti-GD2 immunoliposomes without HPR (anti-GD2-SIL-blank) or anti-GD2 mAb alone (P=0.0297 and P=0.0294, respectively, vs. anti-GD2-SIL-HPR). Moreover, our results clearly demonstrated that, although anti-GD2 mAb had a strong antitumor effect in this in vivo NB model, 100% curability was obtained only following treatment with anti-GD2-SIL-HPR (P<0.0001). Anti-GD2 liposomal HPR should receive clinical evaluation as adjuvant therapy of neuroblastoma.


Assuntos
Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Fenretinida/administração & dosagem , Gangliosídeos/imunologia , Neuroblastoma/tratamento farmacológico , Animais , Divisão Celular , Modelos Animais de Doenças , Humanos , Lipossomos , Camundongos , Neuroblastoma/patologia , Células Tumorais Cultivadas
7.
Cancer Lett ; 197(1-2): 205-9, 2003 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-12880983

RESUMO

In spite of the satisfactory frequency of clinical response to first-line therapy in neuroblastoma (NB), complete eradication of NB cells is rarely achieved. As a consequence, the majority of patients with advanced stage NB undergo relapse, which is often resistant to conventional treatment and rapidly overwhelming. Thus, after induction of the apparent remission, new therapeutic strategies are needed to completely eradicate the small number of surviving NB cells and to prevent relapse. We explored the potential of different doses of the anti-GD2 monoclonal antibody (mAb) 14G2a in an experimental metastatic model where a limited number of HTLA-230 human NB cells are injected i.v. into nude mice, leading to extensive metastases and death of animals within 7-8 weeks. Treatment with 14G2a mAb (1-4 mg/kg cumulative dose given as five i.v. daily administrations) dramatically reduced the metastatic spread of NB cells and prolonged the long-term survival of treated mice in a dose-dependent manner. Neither macrophages nor NK cells appeared to contribute to the protective effect of antibody treatment in vivo, suggesting either an involvement of granulocytes or a complement-mediated cytotoxicity towards NB cells. Whatever the effecting mechanism(s) involved, these results strongly support the clinical use of anti-GD2 mAbs after first-line induction regimens.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Gangliosídeos/imunologia , Imunoterapia , Recidiva Local de Neoplasia/terapia , Neuroblastoma/terapia , Animais , Humanos , Camundongos , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/prevenção & controle , Neuroblastoma/prevenção & controle , Taxa de Sobrevida , Células Tumorais Cultivadas
8.
J Control Release ; 74(1-3): 69-75, 2001 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-11489484

RESUMO

Formulations of antisense oligonucleotides (asODNs) against c-myb or c-myc protooncogenes have been prepared by a new technique that sequesters cationic lipid in the interior of a lipid particle. This technique results in high loading efficiency for the asODNs, small particle size and good stability. When targeted against melanoma cells or neuroblastoma cells via anti-GD(2) coupled at the particle surface, increased cell binding to the cells could be demonstrated. Targeted formulations showed greater inhibition of cell proliferation compared to non-targeted formulations or free drug. Inhibition of cell proliferation was demonstrated to be due to down-regulation of c-myb or c-myc protein expression. The formulations have long-circulation times in vivo, and evaluation for in vivo antitumor activity is currently underway.


Assuntos
Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/uso terapêutico , Western Blotting , Divisão Celular/efeitos dos fármacos , Composição de Medicamentos , Humanos , Lipossomos , Micelas , Neoplasias/metabolismo , Oligonucleotídeos Antissenso/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Células Tumorais Cultivadas
9.
J Natl Cancer Inst ; 92(3): 253-61, 2000 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-10655443

RESUMO

BACKGROUND: Advanced-stage neuroblastoma resists conventional treatment; hence, novel therapeutic approaches are required. We evaluated the use of c-myb antisense oligodeoxynucleotides (asODNs) delivered to cells via targeted immunoliposomes to inhibit c-Myb protein expression and neuroblastoma cell proliferation in vitro. METHODS: Phosphorothioate asODNs and control sequences were encapsulated in cationic lipid, and the resulting particles were coated with neutral lipids to produce coated cationic liposomes (CCLs). Monoclonal antibodies directed against the disialoganglioside GD(2) were covalently coupled to the CCLs. (3)H-labeled liposomes were used to measure cellular binding, and cellular uptake of asODNs was evaluated by dot-blot analysis. Growth inhibition was quantified by counting trypan blue dye-stained cells. Expression of c-Myb protein was examined by western blot analysis. RESULTS: Our methods produced GD(2)-targeted liposomes that stably entrapped 80%-90% of added c-myb asODNs. These liposomes showed concentration-dependent binding to GD(2)-positive neuroblastoma cells that could be blocked by soluble anti-GD(2) monoclonal antibodies. GD(2)-targeted liposomes increased the uptake of asODNs by neuroblastoma cells by a factor of fourfold to 10-fold over that obtained with free asODNs. Neuroblastoma cell proliferation was inhibited to a greater extent by GD(2)-targeted liposomes containing c-myb asODNs than by nontargeted liposomes or free asODNs. GD(2)-targeted liposomes containing c-myb asODNs specifically reduced expression of c-Myb protein by neuroblastoma cells. Enhanced liposome binding and asODN uptake, as well as the antiproliferative effect, were not evident in GD(2)-negative cells. CONCLUSIONS: Encapsulation of asODNs into immunoliposomes appears to enhance their toxicity toward targeted cells while shielding nontargeted cells from antisense effects and may be efficacious for the delivery of drugs with broad therapeutic applications to tumor cells.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Gangliosídeos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Oligodesoxirribonucleotídeos Antissenso/administração & dosagem , Proteínas Proto-Oncogênicas c-myb/antagonistas & inibidores , Western Blotting , Humanos , Lipossomos , Oligodesoxirribonucleotídeos Antissenso/genética , Proteínas Proto-Oncogênicas c-myb/genética , Células Tumorais Cultivadas
10.
Int J Cancer ; 81(2): 262-7, 1999 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-10188729

RESUMO

Melanoma is a highly malignant and increasingly common tumour. Since metastatic melanoma remains incurable, new treatment approaches are needed. Previously, we reported that the synthetic retinoid N-(4-hydroxyphenyl)retinamide (fenretinide, HPR) induces apoptosis in neuroblastoma cells, sharing a neuroectodermal origin with melanoma cells. Since no data exist thus far on the effects of HPR on human melanoma tumours, our purpose was to investigate the in vitro modulation of cell growth and apoptosis by HPR in melanoma cells. Ten human melanoma cell lines were exposed in vitro to increasing concentrations of HPR. Dose-dependent growth inhibition and cytotoxicity were observed. According to cytofluorimetric analysis, propidium iodide staining and TUNEL assay, HPR-treated melanoma cells were shown to undergo apoptosis. However, IC50 values ranged from 5 to 28 microM, while IC90 values were between 10 and 45 microM. These last concentrations are approximately 10-fold higher than those achievable in patients given oral HPR. To explore the potential of new delivery strategies, HPR was loaded at high concentrations into immunoliposomes directed to disialoganglioside GD2, a tumour-specific antigen extensively expressed by neuroectoderma-derived tumours. Treatment of melanoma cells for a short time (2 hr) with HPR-containing immunoliposomes followed by culture in drug-free medium gave rise to apoptosis of target cells, whereas cells treated for 2 hr with equivalent concentrations of the free drug survived. The efficacy of immunoliposomal HPR was strongly dependent on the density of GD2 expression in the different cell lines.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Fenretinida/uso terapêutico , Melanoma/tratamento farmacológico , Especificidade de Anticorpos , Divisão Celular/efeitos dos fármacos , Humanos , Lipossomos , Melanoma/patologia , Células Tumorais Cultivadas
11.
Int J Cancer ; 81(2): 268-74, 1999 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-10188730

RESUMO

Melanoma is a highly malignant and increasingly common neoplasm. Because metastatic melanoma remains incurable, new treatment approaches are needed. Immunoliposomes have been previously shown to enhance the selective localization of immunoliposome-entrapped drugs to solid tumors with improvements in the therapeutic index of the drugs. Previously, we reported that the synthetic retinoid fenretinide (HPR) is an inducer of apoptosis in neuroblastoma (NB) cells, sharing the neuroectodermal origin with melanoma cells. HPR is a strong inducer of apoptosis also in melanoma cells, although at doses 10-fold higher than those achievable clinically. Thus, our purpose was to investigate the in vitro potentiation of its cytotoxic effect on melanoma cells in combination with long-circulating GD2-targeted immunoliposomes. GD2 is a disialoganglioside extensively expressed on tumors of neuroectodermal origin, including melanoma. Murine anti-GD2 antibody (Ab) 14.G2a and its human/mouse chimeric variant ch14.18 have been ligated to sterically stabilized liposomes by covalent coupling of Ab to the polyethylene glycol (PEG) terminus. Ab-bearing liposomes showed specific, competitive binding to and uptake by various melanoma cell lines compared with liposomes bearing non-specific isotype-matched Abs or Ab-free liposomes. Cytotoxicity was evaluated after 2 hr treatment, followed by extensive washing and 72 hr incubation. This treatment protocol was designed to minimize non-specific adsorption of liposomes to the cells, while allowing for maximum Ab-mediated binding. When melanoma cells were incubated with 30 microM HPR entrapped in anti-GD2 liposomes, a significant reduction in cellular growth was observed compared to free HPR, entrapped HPR in Ab-free liposomes or empty liposomes. Cytotoxicity was not evident in tumor cell lines of other origins that did not express GD2. Growth of NB cells was also inhibited by immunoliposomes with entrapped HPR.


Assuntos
Antineoplásicos/uso terapêutico , Fenretinida/uso terapêutico , Gangliosídeos/uso terapêutico , Melanoma/tratamento farmacológico , Especificidade de Anticorpos , Divisão Celular/efeitos dos fármacos , Portadores de Fármacos , Humanos , Lipossomos , Melanoma/patologia , Sensibilidade e Especificidade , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...