Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Drug Deliv ; 14(4): 209-17, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17497353

RESUMO

Oleyl alcohol was complexed with new amphiphilic polyvinylalcohol derivatives with the aim of increasing its aqueous solubility, thus improving bioavailability and favoring its antitumor activity. Water-soluble amphiphilic polymers were prepared by polyvinyl alcohol (PVA) substitution with oleyl chains through a succinyl spacer at 2% and 3% substitution degree. The complexes were obtained by spray-drying hydroalcoholic solutions of the substituted polymers and free oleyl alcohol at different weight ratios (3:1; 5:1; 10:1 w/w). The main physicochemical characteristics of the complexes were analyzed and correlated to the cytotoxic activity of oleyl alcohol toward tumor cell lines. The complexes strongly increased the aqueous solubility of oleyl alcohol and provided oleyl alcohol release in the presence of extractive conditions (simulating in vivo absorption). The complexes obtained by 10:1 polymer:fatty alcohol weight ratio offered higher release rates than the 5:1 and 3:1 ratios, respectively. Complexation also increased oleyl alcohol cytotoxicity toward tumor cells due to increased availability of the active molecule in the aqueous phase. Pure polymers were found to be biocompatible and no toxic effect was detected up to the highest concentration used in the present study (500 mu g/ml). The complexation of oleyl alcohol with the polymers analyzed here efficiently increased the availability of the fatty alcohol in aqueous environment. The enhanced cytotoxicity toward tumor cells of the complexed oleyl alcohol and the polymer biocompatibility make these amphiphilic PVA derivatives interesting candidates for soluble pharmaceutical formulations containing hydrophobic drugs whose therapeutic potential is often underestimated due to unsuitable levels of their aqueous solubilization.


Assuntos
Antineoplásicos/farmacologia , Álcoois Graxos/farmacologia , Álcool de Polivinil/química , Antineoplásicos/química , Varredura Diferencial de Calorimetria , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Química Farmacêutica , Relação Dose-Resposta a Droga , Álcoois Graxos/química , Humanos , Luz , Microscopia Eletrônica de Varredura , Soluções Farmacêuticas , Espalhamento de Radiação , Solubilidade , Solventes/química , Tensoativos/química , Fatores de Tempo , Água/química
2.
J Natl Cancer Inst ; 86(22): 1694-701, 1994 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-7966397

RESUMO

BACKGROUND: Interferon gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF) synergize in inducing human neuroblastoma cells to differentiate terminally in vitro into mature nonproliferating neurons. The mechanisms by which this synergistic activity takes place are still obscure. PURPOSE: To understand the basis of IFN-gamma-TNF synergism, we investigated the constitutive equipment of receptors to IFN-gamma and TNF in two human neuroblastoma cell lines (i.e., LAN-5 and GI-LI-N) and their quantitative and functional variations following treatment with IFN-gamma or TNF. METHODS: IFN-gamma receptors and TNF receptors were assessed and functionally characterized by radioreceptor-binding assay before and after treatment of the cells with IFN-gamma or TNF. The TNF receptor subtypes were identified by the reverse transcriptase-polymerase chain reaction, chemical cross-linking of receptors to iodinated TNF, and inhibition of TNF binding by type-specific anti-TNF receptor monoclonal antibodies. The effects of cytokines on cell differentiation were assessed by thymidine incorporation inhibition and morphologic maturation. RESULTS: No quantitative or functional modification of IFN-gamma receptors was observed in TNF-treated cells. However, after treatment with IFN-gamma, TNF receptor numbers were enhanced to a different extent in both cell lines. The two neuroblastoma cell lines expressed, both constitutively and after IFN-gamma induction, only one species of TNF receptor, i.e., the p80 form in LAN-5 and the p60 form in GI-LI-N. Sequential treatment with IFN-gamma followed by TNF, but not in the opposite order, could reproduce the early effects of differentiation in neuroblastoma cells, supporting a role for TNF receptor up-regulation as a basis for the cooperation between the two cytokines. CONCLUSION: The results strongly suggest that receptor regulation can be at least one mechanism by which IFN-gamma and TNF exert their synergistic effects. Moreover, it appears that the two TNF receptor types are redundant in signaling neuroblastoma cell differentiation. IMPLICATIONS: Our findings can provide a guideline for a rational design of experimental differentiation-based therapeutic protocols in patients with neuroblastoma.


Assuntos
Interferon gama/farmacologia , Neuroblastoma/metabolismo , Receptores de Interferon/efeitos dos fármacos , Receptores do Fator de Necrose Tumoral/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Sequência de Bases , Sinergismo Farmacológico , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Proteínas Recombinantes/farmacologia , Células Tumorais Cultivadas
3.
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
4.
Cancer Res ; 51(16): 4342-6, 1991 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-1868458

RESUMO

The modalities of uptake and storage of iodine-labeled m-iodobenzylguanidine (MIBG) by four human neuroblastoma cell lines have been studied. SK-N-BE(2)C cell line has been shown to possess the specific (type 1) MIBG uptake, as well as an efficient extravesicular storage mechanism. Conversely, LAN-5 cells, which show a nonsaturation kinetic of MIBG incorporation, lack only the ability to efficiently store the MIBG taken up by a mechanism that can be pharmacologically defined as uptake 1. The two other neuroblastoma cell lines tested, GI-LI-N and GI-CA-N, lack both uptake and storage capacity. In view of the fact that the only detailed study on specific MIBG uptake by a human neuroblastoma cell line has been carried out on SK-N-SH, a highly heterogeneous cell line, our report provides new insights on the molecular and cellular pharmacology of radiolabeled MIBG.


Assuntos
Antineoplásicos/metabolismo , Iodobenzenos/metabolismo , 3-Iodobenzilguanidina , Transporte Biológico , Linhagem Celular , Metabolismo Energético , Humanos , Cinética , Neuroblastoma/metabolismo , Ouabaína/farmacologia , Sódio/metabolismo , Fatores de Tempo
5.
Cancer Res ; 52(4): 931-9, 1992 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-1371090

RESUMO

Neuroblastoma (NB), a tumor originating from the sympathetic nervous system, is the most common extracranial neurological tumor of childhood. Human NB cells may differentiate in vitro under treatment with biological agents, as gamma-interferon (IFN-gamma) and tumor necrosis factor (TNF). Unfortunately, NB cell lines resistant to the differentiation-inducing effects of both drugs have been observed. Here we demonstrate that a combination of IFN-gamma plus TNF causes extensive and generalized differentiation of NB cells toward a neuronal phenotype. Both IFN-gamma and TNF, given alone, moderately reduced cell growth and induced partial morphological maturation. Their combination further reduced cell proliferation. The combined treatment gave a synergistic rather than additive cytostatic effect, documented also by a dramatically enhanced differentiation toward a neuronal morphology. Membrane immunofluorescence showed a homologous and heterologous up-regulation of IFN-gamma receptor, as well as a marked induction of HLA Class I antigens and, to a lesser extent, of Class II antigens on NB cells induced to differentiate. Treatment of NB cell lines with IFN-gamma/TNF results in the induction of a differentiated phenotype, as indicated by the increased expression of the Mr 160,000 and 200,000 neurofilament proteins and that of microtubule-associated proteins. Evaluation of biochemical markers of neuronal differentiation confirmed the ability of the combined treatment to induce neuroblast maturation. These results suggest that the combination of IFN-gamma and TNF should be considered for experimental clinical trials in neuroblastoma.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Interferon gama/farmacologia , Neuroblastoma/patologia , Fator de Necrose Tumoral alfa/farmacologia , Acetilcolinesterase/metabolismo , Antígenos de Superfície/análise , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/metabolismo , Replicação do DNA/efeitos dos fármacos , Di-Hidroxifenilalanina/metabolismo , Dopamina/metabolismo , Sinergismo Farmacológico , Imunofluorescência , Humanos , Ácido Hidroxi-Indolacético/metabolismo , Cinética , Neuroblastoma/metabolismo , Norepinefrina/metabolismo , Fosfopiruvato Hidratase/análise , Proteínas Recombinantes/farmacologia , Serotonina/metabolismo , Timidina/metabolismo , Células Tumorais Cultivadas
6.
Cancer Res ; 52(18): 4960-4, 1992 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-1325288

RESUMO

Iodine-labeled m-iodobenzylguanidine (MIBG) is a widely used radiopharmaceutical for both diagnosis and biologically targeted radiotherapy of neuroblastoma. However, resistance to the radiotherapeutic effects of MIBG is often encountered, mainly due to lack of MIBG accumulation by neoplastic cells. We have investigated whether the induction of neuroblastoma cell differentiation modifies MIBG incorporation and retention. LAN-5 cells were selected, due to their moderate ability to take up MIBG. Treatment of these cells with gamma-interferon (IFN-gamma) resulted in morphological changes accompanied by a significant increase in overall cell-associated MIBG. Desimipramine, but not reserpine, easily depleted IFN-gamma-treated LAN-5 cells of their MIBG content. This suggests that the mechanism involved is an uptake enhancement rather than an improved storage ability. Indeed, IFN-gamma induces de nov synthesis of MIBG receptor-transporters, as demonstrated by polymerase chain reaction amplification and semiquantitative analysis. Our results suggest that pretreating neuroblastoma patients with IFN-gamma before MIBG administration may enhance the efficacy of both biologically targeted radioimaging and therapy of this tumor.


Assuntos
Interferon gama/farmacologia , Iodobenzenos/metabolismo , 3-Iodobenzilguanidina , Sequência de Bases , Transporte Biológico , Diferenciação Celular , Expressão Gênica , Humanos , Técnicas In Vitro , Iodobenzenos/química , Dados de Sequência Molecular , Neuroblastoma , Oligodesoxirribonucleotídeos/química , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Receptores de Superfície Celular/metabolismo , Células Tumorais Cultivadas
7.
Cancer Res ; 55(4): 853-61, 1995 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-7850799

RESUMO

Retinoids exert various important biological effects in the control of normal growth, differentiation, and fetal development. While retinoic acid (RA) has entered clinical trials as a differentiation-promoting agent, it is only recently that the synthetic retinoid N-(4-hydroxyphenyl)retinamide (HPR) has been shown to be of potential clinical interest in cancer chemoprevention and treatment. Since thus far no data exist on the effects of HPR on neural crest cell-derived tumors, we have examined its in vitro effects on neuroblastoma (NB) cell lines and found that at relevant pharmacological concentrations it induces a dose-dependent growth inhibition. The antiproliferative effects of HPR were, in six of six cell lines tested, drastically more potent that those induced by an equimolar dose of RA. Time course growth analysis showed that HPR at 3 x 10(-6) M induces a very rapid (24-72 h) fall in thymidine uptake (> 90%), whereas at 3 x 10(-7) M it exhibits cytostatic effects. In contrast to RA, HPR did not show morphological changes typical of NB cell maturation nor did it induce the expression of any cytoskeletal protein associated with neuronal differentiation. DNA flow cytofluorimetric analysis revealed that HPR did not induce an arrest in a specific phase of the cell cycle while triggering apoptosis. This phenomenon was evidenced both by the visualization of "DNA ladders" on gel electrophoresis and by a quantitative assay for evaluating programmed cell death based upon the labeling of DNA breaks with tritiated thymidine. With the latter method, apoptotic cells were detectable as early as 3-6 h after treatment of NB cells with 10(-5) M HPR, while more than 50% of cells were apoptotic by 24-72 h following exposure to 3 x 10(-6) M HPR. In contrast, RA induced a low rate of apoptosis in NB cells only after 3-5 days. Time lapse photomicroscopy showed that NB cells treated with HPR underwent a death process highly reminiscent of apoptosis, with progressive condensation of the cytoplasm around the nucleus and intense cell shrinkage. The cells then rounded up and detached from the plate. Furthermore, propidium iodide staining of the DNA showed that a high proportion of cells treated with HPR displayed a small and brightly staining nucleus; chromatin appeared aggregated into dense masses in the nuclear periphery, a typical feature of apoptotic cells. In conclusion, our study demonstrates that contrary to the differentiation-promoting activity of RA, HPR dramatically suppresses NB cell growth by inducing programmed cell death.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Apoptose/efeitos dos fármacos , Fenretinida/farmacologia , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Tretinoína/farmacologia , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/efeitos dos fármacos , Dano ao DNA , DNA de Neoplasias/análise , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Imunofluorescência , Humanos , Neurônios/citologia , Neurônios/efeitos dos fármacos
8.
Cell Death Differ ; 4(2): 150-8, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16465221

RESUMO

Interferon-gamma (IFN-gamma) has a well known differentiation-promoting activity on several neuroblastoma (NB) cell lines and has also been reported to induce apoptosis in different cellular models. We have investigated the potential of IFN-gamma to trigger, besides differentiation, programmed cell death in NB cells and the relationship between these processes. Nine NB cell lines, characterized by different phenotypic and maturational features, were cultured in the presence of IFN-gamma (1000 IU/ml) for up to 5 days with either only one treatment at the start of the culture or renewing the culture medium (with or without IFN-gamma) every other day. Neuronal differentiation was assessed by evaluation of morphological changes and expression of mature cytoskeletal proteins, while apoptosis was evaluated at the desired times by fluorescent and electronic microscopy, DNA content analysis and DNA fragmentation assay. Our findings show that apoptosis is an early (mainly non post-differentiative) event and is much more evident following a single IFN-gamma administration. Moreover, IFN-gamma-triggered apoptosis is independent of the cellular phenotype (schwannian or neuronal) and appears to be mutually exclusive with respect to differentiation at the single cell level. Our results strengthen the potential of IFN-gamma as a promising therapeutic agent for NB.

9.
J Control Release ; 103(2): 369-80, 2005 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-15763620

RESUMO

All-trans-retinoic acid (ATRA) is now included in many antitumor therapeutic schemes for the treatment of acute promyelocytic leukaemia, Kaposi's sarcoma, head and neck squamous cell carcinoma, ovarian carcinoma, bladder cancer and neuroblastoma. Unfortunately its poor aqueous solubility hampers its parenteral formulation. To date, there is no parenteral formulation of ATRA commercially available and oral administration of ATRA is associated with progressively diminishing ATRA levels in plasma, which is related to induction of retinoic acid-binding protein and increased drug catabolism by cytochrome P-450-mediated reaction. An ATRA formulation, obtained by complexation of the drug into polymeric micelles, might be suitable for parenteral administration overcoming these unwanted effects. To this purpose we prepared an amphiphilic polymer by polyvinylalcohol (PVA) substitution with oleyl amine at 1.5% substitution degree (mol substituent per 100 mol hydroxyvinylmonomer) and evaluated its functional properties with regard to ATRA complexation. The substituted polymer displayed ability to interact with ATRA both in aqueous solution and in the solid state following spray-drying of drug-polymer hydro-alcoholic solutions. The spray-dried complexes rapidly dissolved in water providing high levels of ATRA solubilization as a function of the drug-polymer weight ratio. The complexes characterized by 1:5 drug-polymer weight ratio provided higher levels of ATRA solubilization than 1:3 and 1:10 drug-polymer weight ratios respectively. Pre-formed polymeric micelles in water equilibrated in the presence of excess solid ATRA provided the lowest levels of solubilization. The drug release from the complexes was very slow in PBS, indicating their suitability in antitumor drug targeting where a fundamental requirement is stability towards drug release for at least 24 h, corresponding to the average circulation time period of macromolecular carriers. The cytotoxicity studies against neuroblastoma cell lines outlined increased cytotoxicity of complexed ATRA with respect to free ATRA, likely due to the increased bioavailability of the hydrophobic drug from the complex. We conclude that ATRA entrapped into self-assembling polymer micelles may be a useful parenteral ATRA formulation overcoming the unwanted pharmacological mechanism that lead to acquired retinoid resistance.


Assuntos
Antineoplásicos/química , Composição de Medicamentos , Álcool de Polivinil/química , Tretinoína/química , Aminas/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Estabilidade de Medicamentos , Humanos , Micelas , Tamanho da Partícula , Solubilidade , Tretinoína/farmacologia
10.
Drug Deliv ; 12(1): 15-20, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15801716

RESUMO

Polyvinylalcohol, partially substituted with lipophilic acyl chains, generates polymeric micelles in aqueous phase, containing a hydrophobic core able to encapsulate lipophilic drugs. Two types of polymers were obtained by conjugation of polyvinylalcohol with oleoyl or linoleoyl chains as pendant groups. The polymers, at a substitution degree of approximately 1%, are soluble in water and form polymeric micelles whose size increases with polymer concentration. Doxorubicin was hydrophobized, by linking an oleoyl chain via amide bond, to make the drug more similar to the substituted polymers and promote its encapsulation into the inner core of the micelles. The properties of the drug-polymer systems were evaluated in solution by dynamic light scattering technique and correlated to the physicochemical characteristics of the drug and the substituted polymers. Solubilization tests revealed that the similarity of the chain, in both the polymer and the drug, promotes better drug encapsulation in the oleoyl than linoleoyl derivative. The drug-polymer systems are stable in phosphate buffer saline (pH 7.4) at 37 degrees C, and the release of the drug is activated by the presence of the proteolytic enzyme pronase-E. The enzyme activated drug release and the size of the polymeric micelles, compatible with the pore dimensions of the tumor vessels, make these systems interesting for targeting lipophilic drugs to solid tumors, where the proteolytic enzyme concentration strongly raises with respect to the other body compartments.


Assuntos
Doxorrubicina/administração & dosagem , Álcool de Polivinil/administração & dosagem , Doxorrubicina/química , Micelas
11.
FEBS Lett ; 310(1): 17-21, 1992 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-1526278

RESUMO

The biomolecular mechanisms that mediate signal transduction by type II (gamma) interferon (IFN) are poorly understood. IFN-gamma is a potent growth inhibitory cytokine also endowed with antiviral, immunomodulatory, and differentiating activities on various cell targets, including neural cells. IFN-gamma induced a rapid and transient activation of phospholipase A2 in LAN-5, a human neuroblastoma cell line. A consequence of phospholipase A2 activation was the release of arachidonic acid and the generation of lysophospholipids from membrane phospholipids. Treatment of pre-labeled LAN-5 cells with a receptor-saturating concentration of IFN-gamma led to a time-dependent release of [3H]arachidonic acid into the culture media and generation of [32P]lysophosphatidylcholine. Pretreatment of cultures with the phospholipase A2 inhibitor, bromophenacyl bromide, markedly inhibited both [3H]arachidonic acid release and lysophosphatidylcholine production induced by IFN-gamma treatment. Pretreatment of LAN-5 cells with nordihydroguaiaretic acid, a lipoxygenase inhibitor, or with indomethacin, a cyclooxygenase inhibitor, amplified the release of [3H]arachidonic acid and production of lysophosphatidylcholine induced by non-saturating concentrations of IFN-gamma. In parallel, and with the same time-dependent effect, a significant decrease in phosphatidylcholine labeling was observed in IFN-gamma-treated cells, further indicating that a potential signal transduction mechanism of IFN-gamma is the hydrolysis of membrane phosphatidylcholine by phospholipase A2.


Assuntos
Interferon gama/farmacologia , Fosfolipases A/metabolismo , Ácido Araquidônico/metabolismo , Cromatografia em Camada Fina , Ativação Enzimática , Humanos , Indometacina/farmacologia , Lisofosfatidilcolinas/biossíntese , Masoprocol/farmacologia , Neuroblastoma , Fosfolipases A2 , Transdução de Sinais , Células Tumorais Cultivadas
12.
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
13.
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
14.
Life Sci ; 68(10): 1161-8, 2001 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-11228100

RESUMO

In human tumors changes in angiogenesis and expression of extracellular matrix-degrading enzymes occur simultaneously during invasion and metastasis. Tissues from 20 biopsies of human neuroblastoma (NB) were investigated immunohistochemically by using an antibody against factor VIII to determine their microvessel number, and by in situ hybridisation to determine the expression of mRNA of the matrix metalloproteinase-2 (MMP-2) and MMP-9. The extent of angiogenesis and the expression of the MMP-2 and MMP-9 mRNA were upregulated in advancing stages. These in situ data suggest that angiogenesis and degradation of extracellular matrix occur simultaneously with NB tumor progression.


Assuntos
Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Neovascularização Patológica/etiologia , Neuroblastoma/irrigação sanguínea , RNA Mensageiro/análise , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Neuroblastoma/enzimologia , Neuroblastoma/patologia
15.
Pediatr Med Chir ; 16(3): 227-34, 1994.
Artigo em Italiano | MEDLINE | ID: mdl-7971444

RESUMO

Differentiation of human neuroblastoma (NB) cells is a very interesting biologic event, providing useful insights in both basic neurobiology and clinical management of this malignancy. Investigation of in vitro NB differentiation exploits several NB cell lines that can be induced to differentiate by an array of natural or synthetic chemicals, as well as biological factors such as some cytokines. The hallmarks of neuronal differentiation are represented by a partial or complete block of cell proliferation, morphological alterations and acquisition of biological features typical of mature neurons (for example, induced synthesis and storage of monoamines and neuropeptides, expression of peculiar cytoskeletal proteins and membrane antigens). The possibility to transfer the differentiative approach to the treatment of NB patients opens exciting therapeutic perspectives.


Assuntos
Neuroblastoma/tratamento farmacológico , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Criança , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fatores de Tempo , Células Tumorais Cultivadas
16.
Drug Deliv ; 16(7): 389-98, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19624248

RESUMO

This study relates to the preparation of a series of amphiphilic dextrins and their evaluation as complexing agents for anti-tumor hydrophobic drugs such as fenretinide, paclitaxel, etoposide, and camptothecin. The amphiphilic dextrins were obtained by conjugation of low molecular weight dextrin (average molecular weight 1670, average polymerization degree 9.33 glucose monomer) with hydrocarbon chains at substitution degree of about 0.1 mole hydrocarbon chain per mole of glucose monomer, as confirmed by 1H-NMR spectra. The conjugates were highly soluble in water and dissolved with formation of nano-aggregates endowed with hydrophobic inner cores able to host hydrophobic drugs by complexation. Complexation raised hydrophobic drugs aqueous solubility; the best results were obtained with fenretinide. Solid complexes with fenretinide were prepared by using three different approaches: the kneading method, the co-solubilisation method, and the co-precipitation method. Kneading method provided the complexes endowed with the best functional properties. Thermogravimetric analysis on solid samples suggested a notable thermal stability up to 300 degrees C for both the conjugated dextrins and the solid complexes. In differential scanning calorimetry profiles no significant differences were observed among amphiphilic dextrins and complexed drug, indicating that the guest molecule exists in an amorphous state in the solid matrices. Particle size analysis confirmed the dimensional suitability of the complexes for parenteral administration. Moreover, sustained drug release, in vitro, has been observed from all the complexes analyzed. Regarding the biological effects, the cytotoxicity of complexed fenretinide towards HTLA-230 neuroblastoma cell line was always higher than the free drug, suggesting that complexation increased drug bioavailability. These findings, taken together, indicated that these biodegradable, self-assembling dextrin conjugates may be regarded as new potential complexing agents for hydrophobic drugs and, in particular, for fenretinide, to increase drug solubility, bioavailability, and thus therapeutic efficacy.


Assuntos
Antineoplásicos/química , Dextrinas/química , Fenretinida/química , Interações Hidrofóbicas e Hidrofílicas , Tensoativos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Dextrinas/farmacologia , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Fenretinida/farmacologia , Humanos , Solubilidade , Tensoativos/farmacologia
17.
Drug Deliv ; 16(4): 189-95, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19241235

RESUMO

All-trans-retinoic acid (ATRA) is now included in many antitumor therapeutic schemes for the treatment of acute promyelocytic leukemia, Kaposi's sarcoma, head and neck squamous cell carcinoma, ovarian carcinoma, bladder cancer, and neuroblastoma. Unfortunately, its poor aqueous solubility hampers its parenteral formulation, whereas oral administration of ATRA is associated with progressively diminishing drug levels in plasma, which is related to induction of retinoic acid-binding proteins and increased drug catabolism by cytochrome P450-mediated reactions. An ATRA formulation, obtained by complexation of the drug into polymeric micelles, might be suitable for parenteral administration overcoming these unwanted effects. To this purpose, amphiphilic polymers were prepared by polyvinylalcohol (PVA) partial esterification with nicotinoyl moieties and their functional properties evaluated with regard to ATRA complexation. The physicochemical characteristics of the polymers and the complexes were analyzed by 1H-NMR, Dynamic Light Scattering (DLS), Capillary Electophoresis (CE), and were correlated with the complex ability to improve the drug solubilization and release the free drug in an aqueous environment. Subsequently, the best complex, providing the highest ATRA solubilization and release, was evaluated in vitro to test its cytotoxicity towards neuroblastoma cell lines. The PVA substitution degree calculated from 1H-NMR was found to be 5.0%, 8.2%, 15.3% (nicotinoyl moiety:PVA monomer molar ratio), while capillary electrophoresis analysis on the complexes revealed that the drug loadings were 0.95%, 1.20%, 4.76% (ATRA:polymer w:w) for PVA substitution degrees of 5.0%, 8.2%, and 15.3%, respectively. Complexation strongly increased ATRA aqueous solubility, which reached 1.20 +/- 0.25 mg/mL. The DLS measurements of the polymers and the complexes in aqueous solutions revealed mean sizes always below 400 nm, low polydispersity (min 0.202 +/- 0.013, max 0.450 +/- 0.032), and size almost unaffected by concentration. Drug fractional release did not exceed 8% after 48 h. The cytotoxicity studies against neuroblastoma cell lines outlined a significant growth inhibition effect of complexed ATRA with respect to free ATRA. These data suggest that the systems analyzed may be suitable carriers for parenteral administration of ATRA and other hydrophobic antitumor drugs, where the carriers are required to improve drug aqueous solubility and delay drug release almost after their accumulation in solid tumors.


Assuntos
Antineoplásicos/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Infusões Parenterais , Neuroblastoma/terapia , Tretinoína/administração & dosagem , Química Farmacêutica , Vias de Administração de Medicamentos , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Ésteres/química , Humanos , Micelas , Neuroblastoma/patologia , Álcool de Polivinil/química , Solubilidade , Tretinoína/química , Células Tumorais Cultivadas
18.
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
19.
Biomacromolecules ; 8(10): 3258-62, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17883277

RESUMO

N-(4-hydroxyphenyl)retinamide (fenretinide, 4-HPR) has been shown to be active toward many tumors without appreciable side effects. However its in vitro activity does not match a correspondent efficacy in vivo. The main reason is that the drug's hydrophobicity hinders its bioavailability in the body fluids. Even if the drug is previously dissolved in organic solvents, such as ethanol or DMSO, the subsequent dilution in body fluids trigger its precipitation in fine aggregates characterized by very low dissolution efficiency, never reaching amounts suitable for therapeutic response. To date no intravenous formulation of 4-HPR exists on the market. The 4-HPR linkage to a hydrophilic polymer by a covalent bond easily hydrolyzable in aqueous environment is expected to increase the drug's aqueous solubility, providing the free drug after hydrolysis of the covalent bond. This may be a useful tool for the preparation of aqueous intravenous formulations of 4-HPR. For this purpose, we linked 4-HPR to polyvinylalcohol (PVA) by a carbonate bond at different drug/hydroxy vinyl monomer molar ratios. We demonstrated that conjugation increased 4-HPR aqueous solubility and strongly inhibited neuroblastoma cell proliferation. In addition, in an in vivo neuroblastoma metastatic model, we obtained a significant antitumor effect as a consequence of the improved drug bioavailability.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Fenretinida/administração & dosagem , Fenretinida/química , Álcool de Polivinil/química , Animais , Disponibilidade Biológica , Linhagem Celular Tumoral , Proliferação de Células , Dimetil Sulfóxido/química , Humanos , Infusões Intravenosas , Camundongos , Camundongos Nus , Modelos Químicos , Metástase Neoplásica , Neuroblastoma/metabolismo , Solubilidade
20.
Biomacromolecules ; 7(11): 3157-63, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17096546

RESUMO

Poly(vinyl alcohol) (PVA) substituted with oleyl chains and tetraethyleneglycol monoethyl ether chains (TEGMEE) at 1.5% and 1% degrees of substitution respectively (mol of substituent to mol of hydroxyvinyl monomer) has previously been shown to self-assemble in water, providing aggregates selectively cytotoxic toward tumor cells vs normal cells. These polymers have also been shown to increase the long-term survival of nude mice injected with both human and murine neuroblastoma cell lines. In the present work, we changed the substitution degree of the oleyl chains on the poly(vinyl alcohol) backbone and maintained constant at 1% the degree of TEGMEE substitution. We evaluated the main physicochemical characteristics of the final polymers, their cytotoxicity toward tumor cells, and their complexing ability for hydrophobic molecules. The aim was to investigate the possibility of improving intrinsic antitumor efficacy of the polymer by changing the degree of oleyl chain substitution and further increase activity by complexation with antitumor drugs. The polymers were prepared at oleyl chain substitution degrees ranging from 0.5 to 3% (mol of substituent to mol of hydroxyvinyl monomer). The most active was again the 1.5% substituted polymer. It was further characterized by exhibiting the highest complexing ability toward hydrophobic molecules allowing the formation of a complex with fenretinide (HPR). The polymer-HPR complex was stable in aqueous environment and released the free drug prevalently in the presence of fluid hydrophobic phases. It was cytotoxic toward tumor cells with minimal activity toward normal cells. Antitumor activity exceeded that of the separate complex components resulting from the concomitant effect of the polymer and the HPR solubilized by complexation.


Assuntos
Antineoplásicos/química , Fenretinida/química , Álcool de Polivinil/química , Animais , Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Espectroscopia de Ressonância de Spin Eletrônica , Fenretinida/farmacologia , Humanos , Camundongos , Camundongos Nus , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa